601
|
Senizza A, Rocchetti G, Mosele JI, Patrone V, Callegari ML, Morelli L, Lucini L. Lignans and Gut Microbiota: An Interplay Revealing Potential Health Implications. Molecules 2020; 25:E5709. [PMID: 33287261 PMCID: PMC7731202 DOI: 10.3390/molecules25235709] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/15/2022] Open
Abstract
Plant polyphenols are a broad group of bioactive compounds characterized by different chemical and structural properties, low bioavailability, and several in vitro biological activities. Among these compounds, lignans (a non-flavonoid polyphenolic class found in plant foods for human nutrition) have been recently studied as potential modulators of the gut-brain axis. In particular, gut bacterial metabolism is able to convert dietary lignans into therapeutically relevant polyphenols (i.e., enterolignans), such as enterolactone and enterodiol. Enterolignans are characterized by various biologic activities, including tissue-specific estrogen receptor activation, together with anti-inflammatory and apoptotic effects. However, variation in enterolignans production by the gut microbiota is strictly related to both bioaccessibility and bioavailability of lignans through the entire gastrointestinal tract. Therefore, in this review, we summarized the most important dietary source of lignans, exploring the interesting interplay between gut metabolites, gut microbiota, and the so-called gut-brain axis.
Collapse
Affiliation(s)
- Alice Senizza
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (V.P.); (M.L.C.); (L.M.); (L.L.)
| | - Gabriele Rocchetti
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (V.P.); (M.L.C.); (L.M.); (L.L.)
| | - Juana I. Mosele
- Cátedra de Fisicoquímica, Departamento de Química Analítica y Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires C1113AAD, Argentina;
| | - Vania Patrone
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (V.P.); (M.L.C.); (L.M.); (L.L.)
| | - Maria Luisa Callegari
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (V.P.); (M.L.C.); (L.M.); (L.L.)
| | - Lorenzo Morelli
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (V.P.); (M.L.C.); (L.M.); (L.L.)
| | - Luigi Lucini
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (V.P.); (M.L.C.); (L.M.); (L.L.)
| |
Collapse
|
602
|
Takayama S, Kikuchi A, Makino T, Kainuma M, Namiki T, Ito T. Basic pharmacological mechanisms and clinical evidence of the efficacy of hochuekkito against infectious diseases and its potential for use against COVID‐19. TRADITIONAL & KAMPO MEDICINE 2020. [PMCID: PMC7753421 DOI: 10.1002/tkm2.1264] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Background Hochuekkito extract (HET) has multiple effects through the digestive and immune systems, including for acute viral infection and chronic inflammation. We review basic pharmacological and clinical researches of HET and discuss the effects of HET against the pandemic COVID‐19. Methods We reviewed pharmacological studies from 1996 to 30 April 2020 that used experimental animals orally treated with HET and randomized controlled trials (RCTs) from 2000 to 30 April 2020. Results Altogether, 64 pharmacological studies reported immuno‐stimulatory effects against infection and cancer, immuno‐modulative effects against allergy and some inflammatory diseases, and ameliorating effects against exhaustion and frailty. Nine RCTs showed improvement of pulmonary Mycobacterium avium complex disease on chest X‐ray; improved systemic inflammation, nutrition, and quality of life of patients with chronic obstructive pulmonary disease and a decrease in the number getting common cold and exacerbations; reduction of soluble interleukin‐2 receptor and the serum cortisol concentration of postoperative patients; a reduction of the incidence of inflammatory complications and C‐reactive protein elevation after cerebrovascular disease; a reduction of the volume of steroid and tacrolimus during the treatment of atopic dermatitis; a healing effect for intractable chronic wounds; improvement of the physical status of elderly weak patients; and improvement of the fatigue level of cancer patients. Conclusion CODIV‐19 is characterized by high risk for the aged and people with other disease complications, cytokine hyperactivity in the severe stage, and sequelae in the recovery stage. Considering the immune‐stimulative/modulative effects of HET on inflammatory conditions and against exhaustion and frailty, it may be useful for prevention, treatment, and recovery from COVID‐19.
