1501
|
Craciun CI, Neag MA, Catinean A, Mitre AO, Rusu A, Bala C, Roman G, Buzoianu AD, Muntean DM, Craciun AE. The Relationships between Gut Microbiota and Diabetes Mellitus, and Treatments for Diabetes Mellitus. Biomedicines 2022; 10:biomedicines10020308. [PMID: 35203519 PMCID: PMC8869176 DOI: 10.3390/biomedicines10020308] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/15/2022] [Accepted: 01/26/2022] [Indexed: 02/04/2023] Open
Abstract
Diabetes mellitus is considered to be a global epidemic. The combination of genetic susceptibility and an unhealthy lifestyle is considered to be the main trigger of this metabolic disorder. Recently, there has been increased interest in the roles of gut microbiota as a new potential contributor to this epidemic. Research, in recent years, has contributed to an in-depth characterization of the human microbiome and its associations with various diseases, including metabolic diseases and diabetes mellitus. It is known that diet can change the composition of gut microbiota, but it is unclear how this, in turn, may influence metabolism. The main objective of this review is to evaluate the pathogenetic association between microbiota and diabetes and to explore any new therapeutic agents, including nutraceuticals that may modulate the microbiota. We also look at several mechanisms involved in this process. There is a clear, bidirectional relationship between microbiota and diabetes. Current treatments for diabetes influence microbiota in various ways, some beneficial, but others with still unclear effects. Microbiota-aimed treatments have seen no real-world significant effects on the progression of diabetes and its complications, with more studies needed in order to find a really beneficial agent.
Collapse
Affiliation(s)
- Cristian-Ioan Craciun
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (C.-I.C.); (A.-D.B.)
| | - Maria-Adriana Neag
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (C.-I.C.); (A.-D.B.)
- Correspondence:
| | - Adrian Catinean
- Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania;
| | - Andrei-Otto Mitre
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Adriana Rusu
- Department of Diabetes, Nutrition, Metabolic Diseases, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (A.R.); (C.B.); (G.R.); (A.-E.C.)
| | - Cornelia Bala
- Department of Diabetes, Nutrition, Metabolic Diseases, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (A.R.); (C.B.); (G.R.); (A.-E.C.)
| | - Gabriela Roman
- Department of Diabetes, Nutrition, Metabolic Diseases, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (A.R.); (C.B.); (G.R.); (A.-E.C.)
| | - Anca-Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (C.-I.C.); (A.-D.B.)
| | - Dana-Maria Muntean
- Department of Pharmaceutical Technology and Biopharmaceutics, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Anca-Elena Craciun
- Department of Diabetes, Nutrition, Metabolic Diseases, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (A.R.); (C.B.); (G.R.); (A.-E.C.)
| |
Collapse
|
1502
|
Hillestad EMR, van der Meeren A, Nagaraja BH, Bjørsvik BR, Haleem N, Benitez-Paez A, Sanz Y, Hausken T, Lied GA, Lundervold A, Berentsen B. Gut bless you: The microbiota-gut-brain axis in irritable bowel syndrome. World J Gastroenterol 2022; 28:412-431. [PMID: 35125827 PMCID: PMC8790555 DOI: 10.3748/wjg.v28.i4.412] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 06/24/2021] [Accepted: 01/13/2022] [Indexed: 12/16/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a common clinical label for medically unexplained gastrointestinal symptoms, recently described as a disturbance of the microbiota-gut-brain axis. Despite decades of research, the pathophysiology of this highly heterogeneous disorder remains elusive. However, a dramatic change in the understanding of the underlying pathophysiological mechanisms surfaced when the importance of gut microbiota protruded the scientific picture. Are we getting any closer to understanding IBS' etiology, or are we drowning in unspecific, conflicting data because we possess limited tools to unravel the cluster of secrets our gut microbiota is concealing? In this comprehensive review we are discussing some of the major important features of IBS and their interaction with gut microbiota, clinical microbiota-altering treatment such as the low FODMAP diet and fecal microbiota transplantation, neuroimaging and methods in microbiota analyses, and current and future challenges with big data analysis in IBS.
Collapse
Affiliation(s)
- Eline Margrete Randulff Hillestad
- Department of Clinical Medicine, University of Bergen, Bergen 5021, Norway
- National Center for Functional Gastrointestinal Disorders, Department of Medicine, Haukeland University Hospital, Bergen 5021, Norway
| | - Aina van der Meeren
- National Center for Functional Gastrointestinal Disorders, Department of Medicine, Haukeland University Hospital, Bergen 5021, Norway
| | - Bharat Halandur Nagaraja
- Mohn Medical Imaging and Visualization Center, Department of Radiology, Haukeland University Hospital, Bergen 5021, Norway
| | - Ben René Bjørsvik
- National Center for Functional Gastrointestinal Disorders, Department of Medicine, Haukeland University Hospital, Bergen 5021, Norway
- Mohn Medical Imaging and Visualization Center, Department of Radiology, Haukeland University Hospital, Bergen 5021, Norway
| | - Noman Haleem
- National Center for Functional Gastrointestinal Disorders, Department of Medicine, Haukeland University Hospital, Bergen 5021, Norway
- Mohn Medical Imaging and Visualization Center, Department of Radiology, Haukeland University Hospital, Bergen 5021, Norway
| | - Alfonso Benitez-Paez
- Host-Microbe Interactions in Metabolic Health Laboratory, Principe Felipe Research Center, Valencia 46012, Spain
| | - Yolanda Sanz
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council, Paterna-Valencia 46980, Spain
| | - Trygve Hausken
- Department of Clinical Medicine, University of Bergen, Bergen 5021, Norway
- National Center for Functional Gastrointestinal Disorders, Department of Medicine, Haukeland University Hospital, Bergen 5021, Norway
| | - Gülen Arslan Lied
- National Center for Functional Gastrointestinal Disorders, Department of Medicine, Haukeland University Hospital, Bergen 5021, Norway
- Center for Nutrition, Department of Clinical Medicine, University of Bergen, Bergen 5021, Norway
| | - Arvid Lundervold
- Mohn Medical Imaging and Visualization Center, Department of Radiology, Haukeland University Hospital, Bergen 5021, Norway
- Department of Biomedicine, University of Bergen, Bergen 5021, Norway
| | - Birgitte Berentsen
- Department of Clinical Medicine, University of Bergen, Bergen 5021, Norway
- National Center for Functional Gastrointestinal Disorders, Department of Medicine, Haukeland University Hospital, Bergen 5021, Norway
| |
Collapse
|
1503
|
Siddiqui SH, Khan M, Kang D, Choi HW, Shim K. Meta-Analysis and Systematic Review of the Thermal Stress Response: Gallus gallus domesticus Show Low Immune Responses During Heat Stress. Front Physiol 2022; 13:809648. [PMID: 35153835 PMCID: PMC8832064 DOI: 10.3389/fphys.2022.809648] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Heat stress, which affects broiler growth performance and immunity, is a major concern in the poultry industry. This meta-analysis aimed to demonstrate the significant effect of heat stress on broiler mass gain and immunoglobulin levels, which regulates the mortality rate of broilers. A total of 2,585 studies were downloaded from PubMed, Web of Science, and Google Scholar from January 1, 2015, to September 1, 2021. Eventually, 28 studies were selected based on specific criteria. The results for body mass gain, total mass of immune organs (thymus, spleen, and bursa of Fabricius), immunoglobulin (IgA, IgG, and IgM) levels, and mortality rate were analyzed using odds ratio or the random-effects model (REM) at a confidence interval (CI) of 95%. Compared to the control, heat stress significantly decreased body mass gain (10 trials: REM = 1.35, 95% CI: 1.21, 1.50). Compared to that in the control, heat stress significantly increased immunoglobulin levels: IgA (7 trials: REM = 1.69, 95% CI: 0.90, 3.16), IgG (6 trials: REM = 1.24, 95% CI: 0.85, 1.81), IgM (8 trials: REM = 0.69, 95% CI: 0.44, 1.08), and heat stress also increased the broiler mortality rate (6 trials: REM = 0.06, 95% CI: 0.01, 0.27). However, there were no significant changes in the immune organs between the control and heat-stressed groups. In conclusion, heat stress remarkably alters the mass gain and immunoglobulin levels of broilers, which may be a cause of the high mortality rate.
Collapse
Affiliation(s)
- Sharif Hasan Siddiqui
- Department of Animal Biotechnology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
| | - Mousumee Khan
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, South Korea
| | - Darae Kang
- Department of Animal Biotechnology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
| | - Hyun Woo Choi
- Department of Animal Science, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
- Department of Agricultural Convergence Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
| | - Kwanseob Shim
- Department of Animal Biotechnology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
- Department of Agricultural Convergence Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
- *Correspondence: Kwanseob Shim
| |
Collapse
|
1504
|
Notararigo S, Varela E, Otal A, Antolín M, Guarner F, López P. Anti-Inflammatory Effect of an O-2-Substituted (1-3)-β-D-Glucan Produced by Pediococcus parvulus 2.6 in a Caco-2 PMA-THP-1 Co-Culture Model. Int J Mol Sci 2022; 23:ijms23031527. [PMID: 35163449 PMCID: PMC8835822 DOI: 10.3390/ijms23031527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 01/01/2023] Open
Abstract
Bacterial β-glucans are exopolysaccharides (EPSs), which can protect bacteria or cooperate in biofilm formation or in bacterial cell adhesion. Pediococcus parvulus 2.6 is a lactic acid bacterium that produces an O-2-substituted (1-3)-β-D-glucan. The structural similarity of this EPS to active compounds such as laminarin, together with its ability to modulate the immune system and to adhere in vitro to human enterocytes, led us to investigate, in comparison with laminarin, its potential as an immunomodulator of in vitro co-cultured Caco-2 and PMA-THP-1 cells. O-2-substituted (1-3)-β-D-glucan synthesized by the GTF glycosyl transferase of Pediococcus parvulus 2.6 or that by Lactococcus lactis NZ9000[pGTF] were purified and used in this study. The XTT tests revealed that all β-glucans were non-toxic for both cell lines and activated PMA-THP-1 cells’ metabolisms. The O-2-substituted (1-3)-β-D-glucan modulated production and expression of IL-8 and the IL-10 in Caco-2 and PMA-THP-1 cells. Laminarin also modulated cytokine production by diminishing TNF-α in Caco-2 cells and IL-8 in PMA-THP-1. All these features could be considered with the aim to produce function foods, supplemented with laminarin or with another novel β-glucan-producing strain, in order to ameliorate an individual’s immune system response toward pathogens or to control mild side effects in remission patients affected by inflammatory bowel diseases.
Collapse
Affiliation(s)
- Sara Notararigo
- Molecular Biology of Gram-Positive Bacteria, Margarita Salas Center for Biological Research (CIB-Margarita Salas-CSIC), Department of Microbial and Plant Biotechnology, Ramiro de Maeztu 9, 28040 Madrid, Spain;
- Digestive System Research Unit, Institut de RecercaValld’Hebron (VHIR), University Hospital Valld’Hebron, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.V.); (A.O.); (M.A.); (F.G.)
- Foundation Health Research Institute of Santiago de Compostela (FIDIS), 15706 Santiago de Compostela, Spain
| | - Encarnación Varela
- Digestive System Research Unit, Institut de RecercaValld’Hebron (VHIR), University Hospital Valld’Hebron, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.V.); (A.O.); (M.A.); (F.G.)
- CIBERehd, Instituto Carlos III, 28029 Madrid, Spain
| | - Anna Otal
- Digestive System Research Unit, Institut de RecercaValld’Hebron (VHIR), University Hospital Valld’Hebron, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.V.); (A.O.); (M.A.); (F.G.)
| | - María Antolín
- Digestive System Research Unit, Institut de RecercaValld’Hebron (VHIR), University Hospital Valld’Hebron, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.V.); (A.O.); (M.A.); (F.G.)
- CIBERehd, Instituto Carlos III, 28029 Madrid, Spain
| | - Francisco Guarner
- Digestive System Research Unit, Institut de RecercaValld’Hebron (VHIR), University Hospital Valld’Hebron, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.V.); (A.O.); (M.A.); (F.G.)
- CIBERehd, Instituto Carlos III, 28029 Madrid, Spain
| | - Paloma López
- Molecular Biology of Gram-Positive Bacteria, Margarita Salas Center for Biological Research (CIB-Margarita Salas-CSIC), Department of Microbial and Plant Biotechnology, Ramiro de Maeztu 9, 28040 Madrid, Spain;
- Correspondence: ; Tel.: +34-91-837-31-12; Fax: +34-91-538-04-32
| |
Collapse
|
1505
|
Rannug A. 6-Formylindolo[3,2-b]carbazole, a Potent Ligand for the Aryl Hydrocarbon Receptor Produced Both Endogenously and by Microorganisms, can Either Promote or Restrain Inflammatory Responses. FRONTIERS IN TOXICOLOGY 2022; 4:775010. [PMID: 35295226 PMCID: PMC8915874 DOI: 10.3389/ftox.2022.775010] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) binds major physiological modifiers of the immune system. The endogenous 6-formylindolo[3,2-b]carbazole (FICZ), which binds with higher affinity than any other compound yet tested, including TCDD, plays a well-documented role in maintaining the homeostasis of the intestines and skin. The effects of transient activation of AHR by FICZ differ from those associated with continuous stimulation and, depending on the dose, include either differentiation into T helper 17 cells that express proinflammatory cytokines or into regulatory T cells or macrophages with anti-inflammatory properties. Moreover, in experimental models of human diseases high doses stimulate the production of immunosuppressive cytokines and suppress pathogenic autoimmunity. In our earlier studies we characterized the formation of FICZ from tryptophan via the precursor molecules indole-3-pyruvate and indole-3-acetaldehyde. In the gut formation of these precursor molecules is catalyzed by microbial aromatic-amino-acid transaminase ArAT. Interestingly, tryptophan can also be converted into indole-3-pyruvate by the amino-acid catabolizing enzyme interleukin-4 induced gene 1 (IL4I1), which is secreted by host immune cells. By thus generating derivatives of tryptophan that activate AHR, IL4I1 may have a role to play in anti-inflammatory responses, as well as in a tumor escape mechanism that reduces survival in cancer patients. The realization that FICZ can be produced from tryptophan by sunlight, by enzymes expressed in our cells (IL4I1), and by microorganisms as well makes it highly likely that this compound is ubiquitous in humans. A diurnal oscillation in the level of FICZ that depends on the production by the fluctuating number of microbes might influence not only intestinal and dermal immunity locally, but also systemic immunity.
Collapse
|
1506
|
Paparo L, Maglio MA, Cortese M, Bruno C, Capasso M, Punzo E, Ferrucci V, Lasorsa VA, Viscardi M, Fusco G, Cerino P, Romano A, Troncone R, Zollo M. A New Butyrate Releaser Exerts a Protective Action against SARS-CoV-2 Infection in Human Intestine. Molecules 2022; 27:862. [PMID: 35164139 PMCID: PMC8838168 DOI: 10.3390/molecules27030862] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/12/2022] [Accepted: 01/25/2022] [Indexed: 12/12/2022] Open
Abstract
Butyrate is a major gut microbiome metabolite that regulates several defense mechanisms against infectious diseases. Alterations in the gut microbiome, leading to reduced butyrate production, have been reported in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. A new butyrate releaser, useful for all the known applications of butyrate, presenting physiochemical characteristics suitable for easy oral administration, (N-(1-carbamoyl-2-phenyl-ethyl) butyramide (FBA), has been recently developed. We investigated the protective action of FBA against SARS-CoV-2 infection in the human small intestine and enterocytes. Relevant aspects of SARS-CoV-2 infection were assessed: infectivity, host functional receptor angiotensin-converting enzyme-2 (ACE2), transmembrane protease serine 2 (TMPRSS2), neuropilin-1 (NRP1), pro-inflammatory cytokines expression, genes involved in the antiviral response and the activation of Nf-kB nuclear factor (erythroid-derived 2-like) 2 (Nfr2) pathways. We found that FBA positively modulates the crucial aspects of the infection in small intestinal biopsies and human enterocytes, reducing the expression of ACE2, TMPRSS2 and NRP1, pro-inflammatory cytokines interleukin (IL)-15, monocyte chemoattractant protein-1 (MCP-1) and TNF-α, and regulating several genes involved in antiviral pathways. FBA was also able to reduce the number of SARS-CoV-2-infected cells, and ACE2, TMPRSS2 and NRP1 expression. Lastly, through the inhibition of Nf-kB and the up-regulation of Nfr2, it was also able to reduce the expression of pro-inflammatory cytokines IL-15, MCP-1 and TNF-α in human enterocytes. The new butyrate releaser, FBA, exerts a preventive action against SARS-CoV-2 infection. It could be considered as an innovative strategy to limit COVID-19.
Collapse
Affiliation(s)
- Lorella Paparo
- Dipartimento di Scienze Mediche Translazionali, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.A.M.); (M.C.); (C.B.); (E.P.); (R.T.)
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
| | - Maria Antonia Maglio
- Dipartimento di Scienze Mediche Translazionali, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.A.M.); (M.C.); (C.B.); (E.P.); (R.T.)
| | - Maddalena Cortese
- Dipartimento di Scienze Mediche Translazionali, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.A.M.); (M.C.); (C.B.); (E.P.); (R.T.)
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
| | - Cristina Bruno
- Dipartimento di Scienze Mediche Translazionali, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.A.M.); (M.C.); (C.B.); (E.P.); (R.T.)
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
| | - Mario Capasso
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy
| | - Erika Punzo
- Dipartimento di Scienze Mediche Translazionali, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.A.M.); (M.C.); (C.B.); (E.P.); (R.T.)
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
| | - Veronica Ferrucci
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy
| | - Vito Alessandro Lasorsa
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
| | - Maurizio Viscardi
- DAI Medicina di Laboratorio e Trasfusionale, AOU Azienda Ospedaliera, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.V.); (G.F.); (P.C.)
| | - Giovanna Fusco
- DAI Medicina di Laboratorio e Trasfusionale, AOU Azienda Ospedaliera, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.V.); (G.F.); (P.C.)
| | - Pellegrino Cerino
- DAI Medicina di Laboratorio e Trasfusionale, AOU Azienda Ospedaliera, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.V.); (G.F.); (P.C.)
| | - Alessia Romano
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
| | - Riccardo Troncone
- Dipartimento di Scienze Mediche Translazionali, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.A.M.); (M.C.); (C.B.); (E.P.); (R.T.)
| | - Massimo Zollo
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy
- DAI Medicina di Laboratorio e Trasfusionale, AOU Azienda Ospedaliera, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.V.); (G.F.); (P.C.)
| |
Collapse
|
1507
|
Guryanova SV, Gigani OB, Gudima GO, Kataeva AM, Kolesnikova NV. Dual Effect of Low-Molecular-Weight Bioregulators of Bacterial Origin in Experimental Model of Asthma. Life (Basel) 2022; 12:192. [PMID: 35207480 PMCID: PMC8879587 DOI: 10.3390/life12020192] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 02/07/2023] Open
Abstract
Asthma is one of the most common noncommunicable diseases, affecting over 200 million people. A large number of drugs control asthma attacks, but there is no effective therapy. Identification of reasons for asthma and preventing this disease is a relevant task. The influence of bacterial components is necessary for the normal development of the immune system and the formation of an adequate immune response to antigens. In the absence of microorganisms or their insufficient exposure, the prerequisites are formed for excessive reactivity to harmless antigens. In the present study, we analyzed cellular and humoral factors in a standard mouse model of OVA-induced asthma modified by 5-fold intraperitoneal injection of bacterial cell wall fragments of glucosaminylmuramyl dipeptide (GMDP) 5 μg/animal or 1 μg lipopolysaccharide (LPS) per animal for 5 days before sensitization by ovalbumin (OVA). Preliminary administration of LPS or GMDP to animals significantly reduced goblet cells as well as the number of neutrophils, lymphocytes, and eosinophils in bronchoalveolar lavage, wherein GMDP corrected neutrophilia to a 2-fold degree, and LPS reduced the severity of eosinophilia by 1.9 times. With OVA administration of GMDP or LPS at the sensitization stage, an increase in the total number of bronchoalveolar lavage cells due to neutrophils, macrophages, lymphocytes, and eosinophils in relation to the group with asthma without GMDP or LPS was observed. The administration of GMDP or LPS to normal mice without asthma for 5 days had no statistically significant effect on the change in the number and population composition of cells in bronchoalveolar lavage in comparison with the control group receiving PBS. As a result of a study in a mouse model of asthma, a dual effect of LPS and GMDP was established: the introduction of LPS or GMDP before sensitization reduces neutrophilia and eosinophilia, while the introduction of LPS or GMDP together with an allergen significantly increases neutrophilia and eosinophilia. The study of the immunoglobulin status shows that in normal-asthma mice, GMDP and LPS slightly increase IgA in bronchoalveolar lavage; at the same time, in the asthma model, injections of GMDP or LPS before sensitization contribute to a significant decrease in IgA (2.6 times and 2.1 times, respectively) in BALF and IgE (2.2 times and 2.0 times, respectively) in blood serum. In an experimental model of asthma, the effect of GMDP and LPS was multidirectional: when they are repeatedly administered before sensitization, the bacterial components significantly reduce the severity of the allergic process, while in the case of a joint injection with an allergen, they increase the influx of macrophages, lymphocytes, and neutrophils into the lungs, which can aggravate the course of pathological process. Thus, the insufficient effect of antigens of a bacterial nature, in particular, with prolonged use of antibiotics can be compensated for by substances based on low-molecular-weight bioregulators of bacterial origin to establish the missing signals for innate immunity receptors, whose constant activation at a certain level is necessary to maintain homeostasis.
Collapse
Affiliation(s)
- Svetlana V. Guryanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, Ministry of Science and Higher Education of the Russian Federation, 117997 Moscow, Russia
- Medical Institute, Peoples’ Friendship University of Russia (RUDN University), Ministry of Science and Higher Education of the Russian Federation, 117198 Moscow, Russia; (O.B.G.); (A.M.K.)
| | - Olga B. Gigani
- Medical Institute, Peoples’ Friendship University of Russia (RUDN University), Ministry of Science and Higher Education of the Russian Federation, 117198 Moscow, Russia; (O.B.G.); (A.M.K.)
| | - Georgii O. Gudima
- National Research Center-Institute of Immunology of the Federal Medico-Biological Agency, 115522 Moscow, Russia;
| | - Anastasiya M. Kataeva
- Medical Institute, Peoples’ Friendship University of Russia (RUDN University), Ministry of Science and Higher Education of the Russian Federation, 117198 Moscow, Russia; (O.B.G.); (A.M.K.)
| | - Natalya V. Kolesnikova
- Department of Clinical Immunology, Kuban State Medical University, Ministry of Health of the Russian Federation, 350063 Krasnodar, Russia;
| |
Collapse
|
1508
|
Singh P, Rawat A, Saadaoui M, Elhag D, Tomei S, Elanbari M, Akobeng AK, Mustafa A, Abdelgadir I, Udassi S, Hendaus MA, Al Khodor S. Tipping the Balance: Vitamin D Inadequacy in Children Impacts the Major Gut Bacterial Phyla. Biomedicines 2022; 10:biomedicines10020278. [PMID: 35203487 PMCID: PMC8869474 DOI: 10.3390/biomedicines10020278] [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: 12/14/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
Vitamin D inadequacy appears to be on the rise globally, and it has been linked to an increased risk of osteoporosis, as well as metabolic, cardiovascular, and autoimmune diseases. Vitamin D concentrations are partially determined by genetic factors. Specific single nucleotide polymorphisms (SNPs) in genes involved in vitamin D transport, metabolism, or binding have been found to be associated with its serum concentration, and these SNPs differ among ethnicities. Vitamin D has also been suggested to be a regulator of the gut microbiota and vitamin D deficiency as the possible cause of gut microbial dysbiosis and inflammation. This pilot study aims to fill the gap in our understanding of the prevalence, cause, and implications of vitamin D inadequacy in a pediatric population residing in Qatar. Blood and fecal samples were collected from healthy subjects aged 4–14 years. Blood was used to measure serum metabolite of vitamin D, 25-hydroxycholecalciferol 25(OH)D. To evaluate the composition of the gut microbiota, fecal samples were subjected to 16S rRNA gene sequencing. High levels of vitamin D deficiency/insufficiency were observed in our cohort with 97% of the subjects falling into the inadequate category (with serum 25(OH)D < 75 nmol/L). The CT genotype in rs12512631, an SNP in the GC gene, was associated with low serum levels of vitamin D (ANOVA, p = 0.0356) and was abundant in deficient compared to non-deficient subjects. Overall gut microbial community structure was significantly different between the deficient (D) and non-deficient (ND) groups (Bray Curtis dissimilarity p = 0.049), with deficient subjects also displaying reduced gut microbial diversity. Significant differences were observed among the two major gut phyla, Firmicutes (F) and Bacteroidetes (B), where deficient subjects displayed a higher B/F ratio (p = 0.0097) compared to ND. Vitamin D deficient children also demonstrated gut enterotypes dominated by the genus Prevotella as opposed to Bacteroides. Our findings suggest that pediatric vitamin D inadequacy significantly impacts the gut microbiota. We also highlight the importance of considering host genetics and baseline gut microbiome composition in interpreting the clinical outcomes related to vitamin D deficiency as well as designing better personalized strategies for therapeutic interventions.