Collapse
Affiliation(s)
- Shin Takayama
- Department of Kampo Medicine Tohoku University Hospital Sendai Japan
- Department of Education and Support for Regional Medicine Tohoku University Hospital Sendai Japan
- Department of Kampo and Integrative Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Akiko Kikuchi
- Department of Kampo Medicine Tohoku University Hospital Sendai Japan
- Department of Education and Support for Regional Medicine Tohoku University Hospital Sendai Japan
- Department of Kampo and Integrative Medicine Tohoku University Graduate School of Medicine Sendai Japan
| | - Toshiaki Makino
- Department of Pharmacognosy Graduate School of Pharmaceutical Sciences, Nagoya City University Nagoya Japan
| | - Mosaburo Kainuma
- Community Medicine Education Unit Graduate School of Medical Science, Kyushu University Fukuoka Japan
| | - Takao Namiki
- Department of Japanese‐Oriental (Kampo) Medicine Graduate School of Medicine, Chiba University Chiba Japan
| | | |
Collapse
|
603
|
Faecal microbiota transplantation alleviates symptoms of depression in individuals with irritable bowel syndrome: A case series. MEDICINE IN MICROECOLOGY 2020. [DOI: 10.1016/j.medmic.2020.100029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
604
|
Moon J, Ryu JS, Kim JY, Im SH, Kim MK. Effect of IRT5 probiotics on dry eye in the experimental dry eye mouse model. PLoS One 2020; 15:e0243176. [PMID: 33259525 PMCID: PMC7707591 DOI: 10.1371/journal.pone.0243176] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/16/2020] [Indexed: 12/24/2022] Open
Abstract
Objective To investigate the clinical effects of IRT5 probiotics in the environmental dry eye model. Methods Eight week old male C57BL/6 mice were randomly divided into two groups; control group (n = 16) received oral gavage of 300 μL phosphate-buffered saline (PBS) alone once daily, IRT5 group (n = 9) received oral gavage of 1 x 109 CFU IRT5 probiotics powder in 300 μL PBS once daily, both groups for 11 to 12 days. Simultaneously, all mice underwent dry eye induction. Tear secretion, corneal staining and conjunctival goblet cell density were evaluated. Quantative real-time polymerase chain reaction (RT-PCR) for inflammation-related markers was performed. 16S ribosomal RNA of fecal microbiome was analyzed and compositional difference, alpha and beta diversities were assessed. Results There was no difference in NEI score but significant increase in tear secretion was observed in IRT5 group (p < 0.001). There was no significant difference in goblet cell density between groups. Quantative RT-PCR of cornea and conjunctiva revealed increased TNF-α expression in IRT5 group (p < 0.001) whereas other markers did not significantly differ from control. IRT5 group had significantly increased species diversity by Shannon index (p = 0.041). Beta diversity of genus by UniFrac principle coordinates analysis showed significant distance between groups (p = 0.001). Compositional differences between groups were observed and some were significantly associated with tear secretion. Multivariate linear regression analysis revealed Christensenellaceae (p = 0.009), Lactobacillus Helveticus group (p = 0.002) and PAC001797_s (p = 0.011) to strongly influence tear secretion. Conclusion In experimental dry eye model, IRT5 probiotics treatment partially improves experimental dry eye by increasing tear secretion which was associated with and influenced by the change in intestinal microbiome. Also, intestinal microbiome may affect the lacrimal gland through a different mechanism other than regulating inflammation.
Collapse
Affiliation(s)
- Jayoon Moon
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Laboratory of Ocular Regenerative Medicine and Immunology, Seoul Artificial Eye Center, Seoul National University Hospital Biomedical Research Institute, Seoul, Republic of Korea
| | - Jin Suk Ryu
- Laboratory of Ocular Regenerative Medicine and Immunology, Seoul Artificial Eye Center, Seoul National University Hospital Biomedical Research Institute, Seoul, Republic of Korea
| | - Jun Yeop Kim
- Laboratory of Ocular Regenerative Medicine and Immunology, Seoul Artificial Eye Center, Seoul National University Hospital Biomedical Research Institute, Seoul, Republic of Korea
| | - Sin-Hyeog Im
- Division of Integrative Biosciences and Biotechnology, Department of Life Science, Pohang University of Science and Technology, Pohang, Republic of Korea
- ImmunoBiome Inc. POSTECH Biotech Center, Pohang, Republic of Korea
| | - Mee Kum Kim
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Laboratory of Ocular Regenerative Medicine and Immunology, Seoul Artificial Eye Center, Seoul National University Hospital Biomedical Research Institute, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
605
|
Wright CD, Nelson CI, Brumbaugh JT, McNeil DW. The Role of Distress Tolerance as a Potential Mechanism Between Anxiety Sensitivity and Gut-Specific Anxiety. Int J Behav Med 2020; 27:717-725. [PMID: 32583317 PMCID: PMC7720241 DOI: 10.1007/s12529-020-09912-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND The link between anxiety/fear and gut dysfunction has been robustly documented in both physical and mental health literatures. The current study explored distress tolerance as a potential mechanism in the relation between anxiety sensitivity and gut-specific anxiety. METHOD A cross-sectional sample of 828 adults completed measures of distress tolerance, gut-specific anxiety, and anxiety sensitivity. Multiple linear regression analyses were conducted to determine variable associations, including potential mediating factors. RESULTS The results demonstrated a bidirectional relation between anxiety sensitivity and gut-specific anxiety (ß = 0.23, p < 0.001; ß = 0.22, p < 0.001). Findings suggest distress tolerance is a significant mediator that may partially explain the relation between gut-specific anxiety and anxiety sensitivity more broadly (ß = 0.11, CI [0.07-0.14]). Mediation results were consistent when individual subscales of distress tolerance or anxiety sensitivity were incorporated. CONCLUSION The outcome of the present study merits additional examination of the psychosomatic nature of distress tolerance as a potential clinical target for individuals with both anxiety and gut-related disorders.
Collapse
Affiliation(s)
- Casey D Wright
- Department of Psychology, Eberly College of Arts and Sciences, West Virginia University, 53 Campus Drive, PO Box 6040, Morgantown, WV, 26505, USA.
| | - Cecelia I Nelson
- Department of Psychology, Eberly College of Arts and Sciences, West Virginia University, 53 Campus Drive, PO Box 6040, Morgantown, WV, 26505, USA
| | - Jamey T Brumbaugh
- Department of Psychology, Eberly College of Arts and Sciences, West Virginia University, 53 Campus Drive, PO Box 6040, Morgantown, WV, 26505, USA
| | - Daniel W McNeil
- Department of Psychology, Eberly College of Arts and Sciences, West Virginia University, 53 Campus Drive, PO Box 6040, Morgantown, WV, 26505, USA.
- Department of Dental and Rural Practice, School of Dentistry, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
606
|
Hung TKW, Dong TS, Chen Z, Elashoff D, Sinsheimer JS, Jacobs JP, Lagishetty V, Vora P, Stains J, Mayer EA, Gupta A. Understanding the Heterogeneity of Obesity and the Relationship to the Brain-Gut Axis. Nutrients 2020; 12:nu12123701. [PMID: 33266058 PMCID: PMC7761087 DOI: 10.3390/nu12123701] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/16/2020] [Accepted: 11/20/2020] [Indexed: 12/26/2022] Open
Abstract
Obesity is best understood as a multifactorial metabolic imbalances disorder. In a cross-sectional study, we aimed to explore sociodemographic and dietary determinants of obesity in relation to brain-gut homeostasis among overweight and obese individuals. Multivariate logistic regression models were used to examine obesity and its association with sociodemographic and dietary factors. Biological variables examined included the gut microbiome, fecal amino acid metabolites and brain structural volumes. Among 130 participants, there were higher odds of obesity if individuals were Hispanic (adjusted odds ratio (aOR) 1.56, p = 0.014). Compared to non-Hispanics, Hispanics differed in gut microbial composition (p = 0.046) with lower microbial species richness (Chao1) (p = 0.032) and evenness (Shannon) (p = 0.0029). Fourteen of the twenty fecal amino acids including branch-chain- and aromatic- amino acids were increased among Hispanics (q < 0.05). Brain structural volumes in reward regions were decreased in Hispanics (pallidum, q = 0.036; brainstem, q = 0.011). Correlation patterns suggest complex brain-gut interactions differ by Hispanic ethnicity. In conclusion, Hispanics expressed a unique brain-gut microbial signature, which was associated with obesity despite sociodemographic and dietary differences. Addressing ethnic disparities guided by biologic phenotypes may unlock novel understanding of obesity heterogeneity and treatment strategies.