Collapse
Affiliation(s)
- Parul Singh
- Research Department, Sidra Medicine, Doha P.O. Box 26999, Qatar; (P.S.); (A.R.); (M.S.); (D.E.); (S.T.); (M.E.)
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 5825, Qatar
| | - Arun Rawat
- Research Department, Sidra Medicine, Doha P.O. Box 26999, Qatar; (P.S.); (A.R.); (M.S.); (D.E.); (S.T.); (M.E.)
| | - Marwa Saadaoui
- Research Department, Sidra Medicine, Doha P.O. Box 26999, Qatar; (P.S.); (A.R.); (M.S.); (D.E.); (S.T.); (M.E.)
| | - Duaa Elhag
- Research Department, Sidra Medicine, Doha P.O. Box 26999, Qatar; (P.S.); (A.R.); (M.S.); (D.E.); (S.T.); (M.E.)
| | - Sara Tomei
- Research Department, Sidra Medicine, Doha P.O. Box 26999, Qatar; (P.S.); (A.R.); (M.S.); (D.E.); (S.T.); (M.E.)
| | - Mohammed Elanbari
- Research Department, Sidra Medicine, Doha P.O. Box 26999, Qatar; (P.S.); (A.R.); (M.S.); (D.E.); (S.T.); (M.E.)
| | - Anthony K. Akobeng
- Division of Gastroenterology, Hepatology, and Nutrition, Sidra Medicine, Doha P.O. Box 26999, Qatar;
| | - Amira Mustafa
- Pediatric Department, Sidra Medicine, Doha P.O. Box 26999, Qatar; (A.M.); (S.U.); (M.A.H.)
| | | | - Sharda Udassi
- Pediatric Department, Sidra Medicine, Doha P.O. Box 26999, Qatar; (A.M.); (S.U.); (M.A.H.)
| | - Mohammed A. Hendaus
- Pediatric Department, Sidra Medicine, Doha P.O. Box 26999, Qatar; (A.M.); (S.U.); (M.A.H.)
| | - Souhaila Al Khodor
- Research Department, Sidra Medicine, Doha P.O. Box 26999, Qatar; (P.S.); (A.R.); (M.S.); (D.E.); (S.T.); (M.E.)
- Correspondence: ; Tel.: +974-4003-7397
| |
Collapse
|
1509
|
West AG, DeLaunay A, Marsh P, Perry EK, Jolly M, Gartrell BD, Pas A, Digby A, Taylor MW. Gut microbiota of the threatened takahē: biogeographic patterns and conservation implications. Anim Microbiome 2022; 4:11. [PMID: 35078539 PMCID: PMC8790836 DOI: 10.1186/s42523-021-00158-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 12/22/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The Aotearoa New Zealand takahē (Porphyrio hochstetteri), once thought to be extinct, is a nationally threatened flightless rail under intensive conservation management. While there has been previous research into disease-related microbes in takahē, little is known about the microbes present in the gastrointestinal tract. Given the importance of gut-associated microbes to herbivore nutrition and immunity, knowledge of these communities is likely to be of considerable conservation value. Here we examined the gut microbiotas of 57 takahē at eight separate locations across Aotearoa New Zealand. RESULTS Faecal samples, taken as a proxy for the hindgut bacterial community, were subjected to 16S rRNA gene amplicon sequencing using Illumina MiSeq. Phylogenetic analysis of > 2200 amplicon sequence variants (ASVs) revealed nine main bacterial phyla (Acidobacteriota, Actinobacteriota, Bacteroidota, Campilobacterota, Firmicutes, Fusobacteriota, Planctomycetota, Proteobacteria, and Verrucomicrobiota) that accounted for the majority of sequence reads. Location was a significant effect (p value < 0.001, 9999 permutations) that accounted for 32% of the observed microbiota variation. One ASV, classified as Lactobacillus aviarius, was present in all samples at an average relative abundance of 17% (SD = 23.20). There was strong evidence (p = 0.002) for a difference in the abundance of the genus Lactobacillus between locations. A common commensal bacterium previously described in takahē, Campylobacter spp., was also detected in most faecal samples. CONCLUSIONS Location plays a pivotal role in the observed variation among takahē gut bacterial communities and is potentially due to factors such as supplemental feeding and medical treatment experienced by birds housed in captivity at one of the eight sampled sites. These data present a first glimpse of the previously unexplored takahē gut microbiota and provide a baseline for future microbiological studies and conservation efforts.
Collapse
Affiliation(s)
- Annie G West
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Anne DeLaunay
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Phil Marsh
- Takahē Recovery Programme, Department of Conservation, Lakefront Drive, Te Anau, New Zealand
| | - Elena K Perry
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Megan Jolly
- School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - Brett D Gartrell
- School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - An Pas
- New Zealand Centre for Conservation Medicine, Auckland Zoo, Auckland, New Zealand
| | - Andrew Digby
- Takahē Recovery Programme, Department of Conservation, Lakefront Drive, Te Anau, New Zealand
| | - Michael W Taylor
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| |
Collapse
|
1510
|
van Dorst JM, Tam RY, Ooi CY. What Do We Know about the Microbiome in Cystic Fibrosis? Is There a Role for Probiotics and Prebiotics? Nutrients 2022; 14:nu14030480. [PMID: 35276841 PMCID: PMC8840103 DOI: 10.3390/nu14030480] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Cystic fibrosis (CF) is a life-shortening genetic disorder that affects the cystic fibrosis transmembrane conductance regulator (CFTR) protein. In the gastrointestinal (GI) tract, CFTR dysfunction results in low intestinal pH, thick and inspissated mucus, a lack of endogenous pancreatic enzymes, and reduced motility. These mechanisms, combined with antibiotic therapies, drive GI inflammation and significant alteration of the GI microbiota (dysbiosis). Dysbiosis and inflammation are key factors in systemic inflammation and GI complications including malignancy. The following review examines the potential for probiotic and prebiotic therapies to provide clinical benefits through modulation of the microbiome. Evidence from randomised control trials suggest probiotics are likely to improve GI inflammation and reduce the incidence of CF pulmonary exacerbations. However, the highly variable, low-quality data is a barrier to the implementation of probiotics into routine CF care. Epidemiological studies and clinical trials support the potential of dietary fibre and prebiotic supplements to beneficially modulate the microbiome in gastrointestinal conditions. To date, limited evidence is available on their safety and efficacy in CF. Variable responses to probiotics and prebiotics highlight the need for personalised approaches that consider an individual’s underlying microbiota, diet, and existing medications against the backdrop of the complex nutritional needs in CF.
Collapse
Affiliation(s)
- Josie M. van Dorst
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, UNSW, Sydney 2031, Australia; (J.M.v.D.); (R.Y.T.)
| | - Rachel Y. Tam
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, UNSW, Sydney 2031, Australia; (J.M.v.D.); (R.Y.T.)
| | - Chee Y. Ooi
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, UNSW, Sydney 2031, Australia; (J.M.v.D.); (R.Y.T.)
- Molecular and Integrative Cystic Fibrosis (miCF) Research Centre, Sydney 2031, Australia
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney 2031, Australia
- Correspondence:
| |
Collapse
|
1511
|
Jung SM, Kim S. In vitro Models of the Small Intestine for Studying Intestinal Diseases. Front Microbiol 2022; 12:767038. [PMID: 35058894 PMCID: PMC8765704 DOI: 10.3389/fmicb.2021.767038] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
The small intestine is a digestive organ that has a complex and dynamic ecosystem, which is vulnerable to the risk of pathogen infections and disorders or imbalances. Many studies have focused attention on intestinal mechanisms, such as host–microbiome interactions and pathways, which are associated with its healthy and diseased conditions. This review highlights the intestine models currently used for simulating such normal and diseased states. We introduce the typical models used to simulate the intestine along with its cell composition, structure, cellular functions, and external environment and review the current state of the art for in vitro cell-based models of the small intestine system to replace animal models, including ex vivo, 2D culture, organoid, lab-on-a-chip, and 3D culture models. These models are described in terms of their structure, composition, and co-culture availability with microbiomes. Furthermore, we discuss the potential application for the aforementioned techniques to these in vitro models. The review concludes with a summary of intestine models from the viewpoint of current techniques as well as their main features, highlighting potential future developments and applications.
Collapse
Affiliation(s)
- Sang-Myung Jung
- Jeonbuk Branch Institute, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, South Korea
| | - Seonghun Kim
- Jeonbuk Branch Institute, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, South Korea.,Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, University of Science and Technology (UST), Daejeon, South Korea
| |
Collapse
|
1512
|
Establishment and resilience of transplanted gut microbiota in aged mice. iScience 2022; 25:103654. [PMID: 35024588 PMCID: PMC8733228 DOI: 10.1016/j.isci.2021.103654] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/29/2021] [Accepted: 12/13/2021] [Indexed: 01/04/2023] Open
Abstract
The maintenance of healthy and resilient gut microbiota is critical for the life quality and healthspan of the elderly. Fecal microbiota transplantation (FMT) has been increasingly used to restore healthy gut microbiota. We systemically studied the establishment and resilience of transplanted microbiota after autologous versus heterologous FMT in aged recipients. Gut microbiota of aged mice (20 months old) failed to restore their original diversity and composition over 8 weeks via spontaneous recovery after antibiotics treatment; in contrast, FMT using either autologous or heterologous (2 months old from a different vendor) donors facilitated the recovery successfully, established donor-like microbiota states, and affected host gene expression profile. Furthermore, the transplanted microbiota established by heterologous FMT is not resilient during chemical-induced colonic inflammation, in contrast to that of autologous FMT. Our findings highlighted the need to monitor the long-term stability of transplanted gut microbiota and to perform multiple FMT when necessary. Aged mice microbiota restores slowly after antibiotics treatment Both autologous and heterologous FMT facilitate microbiota restoration in aged mice FMT affects long-term homeostasis of gut metagenome and colon gene expression Established microbiota after heterologous FMT is not resilient against colitis
Collapse
|
1513
|
The Mediating Role of the Gut Microbiota in the Physical Growth of Children. Life (Basel) 2022; 12:life12020152. [PMID: 35207440 PMCID: PMC8880549 DOI: 10.3390/life12020152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota succession overlaps with intensive growth in infancy and early childhood. The multitude of functions performed by intestinal microbes, including participation in metabolic, hormonal, and immune pathways, makes the gut bacterial community an important player in cross-talk between intestinal processes and growth. Long-term disturbances in the colonization pattern may affect the growth trajectory, resulting in stunting or wasting. In this review, we summarize the evidence on the mediating role of gut microbiota in the mechanisms controlling the growth of children.