Collapse
Affiliation(s)
- Tony K. W. Hung
- Division of Hematology and Oncology, University of California, Los Angeles, CA 90095, USA; (T.K.W.H.); (T.S.D.); (Z.C.); (D.E.); (J.P.J.); (V.L.); (P.V.); (J.S.); (E.A.M.)
- David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Tien S. Dong
- Division of Hematology and Oncology, University of California, Los Angeles, CA 90095, USA; (T.K.W.H.); (T.S.D.); (Z.C.); (D.E.); (J.P.J.); (V.L.); (P.V.); (J.S.); (E.A.M.)
- David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, UCLA, Los Angeles, CA 90095, USA
- UCLA Microbiome Center, Los Angeles, CA 90095, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90095, USA
| | - Zixi Chen
- Division of Hematology and Oncology, University of California, Los Angeles, CA 90095, USA; (T.K.W.H.); (T.S.D.); (Z.C.); (D.E.); (J.P.J.); (V.L.); (P.V.); (J.S.); (E.A.M.)
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA, Los Angeles, CA 90095, USA
| | - David Elashoff
- Division of Hematology and Oncology, University of California, Los Angeles, CA 90095, USA; (T.K.W.H.); (T.S.D.); (Z.C.); (D.E.); (J.P.J.); (V.L.); (P.V.); (J.S.); (E.A.M.)
- Department of Computational Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, CA 90095, USA
| | - Janet S. Sinsheimer
- Department of Computational Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Jonathan P. Jacobs
- Division of Hematology and Oncology, University of California, Los Angeles, CA 90095, USA; (T.K.W.H.); (T.S.D.); (Z.C.); (D.E.); (J.P.J.); (V.L.); (P.V.); (J.S.); (E.A.M.)
- David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, UCLA, Los Angeles, CA 90095, USA
- UCLA Microbiome Center, Los Angeles, CA 90095, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90095, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA, Los Angeles, CA 90095, USA
| | - Venu Lagishetty
- Division of Hematology and Oncology, University of California, Los Angeles, CA 90095, USA; (T.K.W.H.); (T.S.D.); (Z.C.); (D.E.); (J.P.J.); (V.L.); (P.V.); (J.S.); (E.A.M.)
- UCLA Microbiome Center, Los Angeles, CA 90095, USA
| | - Priten Vora
- Division of Hematology and Oncology, University of California, Los Angeles, CA 90095, USA; (T.K.W.H.); (T.S.D.); (Z.C.); (D.E.); (J.P.J.); (V.L.); (P.V.); (J.S.); (E.A.M.)
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA, Los Angeles, CA 90095, USA
| | - Jean Stains
- Division of Hematology and Oncology, University of California, Los Angeles, CA 90095, USA; (T.K.W.H.); (T.S.D.); (Z.C.); (D.E.); (J.P.J.); (V.L.); (P.V.); (J.S.); (E.A.M.)
- David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, UCLA, Los Angeles, CA 90095, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA, Los Angeles, CA 90095, USA
| | - Emeran A. Mayer
- Division of Hematology and Oncology, University of California, Los Angeles, CA 90095, USA; (T.K.W.H.); (T.S.D.); (Z.C.); (D.E.); (J.P.J.); (V.L.); (P.V.); (J.S.); (E.A.M.)
- David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, UCLA, Los Angeles, CA 90095, USA
- UCLA Microbiome Center, Los Angeles, CA 90095, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA, Los Angeles, CA 90095, USA
- Ahmanson-Lovelace Brain Mapping Center, UCLA, Los Angeles, CA 90095, USA
| | - Arpana Gupta
- Division of Hematology and Oncology, University of California, Los Angeles, CA 90095, USA; (T.K.W.H.); (T.S.D.); (Z.C.); (D.E.); (J.P.J.); (V.L.); (P.V.); (J.S.); (E.A.M.)