Collapse
|
1514
|
Advances in Microbiome-Derived Solutions and Methodologies Are Founding a New Era in Skin Health and Care. Pathogens 2022; 11:pathogens11020121. [PMID: 35215065 PMCID: PMC8879973 DOI: 10.3390/pathogens11020121] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/04/2022] Open
Abstract
The microbiome, as a community of microorganisms and their structural elements, genomes, metabolites/signal molecules, has been shown to play an important role in human health, with significant beneficial applications for gut health. Skin microbiome has emerged as a new field with high potential to develop disruptive solutions to manage skin health and disease. Despite an incomplete toolbox for skin microbiome analyses, much progress has been made towards functional dissection of microbiomes and host-microbiome interactions. A standardized and robust investigation of the skin microbiome is necessary to provide accurate microbial information and set the base for a successful translation of innovations in the dermo-cosmetic field. This review provides an overview of how the landscape of skin microbiome research has evolved from method development (multi-omics/data-based analytical approaches) to the discovery and development of novel microbiome-derived ingredients. Moreover, it provides a summary of the latest findings on interactions between the microbiomes (gut and skin) and skin health/disease. Solutions derived from these two paths are used to develop novel microbiome-based ingredients or solutions acting on skin homeostasis are proposed. The most promising skin and gut-derived microbiome interventional strategies are presented, along with regulatory, safety, industrial, and technical challenges related to a successful translation of these microbiome-based concepts/technologies in the dermo-cosmetic industry.
Collapse
|
1515
|
Junca H, Pieper DH, Medina E. The emerging potential of microbiome transplantation on human health interventions. Comput Struct Biotechnol J 2022; 20:615-627. [PMID: 35140882 PMCID: PMC8801967 DOI: 10.1016/j.csbj.2022.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/06/2022] [Accepted: 01/08/2022] [Indexed: 02/08/2023] Open
Abstract
The human microbiome has been the subject of intense research over the past few decades, in particular as a promising area for new clinical interventions. The microbiota colonizing the different body surfaces are of benefit for multiple physiological and metabolic processes of the human host and increasing evidence suggests an association between disturbances in the composition and functionality of the microbiota and several pathological conditions. This has provided a rationale for beneficial modulation of the microbiome. One approach being explored for modulating the microbiota in diseased individuals is transferring microbiota or microbiota constituents from healthy donors via microbiome transplantation. The great success of fecal microbiome transplantation for the treatment of Clostridioides difficile infections has encouraged the application of this procedure for the treatment of other diseases such as vaginal disorders via transplantation of vaginal microbiota, or of skin pathologies via the transplantation of skin microbiota. Microbiome modulation could even become a novel strategy for improving the efficacy of cancer therapies. This review discusses the principle, advantages and limitations of microbiome transplantation as well as different clinical contexts where microbiome transplantation has been applied.
Collapse
Affiliation(s)
- Howard Junca
- Microbial Interactions and Processes Research Group, Helmholtz-Zentrum für Infektionsforschung, Braunschweig, Germany
| | - Dietmar H. Pieper
- Microbial Interactions and Processes Research Group, Helmholtz-Zentrum für Infektionsforschung, Braunschweig, Germany
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz-Zentrum für Infektionsforschung, Braunschweig, Germany
| |
Collapse
|
1516
|
Malo ME, Frank C, Khokhoev E, Gorbunov A, Dontsov A, Garg R, Dadachova E. Mitigating effects of sublethal and lethal whole-body gamma irradiation in a mouse model with soluble melanin. JOURNAL OF RADIOLOGICAL PROTECTION : OFFICIAL JOURNAL OF THE SOCIETY FOR RADIOLOGICAL PROTECTION 2022; 42:011508. [PMID: 35037901 DOI: 10.1088/1361-6498/ac3dcf] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/25/2021] [Indexed: 05/27/2023]
Abstract
The field of radiation countermeasures is growing, however, currently there are no effective and non-toxic compounds which could be administered orally to the individuals post exposure to high doses of ionising radiation. The pigment melanin is ubiquitous through all kingdoms of life and provides selective advantage under radiation stress through its role as a chemical and physical shield, and its capacity to respond and react to exposures. Soluble allomelanin was administered to mice following whole-body exposure to lethal or sublethal doses of gamma radiation to determine its capacity to mitigate the effects of acute radiation syndrome, and its utility as a radiation countermeasure. Allomelanin has shown a trend to improve survival post an 8 Gy sublethal radiation exposure when administered up to 48 h post-irradiation. Furthermore, it improved median and overall survival to a 10 Gy lethal radiation exposure, specifically when administered at 24 h post-irradiation. Histological analysis on the jejunum region of the small intestine of this treatment group indicated that alterations of the mucosal and submucosal architecture, and disruption of the lymphatic system associated with lethal radiation exposure were mitigated when allomelanin was administered at 24 h post-irradiation. Based on this work soluble allomelanin derived from a fungal source could serve as an easily sourced, cost-effective, and viable countermeasure to accidental radiation exposure and merits further investigation.
Collapse
Affiliation(s)
- Mackenzie E Malo
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | - Connor Frank
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | | | | | - Alexander Dontsov
- Emanuel Institute of Biochemical Physics, Russian Academy of Science, Moscow, Russia
| | - Ravendra Garg
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
1517
|
Wang Y, van de Wouw M, Drogos L, Vaghef-Mehrabani E, Reimer RA, Tomfohr-Madsen L, Giesbrecht GF. Sleep and the gut microbiota in preschool-aged children. Sleep 2022; 45:6509073. [PMID: 35037059 PMCID: PMC9189981 DOI: 10.1093/sleep/zsac020] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/24/2021] [Indexed: 01/19/2023] Open
Abstract
Sleep plays a significant role in the mental and physical development of children. Emerging evidence in animals and human adults indicates a relationship between sleep and the gut microbiota; however, it is unclear whether the sleep of preschoolers during a key developmental period, associates with features of their gut microbiota. The objective of this study was to assess the relationship between sleep and gut microbiota in preschool-aged children (4.37 ± 0.48 years, n = 143). Sleep measures included total night-time sleep (TST), sleep efficiency (SE), and wake-time after sleep onset (WASO) assessed using actigraphy. Beta-diversity differences between children with low and high TST (p = .048) suggest gut microbiota community differences. Particularly, relative abundance of Bifidobacterium was higher in the high TST group and Bacteroides, was higher in children who had greater SE and less WASO (LDA score >2). In contrast, some Lachnospiraceae members including Blautia and Coprococcus 1 were associated with shorter night-time sleep duration and less efficiency, respectively. We also found a group of fecal metabolites, including specific neuroactive compounds and immunomodulating metabolites were associated with greater sleep efficiency and less time awake at night. Notably, tryptophan and its metabolizing products were higher in children who had higher SE or lower WASO (LDA score >2); concentration of propionate was higher in children with less WASO (p = .036). Overall, our results reveal a novel association between sleep and gut microbiota in preschool-aged children. Longer night-time sleep and greater sleep efficiency were associated with specific commensal bacteria that may regulate sleep through modulating neurotransmitter metabolism and the immune system.
Collapse
Affiliation(s)
- Yanan Wang
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
| | | | - Lauren Drogos
- Department of Psychology, University of Calgary, Calgary, AB, Canada,Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | | | - Raylene A Reimer
- Alberta Children’s Hospital Research Institute (ACHRI), Calgary, AB, Canada,Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Lianne Tomfohr-Madsen
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada,Department of Psychology, University of Calgary, Calgary, AB, Canada,Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada
| | - Gerald F Giesbrecht
- Corresponding author. Gerry Giesbrecht, Department of Psychology, University of Calgary, 2500 University Drive, NW, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
1518
|
Kim JE, Lee JY, Kang CH. Limosilactobacillus fermentum MG4295 Improves Hyperglycemia in High-Fat Diet-Induced Mice. Foods 2022; 11:foods11020231. [PMID: 35053962 PMCID: PMC8774940 DOI: 10.3390/foods11020231] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/15/2022] Open
Abstract
Hyperglycemia due to uncontrolled glucose regulation is widely known as cause of diabetes, non-alcoholic fatty liver disease (NAFLD), and other complications. NAFLD refers to a condition in which fat is excessively accumulated, whether inflamed or not, and has caused serious medical problems in recent years. The aim of this study was to explore the antihyperglycemia effects of Limosilactobacillus fermentum MG4295 (L. fermentum MG4295) in high-fat diet (HFD)-induced in vivo. We demonstrated the suitability of L. fermentum MG4295 as a probiotic by observing its stability, survivability, and proliferation under simulated gastrointestinal conditions, and safety, antibiotic susceptibility, hemolysis, and enzyme activity. The potential antihyperglycemic activity of L. fermentum MG4295 was investigated in an HFD and sugar-water-induced mouse model. Administration of this strain for 12 weeks showed an improved trend in glucose tolerance, insulin, alanine amino transferase, total cholesterol, low-density lipoprotein cholesterol, and glucagon-like peptide-1. Histopathological analysis revealed that L. fermentum MG4295 significantly reduced the histopathological scores of hepatic steatosis, inflammation, and hepatocellular hypertrophy in liver tissues and lipid content in adipose tissues. Administration of L. fermentum MG4295 upregulated IRS-1, AKT, and GLUT4 and downregulated G6Pc and PEPCK expression in liver and/or muscle tissues. Our results suggest that L. fermentum MG4295 can improve hyperglycemia. Furthermore, it can be used as a dietary functional supplement to manage blood glucose.
Collapse
|
1519
|
A More Diverse Cervical Microbiome Associates with Better Clinical Outcomes in Patients with Endometriosis: A Pilot Study. Biomedicines 2022; 10:biomedicines10010174. [PMID: 35052854 PMCID: PMC8774211 DOI: 10.3390/biomedicines10010174] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 02/04/2023] Open
Abstract
Infection-induced chronic inflammation is common in patients with endometriosis. Although microbial communities in the reproductive tracts of patients have been reported, little was known about their dynamic profiles during disease progression and complication development. Microbial communities in cervical mucus were collected by cervical swabs from 10 healthy women and 23 patients, and analyzed by 16S rRNA amplicon sequencing. The abundance, ecological relationships and functional networks of microbiota were characterized according to their prevalence, clinical stages, and clinical features including deeply infiltrating endometriosis (DIE), CA125, pain score and infertility. Cervical microbiome can be altered during endometriosis development and progression with a tendency of increased Firmicutes and decreased Actinobacteria and Bacteroidetes. Distinct from vaginal microbiome, upregulation of Lactobacillus, in combination with increased Streptococcus and decreased Dialister, was frequently associated with advanced endometriosis stages, DIE, higher CA125 levels, severe pain, and infertility. Significantly, reduced richness and diversity of cervical microbiome were detected in patients with more severe clinical symptoms. Clinical treatments against infertility can partially reverse the ecological balance of microbes through remodeling nutrition metabolism and transport and cell-cell/cell-matrix interaction. This study provides a new understanding on endometriosis development and a more diverse cervical microbiome may be beneficial for patients to have better clinical outcomes.
Collapse
|
1520
|
Salazar-Robles E, Kalantar-Zadeh K, Badillo H, Calderón-Juárez M, García-Bárcenas CA, Ledesma-Pérez PD, Lerma A, Lerma C. Association between severity of COVID-19 symptoms and habitual food intake in adult outpatients. BMJ Nutr Prev Health 2022; 4:469-478. [PMID: 35024547 PMCID: PMC8594975 DOI: 10.1136/bmjnph-2021-000348] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/05/2021] [Indexed: 12/15/2022] Open
Abstract
Objective To evaluate the association between habitual frequency of food intake of certain food groups during the COVID-19 pandemic and manifestations of COVID-19 symptoms in adult outpatients with suspected SARS-CoV-2 infection. Design We included 236 patients who attended an outpatient clinic for suspected COVID-19 evaluation. Severity of symptoms, habitual food intake frequency, demographics and Bristol chart scores were obtained before diagnostic confirmation with real-time reverse transcriptase PCR using nasopharyngeal swab. Results The results of the COVID-19 diagnostic tests were positive for 103 patients (44%) and negative for 133 patients (56%). In the SARS-CoV-2-positive group, symptom severity scores had significant negative correlations with habitual intake frequency of specific food groups. Multivariate binary logistic regression analysis adjusted for age, sex and occupation confirmed that SARS-CoV-2-positive patients showed a significant negative association between having higher symptom severity and the habitual intake frequency of ‘legumes’ and ‘grains, bread and cereals’. Conclusions Increase in habitual frequency of intake of ‘legumes’, and ‘grains, bread and cereals’ food groups decreased overall symptom severity in patients with COVID-19. This study provides a framework for designing a protective diet during the COVID-19 pandemic and also establishes a hypothesis of using a diet-based intervention in the management of SARS-CoV-2 infection, which may be explored in future studies.
Collapse
Affiliation(s)
- Elihud Salazar-Robles
- Centro Universitario de la Costa, Department of Medical Sciences, Universidad de Guadalajara, Puerto Vallarta, Mexico
| | - Kourosh Kalantar-Zadeh
- Chemical Engineering (Food Science and Technology), University of New South Wales, Sydney, New South Wales, Australia
| | - Humberto Badillo
- Centro de Salud Jalalpa el Grande, Secretaría de Salud de la Ciudad de México, Mexico City, Mexico
| | - Martín Calderón-Juárez
- Plan de Estudios Combinados en Medicina (PECEM), Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Cesar Alberto García-Bárcenas
- Centro Universitario de la Costa, Department of Medical Sciences, Universidad de Guadalajara, Puerto Vallarta, Mexico
| | | | - Abel Lerma
- Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca, Mexico
| | - Claudia Lerma
- Department of Electromechanical Instrumentation, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| |
Collapse
|
1521
|
Gut Microbiome and Organ Fibrosis. Nutrients 2022; 14:nu14020352. [PMID: 35057530 PMCID: PMC8781069 DOI: 10.3390/nu14020352] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 02/07/2023] Open
Abstract
Fibrosis is a pathological process associated with most chronic inflammatory diseases. It is defined by an excessive deposition of extracellular matrix proteins and can affect nearly every tissue and organ system in the body. Fibroproliferative diseases, such as intestinal fibrosis, liver cirrhosis, progressive kidney disease and cardiovascular disease, often lead to severe organ damage and are a leading cause of morbidity and mortality worldwide, for which there are currently no effective therapies available. In the past decade, a growing body of evidence has highlighted the gut microbiome as a major player in the regulation of the innate and adaptive immune system, with severe implications in the pathogenesis of multiple immune-mediated disorders. Gut microbiota dysbiosis has been associated with the development and progression of fibrotic processes in various organs and is predicted to be a potential therapeutic target for fibrosis management. In this review we summarize the state of the art concerning the crosstalk between intestinal microbiota and organ fibrosis, address the relevance of diet in different fibrotic diseases and discuss gut microbiome-targeted therapeutic approaches that are current being explored.
Collapse
|
1522
|
Food Additives, a Key Environmental Factor in the Development of IBD through Gut Dysbiosis. Microorganisms 2022; 10:microorganisms10010167. [PMID: 35056616 PMCID: PMC8780106 DOI: 10.3390/microorganisms10010167] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/04/2022] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Diet is a key environmental factor in inflammatory bowel disease (IBD) and, at the same time, represents one of the most promising therapies for IBD. Our daily diet often contains food additives present in numerous processed foods and even in dietary supplements. Recently, researchers and national authorities have been paying much attention to their toxicity and effects on gut microbiota and health. This review aims to gather the latest data focusing on the potential role of food additives in the pathogenesis of IBDs through gut microbiota modulation. Some artificial emulsifiers and sweeteners can induce the dysbiosis associated with an alteration of the intestinal barrier, an activation of chronic inflammation, and abnormal immune response accelerating the onset of IBD. Even if most of these results are retrieved from in vivo and in vitro studies, many artificial food additives can represent a potential hidden driver of gut chronic inflammation through gut microbiota alterations, especially in a population with IBD predisposition. In this context, pending the confirmation of these results by large human studies, it would be advisable that IBD patients avoid the consumption of processed food containing artificial food additives and follow a personalized nutritional therapy prescribed by a clinical nutritionist.
Collapse
|
1523
|
Amir A, Erez-Granat O, Braun T, Sosnovski K, Hadar R, BenShoshan M, Heiman S, Abbas-Egbariya H, Glick Saar E, Efroni G, Haberman Y. Gut microbiome development in early childhood is affected by day care attendance. NPJ Biofilms Microbiomes 2022; 8:2. [PMID: 35017536 PMCID: PMC8752763 DOI: 10.1038/s41522-021-00265-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/11/2021] [Indexed: 11/09/2022] Open
Abstract
The human gut microbiome develops during the first years of life, followed by a relatively stable adult microbiome. Day care attendance is a drastic change that exposes children to a large group of peers in a diverse environment for prolonged periods, at this critical time of microbial development, and therefore has the potential to affect microbial composition. We characterize the effect of day care on the gut microbial development throughout a single school year in 61 children from 4 different day care facilities, and in additional 24 age-matched home care children (n = 268 samples, median age of entering the study was 12 months). We show that day care attendance is a significant and impactful factor in shaping the microbial composition of the growing child, the specific daycare facility and class influence the gut microbiome, and each child becomes more similar to others in their day care. Furthermore, in comparison to home care children, day care children have a different gut microbial composition, with enrichment of taxa more frequently observed in older populations. Our results provide evidence that daycare may be an external factor that contributes to gut microbiome maturation and make-up in early childhood.
Collapse
Affiliation(s)
- Amnon Amir
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Ortal Erez-Granat
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Tzipi Braun
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Katya Sosnovski
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Rotem Hadar
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Marina BenShoshan
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Sophia Heiman
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Haya Abbas-Egbariya
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Efrat Glick Saar
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Gilat Efroni
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel
| | - Yael Haberman
- Sheba Medical Center, Tel-HaShomer, affiliated with the Tel-Aviv University, Tel-Aviv, Israel. .,Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
1524
|
Shoubridge AP, Fourrier C, Choo JM, Proud CG, Sargeant TJ, Rogers GB. Gut Microbiome Regulation of Autophagic Flux and Neurodegenerative Disease Risks. Front Microbiol 2022; 12:817433. [PMID: 35003048 PMCID: PMC8733410 DOI: 10.3389/fmicb.2021.817433] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/02/2021] [Indexed: 11/21/2022] Open
Abstract
The gut microbiome-brain axis exerts considerable influence on the development and regulation of the central nervous system. Numerous pathways have been identified by which the gut microbiome communicates with the brain, falling largely into the two broad categories of neuronal innervation and immune-mediated mechanisms. We describe an additional route by which intestinal microbiology could mediate modifiable risk for neuropathology and neurodegeneration in particular. Autophagy, a ubiquitous cellular process involved in the prevention of cell damage and maintenance of effective cellular function, acts to clear and recycle cellular debris. In doing so, autophagy prevents the accumulation of toxic proteins and the development of neuroinflammation, both common features of dementia. Levels of autophagy are influenced by a range of extrinsic exposures, including nutrient deprivation, infection, and hypoxia. These relationships between exposures and rates of autophagy are likely to be mediated, as least in part, by the gut microbiome. For example, the suppression of histone acetylation by microbiome-derived short-chain fatty acids appears to be a major contributor to upregulation of autophagic function. We discuss the potential contribution of the microbiome-autophagy axis to neurological health and examine the potential of exploiting this link to predict and prevent neurodegenerative diseases.
Collapse
Affiliation(s)
- Andrew P Shoubridge
- Microbiome and Host Health, Lifelong Health, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Infection and Immunity, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Célia Fourrier
- Lysosomal Health in Ageing, Hopwood Centre for Neurobiology, Lifelong Health, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Jocelyn M Choo
- Microbiome and Host Health, Lifelong Health, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Infection and Immunity, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Christopher G Proud
- Nutrition, Diabetes and Gut Health, Lifelong Health, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Timothy J Sargeant
- Lysosomal Health in Ageing, Hopwood Centre for Neurobiology, Lifelong Health, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Geraint B Rogers
- Microbiome and Host Health, Lifelong Health, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Infection and Immunity, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| |
Collapse
|
1525
|
Balcioglu H. The War between the Brain and the Body Caused by Stress. JOURNAL OF CLINICAL AND EXPERIMENTAL INVESTIGATIONS 2022. [DOI: 10.29333/jcei/11513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
1526
|
Microenvironmental Metabolites in the Intestine: Messengers between Health and Disease. Metabolites 2022; 12:metabo12010046. [PMID: 35050167 PMCID: PMC8778376 DOI: 10.3390/metabo12010046] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/04/2022] [Indexed: 02/01/2023] Open
Abstract
The intestinal mucosa is a highly absorptive organ and simultaneously constitutes the physical barrier between the host and a complex outer ecosystem. Intestinal epithelial cells (IECs) represent a special node that receives signals from the host and the environment and translates them into corresponding responses. Specific molecular communication systems such as metabolites are known to transmit information across the intestinal boundary. The gut microbiota or food-derived metabolites are extrinsic factors that influence the homeostasis of the intestinal epithelium, while mitochondrial and host-derived cellular metabolites determine the identity, fitness, and regenerative capacity of IECs. Little is known, however, about the role of intrinsic and extrinsic metabolites of IECs in the initiation and progression of pathological processes such as inflammatory bowel disease and colorectal cancer as well as about their impact on intestinal immunity. In this review, we will highlight the most recent contributions on the modulatory effects of intestinal metabolites in gut pathophysiology, with a particular focus on metabolites in promoting intestinal inflammation or colorectal tumorigenesis. In addition, we will provide a perspective on the role of newly identified oncometabolites from the commensal and opportunistic microbiota in shaping response and resistance to antitumor therapy.
Collapse
|
1527
|
Zhang P, Zheng L, Duan Y, Gao Y, Gao H, Mao D, Luo Y. Gut microbiota exaggerates triclosan-induced liver injury via gut-liver axis. JOURNAL OF HAZARDOUS MATERIALS 2022; 421:126707. [PMID: 34315018 DOI: 10.1016/j.jhazmat.2021.126707] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 06/26/2021] [Accepted: 07/18/2021] [Indexed: 06/13/2023]
Abstract
Triclosan (TCS) is an antimicrobial ingredient that has been widely incorporated in consumer products. TCS can cause hepatic damage by disturbing lipid metabolism, which is often accompanied with gut microbiota dysbiosis. However, the effects of gut microbiota on the TCS-induced liver injury are still unknown. Therefore, we constructed a mouse model based on five-week-old male C57BL/6 mice to investigate the effects of dietary TCS exposure (40 ppm) on liver injury. We found that TCS treatment for 4 weeks dramatically disturbed gut microbiota homeostasis, resulting in overproduction of lipopolysaccharides (LPS) and deficiency of secondary bile acids such as deoxycholic acid (DCA) and lithocholic acid (LCA). In addition, TCS considerably increased intestinal permeability by reducing mucus excretion and expression of tight junction proteins (ZO-1, occludin and claudin 4), which facilitated translocation of LPS. The LPS accumulation in blood contributed to liver injury by triggering the inflammatory response via TLR4 pathway. In summary, this study provides novel insights into the underlying mechanisms of TCS-associated liver injury induced by gut microbiota via the gut-liver axis, and contributes to better interpretation of the health impact of the environmentally emerging contaminant TCS.