- David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, UCLA, Los Angeles, CA 90095, USA
- UCLA Microbiome Center, Los Angeles, CA 90095, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA, Los Angeles, CA 90095, USA
- Correspondence:
| |
Collapse
|
607
|
Maitre Y, Micheneau P, Delpierre A, Mahalli R, Guerin M, Amador G, Denis F. Did the Brain and Oral Microbiota Talk to Each Other? A Review of the Literature. J Clin Med 2020; 9:jcm9123876. [PMID: 33260581 PMCID: PMC7760025 DOI: 10.3390/jcm9123876] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023] Open
Abstract
This systematic review aims to investigate the role of the oral microbiome in the pathophysiology of mental health disorders and to appraise the methodological quality of research of the oral–brain axis which is a growing interest area. The PRISMA guideline was adopted, to carry out an electronic search through the MEDLINE database, to identify studies that have explored the role of the oral microbiome in the pathophysiology of mental health disorders published from 2000 up to June 2020. The search resulted in 140 records; after exclusions, a total of 22 papers were included in the present review. In accordance with the role of the oral microbiome in the pathophysiology of mental disorders, four mental disorders were identified: Alzheimer’s disease, dementia, and cognitive disorders; autism spectrum disorder; Down’s syndrome and mental retardation; and Bipolar disorders. Studies argue for correlations between oral microbiota and Alzheimer’s disease, autism spectrum disorders, Down’s syndrome, and bipolar disorders. This field is still under-studied, and studies are needed to clarify the biological links and interconnections between the oral microbiota and the pathophysiology of all mental health disorders. Researchers should focus their efforts to develop research on the oral–brain axis in the future.
Collapse
Affiliation(s)
- Yoann Maitre
- Emergency Department, Montpellier University Hospital, 34259 Montpellier, France;
- EA 2415, Aide à la Décision pour une Médecine Personnalisée, Université de Montpellier, 34090 Montpellier, France
| | - Pierre Micheneau
- Department of Odontology, Tours University Hospital, 37261 Tours, France; (P.M.); (A.D.); (R.M.)
| | - Alexis Delpierre
- Department of Odontology, Tours University Hospital, 37261 Tours, France; (P.M.); (A.D.); (R.M.)
| | - Rachid Mahalli
- Department of Odontology, Tours University Hospital, 37261 Tours, France; (P.M.); (A.D.); (R.M.)
| | - Marie Guerin
- Faculty of Dentistry, Clermont-Ferrand University, 63000 Clermont-Ferrand, France;
| | - Gilles Amador
- Faculty of Dentistry, Nantes University, 44035 Nantes, France;
| | - Frederic Denis
- Department of Odontology, Tours University Hospital, 37261 Tours, France; (P.M.); (A.D.); (R.M.)
- Faculty of Dentistry, Nantes University, 44035 Nantes, France;
- EA 75-05 Education, Ethique, Santé, Faculté de Médecine, Université François-Rabelais, 37000 Tours, France
- Correspondence: ; Tel.: +33-6-7715-6968
| |
Collapse
|
608
|
O'Sullivan JN, Rea MC, Hill C, Ross RP. Protecting the outside: biological tools to manipulate the skin microbiota. FEMS Microbiol Ecol 2020; 96:5836215. [PMID: 32396198 DOI: 10.1093/femsec/fiaa085] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 05/11/2020] [Indexed: 12/18/2022] Open
Abstract
Interest surrounding the role that skin microbes play in various aspects of human health has recently experienced a timely surge, particularly among researchers, clinicians and consumer-focused industries. The world is now approaching a post-antibiotic era where conventional antibacterial therapeutics have shown a loss in effectiveness due to overuse, leading to the looming antibiotic resistance crisis. The increasing threat posed by antibiotic resistance is compounded by an inadequate discovery rate of new antibiotics and has, in turn, resulted in global interest for alternative solutions. Recent studies have demonstrated that imbalances in skin microbiota are associated with assorted skin diseases and infections. Specifically, restoration of this ecosystem imbalance results in an alleviation of symptoms, achieved simply by applying bacteria normally found in abundance on healthy skin to the skin of those deficient in beneficial bacteria. The aim of this review is to discuss the currently available literature on biological tools that have the potential to manipulate the skin microbiota, with particular focus on bacteriocins, phage therapy, antibiotics, probiotics and targets of the gut-skin axis. This review will also address how the skin microbiota protects humans from invading pathogens in the external environment while discussing novel strategies to manipulate the skin microbiota to avoid and/or treat various disease states.