Collapse
Affiliation(s)
- Peng Zhang
- College of Environmental Sciences and Engineering, Nankai University, Tianjin 300350, China; State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210046, China
| | - Liyang Zheng
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Yitao Duan
- College of Environmental Sciences and Engineering, Nankai University, Tianjin 300350, China
| | - Yuting Gao
- College of Environmental Sciences and Engineering, Nankai University, Tianjin 300350, China
| | - Huihui Gao
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Daqing Mao
- School of Medicine, Nankai University, Tianjin 300071, China.
| | - Yi Luo
- College of Environmental Sciences and Engineering, Nankai University, Tianjin 300350, China; State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210046, China.
| |
Collapse
|
1528
|
Sadri H, Aghaei M, Akbari V. Nisin induces apoptosis in cervical cancer cells via reactive oxygen species generation and mitochondrial membrane potential changes. Biochem Cell Biol 2022; 100:136-141. [PMID: 34986025 DOI: 10.1139/bcb-2021-0225] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nisin, an antimicrobial peptide produced by Lactococcus lactis, is widely used as a safe food preservative and has been recently attracting the attention of many researchers as a potential anticancer agent. The cytotoxicity of nisin against HeLa, OVCAR-3, SK-OV-3, and HUVEC cells was evaluated using MTT assay. The apoptotic effect of nisin was identified by Annexin-V/propidium iodide assay, and then it was further confirmed by western blotting analysis, mitochondrial membrane potential (ΔΨm) analysis, and reactive oxygen species (ROS) assay. The MTT assay showed concentration-dependent cytotoxicity of nisin towards cancer cell lines, with the IC50 values of 11.5-23 µM, but less toxicity against normal endothelial cells. Furthermore, treatment of cervical cancer cells with 12 µM nisin significantly (P<0.05) increased the Bax/Bcl-2 ratio (4.9-fold), reduced ΔΨm (70%), and elevated ROS levels (1.7-fold). These findings indicated that nisin might have anticancer and apoptogenic activities through mitochondrial dysfunction and oxidative stress damage in cervical cancer cells.
Collapse
Affiliation(s)
- Houri Sadri
- Isfahan University of Medical Sciences, 48455, Department of Pharmaceutical Biotechnology, Isfahan, Iran (the Islamic Republic of);
| | - Mahmoud Aghaei
- Isfahan University of Medical Sciences, 48455, Isfahan, Isfahan, Iran (the Islamic Republic of);
| | - Vajihe Akbari
- Isfahan University of Medical Sciences, 48455, Department of Pharmaceutical Biotechnology, Isfahan, Iran (the Islamic Republic of);
| |
Collapse
|
1529
|
Plesniarski A, Siddik AB, Su RC. The Microbiome as a Key Regulator of Female Genital Tract Barrier Function. Front Cell Infect Microbiol 2022; 11:790627. [PMID: 34976864 PMCID: PMC8719631 DOI: 10.3389/fcimb.2021.790627] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
The microbiome, the collection of microbial species at a site or compartment, has been an underappreciated realm of human health up until the last decade. Mounting evidence suggests the microbiome has a critical role in regulating the female genital tract (FGT) mucosa's function as a barrier against sexually transmitted infections (STIs) and pathogens. In this review, we provide the most recent experimental systems and studies for analyzing the interplay between the microbiome and host cells and soluble factors with an influence on barrier function. Key components, such as microbial diversity, soluble factors secreted by host and microbe, as well as host immune system, all contribute to both the physical and immunologic aspects of the FGT mucosal barrier. Current gaps in what is known about the effects of the microbiome on FGT mucosal barrier function are compared and contrasted with the literature of the gut and respiratory mucosa. This review article presents evidence supporting that the vaginal microbiome, directly and indirectly, contributes to how well the FGT protects against infection.
Collapse
Affiliation(s)
- Andrew Plesniarski
- JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Abu Bakar Siddik
- JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Ruey-Chyi Su
- JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
1530
|
Fonseca JR, Lucio M, Harir M, Schmitt-Kopplin P. Mining for Active Molecules in Probiotic Supernatant by Combining Non-Targeted Metabolomics and Immunoregulation Testing. Metabolites 2022; 12:metabo12010035. [PMID: 35050158 PMCID: PMC8778235 DOI: 10.3390/metabo12010035] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/23/2021] [Accepted: 12/25/2021] [Indexed: 11/16/2022] Open
Abstract
Chronic respiratory diseases such as asthma are highly prevalent in industrialized countries. As cases are expected to rise, there is a growing demand for alternative therapies. Our recent research on the potential benefits of probiotics suggests that they could prevent and reduce the symptoms of many diseases by modulating the host immune system with secreted metabolites. This article presents the first steps of the research that led us to identify the immunoregulatory bioactivity of the amino acid d-Trp reported in our previous study. Here we analyzed the cell culture metabolic footprinting of 25 commercially available probiotic strains to associate metabolic pathway activity information with their respective immune modulatory activity observed in vitro. Crude probiotic supernatant samples were processed in three different ways prior to untargeted analysis in positive and negative ionization mode by direct infusion ESI-FT-ICR-MS: protein precipitation and solid phase extraction (SPE) using HLB and CN-E sorbent cartridges. The data obtained were submitted to multivariate statistical analyses to distinguish supernatant samples into the bioactive and non-bioactive group. Pathway analysis using discriminant molecular features showed an overrepresentation of the tryptophan metabolic pathway for the bioactive supernatant class, suggesting that molecules taking part in that pathway may be involved in the immunomodulatory activity observed in vitro. This work showcases the potential of metabolomics to drive product development and novel bioactive compound discovery out of complex biological samples in a top-down manner.
Collapse
Affiliation(s)
- Juliano Roldan Fonseca
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (J.R.F.); (M.H.); (P.S.-K.)
| | - Marianna Lucio
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (J.R.F.); (M.H.); (P.S.-K.)
- Correspondence: ; Tel.: +49-89-3187-3775
| | - Mourad Harir
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (J.R.F.); (M.H.); (P.S.-K.)
| | - Philippe Schmitt-Kopplin
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (J.R.F.); (M.H.); (P.S.-K.)
- Analytical Food Chemistry, Technical University of Munich, 85354 Freising, Germany
| |
Collapse
|
1531
|
Shelton CD, Yoo W, Shealy NG, Torres TP, Zieba JK, Calcutt MW, Foegeding NJ, Kim D, Kim J, Ryu S, Byndloss MX. Salmonella enterica serovar Typhimurium uses anaerobic respiration to overcome propionate-mediated colonization resistance. Cell Rep 2022; 38:110180. [PMID: 34986344 PMCID: PMC8800556 DOI: 10.1016/j.celrep.2021.110180] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 10/23/2021] [Accepted: 12/06/2021] [Indexed: 12/18/2022] Open
Abstract
The gut microbiota benefits the host by limiting enteric pathogen expansion (colonization resistance), partially via the production of inhibitory metabolites. Propionate, a short-chain fatty acid produced by microbiota members, is proposed to mediate colonization resistance against Salmonella enterica serovar Typhimurium (S. Tm). Here, we show that S. Tm overcomes the inhibitory effects of propionate by using it as a carbon source for anaerobic respiration. We determine that propionate metabolism provides an inflammation-dependent colonization advantage to S. Tm during infection. Such benefit is abolished in the intestinal lumen of Salmonella-infected germ-free mice. Interestingly, S. Tm propionate-mediated intestinal expansion is restored when germ-free mice are monocolonized with Bacteroides thetaiotaomicron (B. theta), a prominent propionate producer in the gut, but not when mice are monocolonized with a propionate-production-deficient B. theta strain. Taken together, our results reveal a strategy used by S. Tm to mitigate colonization resistance by metabolizing microbiota-derived propionate Propionate, a short-chain fatty acid produced by the gut microbiota, is proposed to mediate colonization resistance against Salmonella enterica serovar Typhimurium (S. Tm). Here, Shelton et al. show that nitrate-dependent propionate metabolism fuels pathogen expansion in the inflamed gut, allowing S. Tm to overcome propionate’s inhibitory effects.
Collapse
Affiliation(s)
- Catherine D Shelton
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Woongjae Yoo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nicolas G Shealy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Teresa P Torres
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jacob K Zieba
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - M Wade Calcutt
- Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Nora J Foegeding
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Dajeong Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jinshil Kim
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea; Center for Food Bioconvergence, Seoul National University, Seoul 08826, Republic of Korea
| | - Sangryeol Ryu
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea; Center for Food Bioconvergence, Seoul National University, Seoul 08826, Republic of Korea
| | - Mariana X Byndloss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute of Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Digestive Disease Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Microbiome Innovation Center, Vanderbilt University, Nashville, TN 37235, USA.
| |
Collapse
|
1532
|
Naghizadeh M, Klaver L, Schönherz AA, Rani S, Dalgaard TS, Engberg RM. Impact of Dietary Sodium Butyrate and Salinomycin on Performance and Intestinal Microbiota in a Broiler Gut Leakage Model. Animals (Basel) 2022; 12:111. [PMID: 35011218 PMCID: PMC8749775 DOI: 10.3390/ani12010111] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/21/2021] [Accepted: 12/31/2021] [Indexed: 12/15/2022] Open
Abstract
Unfavorable alterations of the commensal gut microbiota and dysbacteriosis is a major health problem in the poultry industry. Understanding how dietary intervention alters the microbial ecology of broiler chickens is important for prevention strategies. A trial was conducted with 672 Ross 308 day-old male broilers fed a basic diet (no additives, control) or the basic diet supplemented with 500 mg/kg encapsulated butyrate or 68 mg/kg salinomycin. Enteric challenge was induced by inclusion of 50 g/kg rye in a grower diet and oral gavage of a 10 times overdose of a vaccine against coccidiosis. Compared to control and butyrate-supplemented birds, salinomycin supplementation alleviated growth depression. Compared to butyrate and non-supplemented control, salinomycin increased potentially beneficial Ruminococcaceae and reduced potentially pathogenic Enterobacteriaceae and counts of Lactobacillus salivarius and Clostridium perfringens. Further, salinomycin supplementation was accompanied by a pH decrease and succinic acid increase in ceca, while coated butyrate (0.5 g/kg) showed no or limited effects. Salinomycin alleviated growth depression and maintained intestinal homeostasis in the challenged broilers, while butyrate in the tested concentration showed limited effects. Thus, further investigations are required to identify optimal dietary inclusion rates for butyrate used as alternative to ionophore coccidiostats in broiler production.
Collapse
Affiliation(s)
- Mohammad Naghizadeh
- Department of Animal Science, Aarhus University, Blichers Allé 20, 8830 Tjele, Denmark; (L.K.); (A.A.S.); (S.R.); (T.S.D.)
| | - Laura Klaver
- Department of Animal Science, Aarhus University, Blichers Allé 20, 8830 Tjele, Denmark; (L.K.); (A.A.S.); (S.R.); (T.S.D.)
| | - Anna A. Schönherz
- Department of Animal Science, Aarhus University, Blichers Allé 20, 8830 Tjele, Denmark; (L.K.); (A.A.S.); (S.R.); (T.S.D.)
| | - Sundas Rani
- Department of Animal Science, Aarhus University, Blichers Allé 20, 8830 Tjele, Denmark; (L.K.); (A.A.S.); (S.R.); (T.S.D.)
- SA-Center for Interdisciplinary Research in Basic Sciences, Faculty of Basic and Applied Sciences, International Islamic University, Islamabad 44000, Pakistan
| | - Tina Sørensen Dalgaard
- Department of Animal Science, Aarhus University, Blichers Allé 20, 8830 Tjele, Denmark; (L.K.); (A.A.S.); (S.R.); (T.S.D.)
| | - Ricarda Margarete Engberg
- Department of Animal Science, Aarhus University, Blichers Allé 20, 8830 Tjele, Denmark; (L.K.); (A.A.S.); (S.R.); (T.S.D.)
| |
Collapse
|
1533
|
Siddiqui R, Maciver SK, Khan NA. Gut microbiome-immune system interaction in reptiles. J Appl Microbiol 2022; 132:2558-2571. [PMID: 34984778 DOI: 10.1111/jam.15438] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/12/2021] [Accepted: 12/31/2021] [Indexed: 12/17/2022]
Abstract
Reptiles are ectothermic amniotes in a world dominated by endotherms. Reptiles originated more than 300 million years ago and they often dwell in polluted environments which may expose them to pathogenic micro-organisms, radiation and/or heavy metals. Reptiles also possess greater longevity and may live much longer than similar-sized land mammals, for example, turtles, tortoises, crocodiles and tuatara are long-lived reptiles living up to 100 years or more. Many recent studies have emphasized the pivotal role of the gut microbiome on its host; thus, we postulated that reptilian gut microbiome and/or its metabolites and the interplay with their robust immune system may contribute to their longevity and overall hardiness. Herein, we discuss the composition of the reptilian gut microbiome, immune system-gut microbiome cross-talk, antimicrobial peptides, reptilian resistance to infectious diseases and cancer, ageing, as well the current knowledge of the genome and epigenome of these remarkable species. Preliminary studies have demonstrated that microbial gut flora of reptiles such as crocodiles, tortoises, water monitor lizard and python exhibit remarkable anticancer and antibacterial properties, as well as comprise novel gut bacterial metabolites and antimicrobial peptides. The underlying mechanisms between the gut microbiome and the immune system may hold clues to developing new therapies overall for health, and possible extrapolation to exploit the ancient defence systems of reptiles for Homo sapiens benefit.
Collapse
Affiliation(s)
- Ruqaiyyah Siddiqui
- College of Arts and Sciences, American University of Sharjah, Sharjah, United Arab Emirates
| | - Sutherland K Maciver
- Centre for Discovery Brain Science, Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Naveed Ahmed Khan
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
1534
|
Cutuli D, Giacovazzo G, Decandia D, Coccurello R. Alzheimer's disease and depression in the elderly: A trajectory linking gut microbiota and serotonin signaling. Front Psychiatry 2022; 13:1010169. [PMID: 36532180 PMCID: PMC9750201 DOI: 10.3389/fpsyt.2022.1010169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/28/2022] [Indexed: 12/05/2022] Open
Abstract
The occurrence of neuropsychiatric symptoms in the elderly is viewed as an early sign of subsequent cognitive deterioration and conversion from mild cognitive impairment to Alzheimer's disease. The prognosis in terms of both the severity and progression of clinical dementia is generally aggravated by the comorbidity of neuropsychiatric symptoms and decline in cognitive function. Undeniably, aging and in particular unhealthy aging, is a silent "engine of neuropathology" over which multiple changes take place, including drastic alterations of the gut microbial ecosystem. This narrative review evaluates the role of gut microbiota changes as a possible unifying concept through which the comorbidity of neuropsychiatric symptoms and Alzheimer's disease can be considered. However, since the heterogeneity of neuropsychiatric symptoms, it is improbable to describe the same type of alterations in the bacteria population observed in patients with Alzheimer's disease, as well as it is improbable that the variety of drugs used to treat neuropsychiatric symptoms might produce changes in gut bacterial diversity similar to that observed in the pathophysiology of Alzheimer's disease. Depression seems to be another very intriguing exception, as it is one of the most frequent neuropsychiatric symptoms in dementia and a mood disorder frequently associated with brain aging. Antidepressants (i.e., serotonin reuptake inhibitors) or tryptophan dietary supplementation have been shown to reduce Amyloid β-loading, reinstate microbial diversity and reduce the abundance of bacterial taxa dominant in depression and Alzheimer's disease. This review briefly examines this trajectory by discussing the dysfunction of gut microbiota composition, selected bacterial taxa, and alteration of tryptophan and serotonin metabolism/neurotransmission as overlapping in-common mechanisms involved with depression, Alzheimer's disease, and unhealthy aging.
Collapse
Affiliation(s)
- Debora Cutuli
- Department of Psychology, University of Rome La Sapienza, Rome, Italy.,European Center for Brain Research, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Giacomo Giacovazzo
- European Center for Brain Research, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Davide Decandia
- Department of Psychology, University of Rome La Sapienza, Rome, Italy.,European Center for Brain Research, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Roberto Coccurello
- European Center for Brain Research, Santa Lucia Foundation IRCCS, Rome, Italy.,Institute for Complex Systems (ISC), National Council of Research (CNR), Rome, Italy
| |
Collapse
|
1535
|
Sharma G, Khanna G, Sharma P, Deol PK, Kaur IP. Mechanistic Role of Probiotics in Improving Skin Health. PROBIOTIC RESEARCH IN THERAPEUTICS 2022:27-47. [DOI: 10.1007/978-981-16-5628-6_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
1536
|
Wang T, Ishikawa T, Sasaki M, Chiba T. Oral and Gut Microbial Dysbiosis and Non-alcoholic Fatty Liver Disease: The Central Role of Porphyromonas gingivalis. Front Med (Lausanne) 2022; 9:822190. [PMID: 35308549 PMCID: PMC8924514 DOI: 10.3389/fmed.2022.822190] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/19/2022] [Indexed: 02/05/2023] Open
Abstract
Gut microbiota play many important roles, such as the regulation of immunity and barrier function in the intestine, and are crucial for maintaining homeostasis in living organisms. The disruption in microbiota is called dysbiosis, which has been associated with various chronic inflammatory conditions, food allergies, colorectal cancer, etc. The gut microbiota is also affected by several other factors such as diet, antibiotics and other medications, or bacterial and viral infections. Moreover, there are some reports on the oral-gut-liver axis indicating that the disruption of oral microbiota affects the intestinal biota. Non-alcoholic fatty liver disease (NAFLD) is one of the systemic diseases caused due to the dysregulation of the oral-gut-liver axis. NAFLD is the most common liver disease reported in the developed countries. It includes liver damage ranging from simple steatosis to nonalcoholic steatohepatitis (NASH), cirrhosis, and cancer. Recently, accumulating evidence supports an association between NAFLD and dysbiosis of oral and gut microbiota. Periodontopathic bacteria, especially Porphyromonas gingivalis, have been correlated with the pathogenesis and development of NAFLD based on the clinical and basic research, and immunology. P. gingivalis was detected in the liver, and lipopolysaccharide from this bacteria has been shown to be involved in the progression of NAFLD, thereby indicating a direct role of P. gingivalis in NAFLD. Moreover, P. gingivalis induces dysbiosis of gut microbiota, which promotes the progression of NAFLD, through disrupting both metabolic and immunologic pathways. Here, we review the roles of microbial dysbiosis in NAFLD. Focusing on P. gingivalis, we evaluate and summarize the most recent advances in our understanding of the relationship between oral-gut microbiome symbiosis and the pathogenesis and progression of non-alcoholic fatty liver disease, as well as discuss novel strategies targeting both P. gingivalis and microbial dysbiosis.
Collapse
Affiliation(s)
- Ting Wang
- Division of Internal Medicine, Department of Oral Medicine, Iwate Medical University, Morioka, Japan
- Ting Wang
| | - Taichi Ishikawa
- Division of Molecular Microbiology, Department of Microbiology, Iwate Medical University, Morioka, Japan
| | - Minoru Sasaki
- Division of Molecular Microbiology, Department of Microbiology, Iwate Medical University, Morioka, Japan
| | - Toshimi Chiba
- Division of Internal Medicine, Department of Oral Medicine, Iwate Medical University, Morioka, Japan
- *Correspondence: Toshimi Chiba
| |
Collapse
|
1537
|
Moutsoglou DM. 2021 American Thoracic Society BEAR Cage Winning Proposal: Microbiome Transplant in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2022; 205:13-16. [PMID: 34758276 PMCID: PMC8865595 DOI: 10.1164/rccm.202108-1833ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
1538
|
Suess PM. Effects of Polyphosphate on Leukocyte Function. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2022; 61:131-143. [PMID: 35697939 DOI: 10.1007/978-3-031-01237-2_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Leukocytes are immune cells derived from hematopoietic stem cells of the bone marrow which play essential roles in inflammatory and immune responses. In contrast to anucleate platelets and erythrocytes, leukocytes are differentiated from other blood cells by the presence of a nucleus, and consist of monocytes, neutrophils, lymphocytes, basophils, and eosinophils. Factors released from platelets mediate immune responses in part by recruitment and regulation of leukocyte activity. Platelet dense granules contain the highly anionic polymer polyphosphate (polyP) with monomer chain lengths of approximately 60-100 phosphates long, which are released into the microenvironment upon platelet activation. Recent studies suggest that polyP released from platelets plays roles in leukocyte migration, recruitment, accumulation, differentiation, and activation. Furthermore, bacterial-derived polyphosphate, generally consisting of phosphate monomer lengths in the hundreds to thousands, appear to play a role in pathogenic evasion of the host immune response. This review will discuss the effects of host and pathogenic-derived polyphosphate on leukocyte function.
Collapse
Affiliation(s)
- Patrick M Suess
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
1539
|
Eng C, Jácome AA, Agarwal R, Hayat MH, Byndloss MX, Holowatyj AN, Bailey C, Lieu CH. A comprehensive framework for early-onset colorectal cancer research. Lancet Oncol 2022; 23:e116-e128. [PMID: 35090673 DOI: 10.1016/s1470-2045(21)00588-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 02/07/2023]
Abstract
Sporadic colorectal cancer has traditionally been viewed as a malignancy of older individuals. However, as the global prevalence of the disease diagnosed in younger individuals (<50 years) is expected to increase within the next decade, greater recognition is now being given to early-onset colorectal cancer. The cause of the predicted rise in prevalence is largely unknown and probably multifactorial. In this Series paper, we discuss the potential underlying causes of early-onset colorectal cancer, the role of energy balance, biological and genomic mechanisms (including microbiome aspects), and the treatment of early-onset colorectal cancer. We have specifically considered the psychosocial challenges of being diagnosed with colorectal cancer at younger age and the potential financial toxicity that might ensue. This Series paper brings a comprehensive review based on the existing data in the hopes of optimising the overall outcomes for patients with early-onset colorectal cancer.
Collapse
|
1540
|
Aan FJ, Glibetic N, Montoya-Uribe V, Matter ML. COVID-19 and the Microbiome: The Gut-Lung Connection. COMPREHENSIVE GUT MICROBIOTA 2022. [PMCID: PMC8131000 DOI: 10.1016/b978-0-12-819265-8.00048-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
1541
|
Liu J, Li X, Song F, Cui S, Lu W, Zhao J, Zhang H, Gu Z, Chen W. Dietary supplementation with low-dose xylooligosaccharide promotes the anti-Salmonella activity of probiotic Lactiplantibacillus plantarum ZS2058 in a murine model. Food Res Int 2022; 151:110858. [PMID: 34980394 DOI: 10.1016/j.foodres.2021.110858] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/22/2021] [Accepted: 12/01/2021] [Indexed: 11/17/2022]
Abstract
Oligosaccharides have been previously reported to cause an aggravation of Salmonella infection. In this study, we reduced the dietary supplementation of oligosaccharides (1% w/w) and studied their effects on the anti-Salmonella activity of probiotic Lactiplantibacillus plantarum (L. plantarum) ZS2058. The results showed that among all five studied oligosaccharides, only xylooligosaccharide (XOS) promoted the anti-Salmonella activity of L. plantarum ZS2058 by increasing the survival rate of the infected mice (66.7% vs. 53.3%). Further study revealed that XOS did not function synergistically with L. plantarum ZS2058, as XOS itself did not improve the survival rate of the infected mice. In an in vitro coculture system, XOS significantly promoted the antagonistic activity (92% increase) of L. plantarum ZS2058 against Salmonella. In Salmonella-infected mice, the combination of XOS and L. plantarum ZS2058 significantly increased the faecal content of short-chain fatty acids (SCFAs) and restored the production of proinflammatory cytokines. More importantly, XOS, L. plantarum ZS2058 and their combination changed the gut microbiota into distinct profiles. Linear Discriminant Analysis (LDA) effect size (LEfSe) analysis identified five taxa as marker bacteria for mice treated with a combination of XOS and L. plantarum ZS2058. In particular, Mucispirillum, which was previously reported to protect the host from Salmonella infection, was increased. Here, we showed that low dose XOS could promote the anti-Salmonella activity of the probiotic L. plantarum ZS2058. These results offer new opportunities to cope with this predominant food-borne pathogen with great efficiency and to lay a foundation for developing functional foods with anti-Salmonella potential.
Collapse
Affiliation(s)
- Junsheng Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Xiangfei Li
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, PR China
| | - Fanfen Song
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Shumao Cui
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Zhennan Gu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, PR China.
| |
Collapse
|
1542
|
Forssten SD, Ouwehand AC. Contribution of the Microbiota to Healthy Aging. COMPREHENSIVE GUT MICROBIOTA 2022:69-84. [DOI: 10.1016/b978-0-12-819265-8.00059-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
1543
|
Aganetti MA, Cruz CS, Galvão I, Engels DF, Ricci MF, Vieira AT. The Gut Microbiota and Immunopathophysiology. COMPREHENSIVE PHARMACOLOGY 2022:492-514. [DOI: 10.1016/b978-0-12-820472-6.00128-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
1544
|
Tissue-resident immunity in the female and male reproductive tract. Semin Immunopathol 2022; 44:785-799. [PMID: 35488095 PMCID: PMC9053558 DOI: 10.1007/s00281-022-00934-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/21/2022] [Indexed: 02/07/2023]
Abstract
The conception of how the immune system is organized has been significantly challenged over the last years. It became evident that not all lymphocytes are mobile and recirculate through secondary lymphoid organs. Instead, subsets of immune cells continuously reside in tissues until being reactivated, e.g., by a recurring pathogen or other stimuli. Consequently, the concept of tissue-resident immunity has emerged, and substantial evidence is now available to support its pivotal function in maintaining tissue homeostasis, sensing challenges and providing antimicrobial protection. Surprisingly, insights on tissue-resident immunity in the barrier tissues of the female reproductive tract are sparse and only slowly emerging. The need for protection from vaginal and amniotic infections, the uniqueness of periodic tissue shedding and renewal of the endometrial barrier tissue, and the demand for a tailored decidual immune adaptation during pregnancy highlight that tissue-resident immunity may play a crucial role in distinct compartments of the female reproductive tract. This review accentuates the characteristics of tissue-resident immune cells in the vagina, endometrium, and the decidua during pregnancy and discusses their functional role in modulating the risk for infertility, pregnancy complications, infections, or cancer. We here also review data published to date on tissue-resident immunity in the male reproductive organs, which is still a largely uncharted territory.
Collapse
|
1545
|
Jensen SK, Pærregaard SI, Brandum EP, Jørgensen AS, Hjortø GM, Jensen BAH. OUP accepted manuscript. Gastroenterol Rep (Oxf) 2022; 10:goac008. [PMID: 35291443 PMCID: PMC8915887 DOI: 10.1093/gastro/goac008] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/07/2022] [Accepted: 02/16/2022] [Indexed: 11/24/2022] Open
Abstract
Organismal survival depends on a well-balanced immune system and maintenance of host–microbe mutualism. The fine-tuned relationship between the gut microbiota and host immunity is constantly challenged by opportunistic bacteria testing the integrity of gastrointestinal (GI) barrier defenses. Barrier dysfunction reduces immunological tolerance towards otherwise innocuous microbes; it is a process that may instigate chronic inflammation. Paradoxically, sustained inflammation further diminishes barrier function, enabling bacterial translocation to extra-intestinal tissues. Once translocated, these bacteria stimulate systemic inflammation, thereby compromising organ function. While genetic risk alleles associate with barrier dysfunction, environmental stressors are key triggers of GI inflammation and associated breakdown in immune tolerance towards resident gut microbes. As dietary components dictate substrate availability, they also orchestrate microbiota composition and function, including migratory and pro-inflammatory potential, thus holding the capacity to fuel both GI and extra-intestinal inflammation. Additionally, Western diet consumption may weaken barrier defenses via curbed Paneth cell function and diminished host-defense peptide secretion. This review focuses on intervenable niches of host–microbe interactions and mucosal immunity with the ambition to provide a framework of plausible strategies to improve barrier function and regain tolerance in the inflamed mucosa via nutritional intervention.
Collapse
Affiliation(s)
- Sune K Jensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simone I Pærregaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emma P Brandum
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Astrid S Jørgensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gertrud M Hjortø
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Benjamin A H Jensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Corresponding author. Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, Build. 22.5.39, Copenhagen N 2200, Denmark. Tel: +45-35330188;
| |
Collapse
|
1546
|
Alam MJ, Puppala V, Uppulapu SK, Das B, Banerjee SK. Human microbiome and cardiovascular diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 192:231-279. [PMID: 36280321 DOI: 10.1016/bs.pmbts.2022.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
1547
|
Kim SJ, Khadka D, Seo JH. Interplay between Solid Tumors and Tumor Microenvironment. Front Immunol 2022; 13:882718. [PMID: 35707536 PMCID: PMC9189309 DOI: 10.3389/fimmu.2022.882718] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/04/2022] [Indexed: 12/11/2022] Open
Abstract
Over the past few decades, basic studies aimed at curing patients with cancer have been constantly evolving. A myriad of mechanistic studies on physiological changes and related factors in tumor growth and metastasis have been reported. Recently, several studies have been considerate to how tumors adapt to unfavorable environments, such as glucose deprivation, oxidative stress, hypoxic conditions, and immune responses. Tumors attempt to adapt to unfavorable environments with genetic or non-genetic changes, the alteration of metabolic signals, or the reconfiguration of their environment through migration to other organs. One of the distinct features in solid tumors is heterogeneity because their environments vary due to the characteristics of colony growth. For this reason, researchers are paying attention to the communication between growing tumors and neighboring environments, including stromal cells, immune cells, fibroblasts, and secreted molecules, such as proteins and RNAs. During cancer survival and progression, tumor cells undergo phenotype and molecular changes collectively referred to as cellular plasticity, which result from microenvironment signals, genetics and epigenetic alterations thereby contributing to tumor heterogeneity and therapy response. In this review, we herein discuss the adaptation process of tumors to adverse environments via communication with neighboring cells for overcoming unfavorable growth conditions. Understanding the physiology of these tumors and their communication with the tumor environment can help to develop promising tumor treatment strategies.
Collapse
Affiliation(s)
- Seung-Jin Kim
- Department of Biochemistry, College of Natural Sciences, and Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, South Korea
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, Chuncheon, South Korea
| | - Dipendra Khadka
- NADIANBIO Ltd., Wonkwang University, Business Incubation Center R201-1, Iksan, South Korea
| | - Jae Ho Seo
- Department of Biochemistry, Wonkwang University School of Medicine, Iksan, South Korea
- Sarcopenia Total Solution Center, Wonkwang University School of Medicine, Iksan, South Korea
- *Correspondence: Jae Ho Seo,
| |
Collapse
|
1548
|
Zhao L, Cho WC, Luo JL. Exploring the patient-microbiome interaction patterns for pan-cancer. Comput Struct Biotechnol J 2022; 20:3068-3079. [PMID: 35782745 PMCID: PMC9233187 DOI: 10.1016/j.csbj.2022.06.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/06/2022] [Accepted: 06/06/2022] [Indexed: 11/03/2022] Open
Abstract
Cancer subtype-specific sets of microbiomes, making pan-cancer heterogeneity at the microbial level. Approximately 60% of the untreated cancer patients have experienced microbial composition changes in their tumor tissues. Colorectal cancer (CRC) was largely composed of two subtypes (S4 and S6) driven by different microbial profiles. The identified seven pan-cancer subtypes with 424 subtype-specific microbial signatures will help us find new therapeutic targets and better treatment strategies for cancer patients.
Microbes play important roles in human health and disease. Immunocompromised cancer patients are more vulnerable to getting microbial infections. Regions of hypoxia and acidic tumor microenvironment shape the microbial community diversity and abundance. Each cancer has its own microbiome, making cancer-specific sets of microbiomes. High-throughput profiling technologies provide a culture-free approach for microbial profiling in tumor samples. Microbial compositional data was extracted and examined from the TCGA unmapped transcriptome data. Biclustering, correlation, and statistical analyses were performed to determine the seven patient-microbe interaction patterns. These two-dimensional patterns consist of a group of microbial species that show significant over-representation over the 7 pan-cancer subtypes (S1-S7), respectively. Approximately 60% of the untreated cancer patients have experienced tissue microbial composition and functional changes between subtypes and normal controls. Among these changes, subtype S5 had loss of microbial diversity as well as impaired immune functions. S1, S2, and S3 had been enriched with microbial signatures derived from the Gammaproteobacteria, Actinobacteria and Betaproteobacteria, respectively. Colorectal cancer (CRC) was largely composed of two subtypes, namely S4 and S6, driven by different microbial profiles. S4 patients had increased microbial load, and were enriched with CRC-related oncogenic pathways. S6 CRC together with other cancer patients, making up almost 40% of all cases were classified into the S6 subtype, which not only resembled the normal control’s microbiota but also retained their original “normal-like” functions. Lastly, the S7 was a rare and understudied subtype. Our study investigated the pan-cancer heterogeneity at the microbial level. The identified seven pan-cancer subtypes with 424 subtype-specific microbial signatures will help us find new therapeutic targets and better treatment strategies for cancer patients.
Collapse
|
1549
|
SANTANA LUCASA, CONCEIÇÃO ROBERTYN, REZENDE-SILVA ERIKA, BARBOSA BRENOF, BORGES LYSANDROP, TRENTO CLEVERSONLUCIANO. Is dysbiosis a preponderant factor for the occurrence of oral lesions in SARS-CoV- 2 positive patients? A critical analysis. AN ACAD BRAS CIENC 2022; 94:e20211635. [DOI: 10.1590/0001-3765202220211635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/26/2022] [Indexed: 11/22/2022] Open
|
1550
|
Kim S, Han SY, Lee J, Kim NR, Lee BR, Kim H, Kwon M, Ahn K, Noh Y, Kim SJ, Lee P, Kim D, Kim BE, Kim J. Bifidobacterium longum and Galactooligosaccharide Improve Skin Barrier Dysfunction and Atopic Dermatitis-like Skin. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2022; 14:549-564. [PMID: 36174995 PMCID: PMC9523416 DOI: 10.4168/aair.2022.14.5.549] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 11/20/2022]
Abstract
Purpose The beneficial effects of a combination therapy using Bifidobacterium longum and galactooligosaccharide (GOS) for the treatment of atopic dermatitis (AD) have not been elucidated. Methods Gene expressions of interleukin (IL)-4 and IL-13 from peripheral blood mononuclear cells and fecal abundance of B. longum from 12-month-old infants were evaluated. Human primary epidermal keratinocytes (HEKs) and hairless mice were treated with B. longum, GOS, B. longum-derived extracellular vesicles (BLEVs), dinitrochlorobenzene (DNCB), or a synbiotic mixture of B. longum and GOS. Expression of epidermal barrier proteins and cytokines as well as serum immunoglobulin E (IgE) levels were analyzed in HEKs and mice. Dermatitis scores, transepidermal water loss (TEWL), epidermal thickness, and fecal B. longum abundance were evaluated in mice. Results Fecal abundance of B. longum was negatively correlated with blood IL-13 expression in infants. B. longum or BLEVs increased expression of filaggrin (FLG) and loricrin (LOR) in HEKs. B. longum increased the efficacy of GOS to upregulate FLG and LOR expressions in HEKs. Oral administration of GOS increased fecal abundance of B. longum in mice. Oral administration of B. longum attenuated DNCB-induced skin inflammation, abnormal TEWL, AD-like skin, and deficiency of epidermal barrier proteins. Moreover, the combination of B. longum and GOS showed greater effects to improve DNCB-induced skin inflammation, abnormal TEWL, AD-like skin, serum IgE levels, IL-4 over-expression, and the deficiency of epidermal barrier proteins than the administration of B. longum alone. Conclusions B. longum and GOS improve DNCB-induced skin barrier dysfunction and AD-like skin.
Collapse
Affiliation(s)
- Sukyung Kim
- Department of Pediatrics, Hallym University Dongtan Sacred Heart Hospital, Hallym University School of Medicine, Hwaseong, Korea
| | - Song-Yi Han
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jinyoung Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Na-Rae Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Bo Ra Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyunmi Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Mijeoung Kwon
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kangmo Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | - Phyrim Lee
- Dairyteam, Lotte R&D Center, Seoul, Korea
| | - Dongki Kim
- Department of Translational Research and Cellular Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Byung Eui Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Jihyun Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|