Collapse
Affiliation(s)
- Julie N O'Sullivan
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland, P61 C996.,School of Microbiology, Food Science & Technology Building, University College Cork, College Road, Cork, Ireland, T12 K8AF.,APC Microbiome Ireland, Biosciences Institute, University College Cork, College Road, Cork, Ireland, T12 YT20
| | - Mary C Rea
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland, P61 C996.,APC Microbiome Ireland, Biosciences Institute, University College Cork, College Road, Cork, Ireland, T12 YT20
| | - Colin Hill
- School of Microbiology, Food Science & Technology Building, University College Cork, College Road, Cork, Ireland, T12 K8AF.,APC Microbiome Ireland, Biosciences Institute, University College Cork, College Road, Cork, Ireland, T12 YT20
| | - R Paul Ross
- School of Microbiology, Food Science & Technology Building, University College Cork, College Road, Cork, Ireland, T12 K8AF.,APC Microbiome Ireland, Biosciences Institute, University College Cork, College Road, Cork, Ireland, T12 YT20
| |
Collapse
|
609
|
López-Taboada I, González-Pardo H, Conejo NM. Western Diet: Implications for Brain Function and Behavior. Front Psychol 2020; 11:564413. [PMID: 33329193 PMCID: PMC7719696 DOI: 10.3389/fpsyg.2020.564413] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 10/02/2020] [Indexed: 12/12/2022] Open
Abstract
The Western diet (WD) pattern characterized by high daily intake of saturated fats and refined carbohydrates often leads to obesity and overweight, and it has been linked to cognitive impairment and emotional disorders in both animal models and humans. This dietary pattern alters the composition of gut microbiota, influencing brain function by different mechanisms involving the gut-brain axis. In addition, long-term exposure to highly palatable foods typical of WD could induce addictive-like eating behaviors and hypothalamic-pituitary-adrenal (HPA) axis dysregulation associated with chronic stress, anxiety, and depression. In turn, chronic stress modulates eating behavior, and it could have detrimental effects on different brain regions such as the hippocampus, hypothalamus, amygdala, and several cortical regions. Moreover, obesity and overweight induce neuroinflammation, causing neuronal dysfunction. In this review, we summarize the current scientific evidence about the mechanisms and factors relating WD consumption with altered brain function and behavior. Possible therapeutic interventions and limitations are also discussed, aiming to tackle and prevent this current pandemic.
Collapse
Affiliation(s)
| | | | - Nélida María Conejo
- Laboratory of Neuroscience, Department of Psychology, Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Oviedo, Spain
| |
Collapse
|
610
|
Atypical immunometabolism and metabolic reprogramming in liver cancer: Deciphering the role of gut microbiome. Adv Cancer Res 2020; 149:171-255. [PMID: 33579424 DOI: 10.1016/bs.acr.2020.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related mortality worldwide. Much recent research has delved into understanding the underlying molecular mechanisms of HCC pathogenesis, which has revealed to be heterogenous and complex. Two major hallmarks of HCC include: (i) a hijacked immunometabolism and (ii) a reprogramming in metabolic processes. We posit that the gut microbiota is a third component in an entanglement triangle contributing to HCC progression. Besides metagenomic studies highlighting the diagnostic potential in the gut microbiota profile, recent research is pinpointing the gut microbiota as an instigator, not just a mere bystander, in HCC. In this chapter, we discuss mechanistic insights on atypical immunometabolism and metabolic reprogramming in HCC, including the examination of tumor-associated macrophages and neutrophils, tumor-infiltrating lymphocytes (e.g., T-cell exhaustion, regulatory T-cells, natural killer T-cells), the Warburg effect, rewiring of the tricarboxylic acid cycle, and glutamine addiction. We further discuss the potential involvement of the gut microbiota in these characteristics of hepatocarcinogenesis. An immediate highlight is that microbiota metabolites (e.g., short chain fatty acids, secondary bile acids) can impair anti-tumor responses, which aggravates HCC. Lastly, we describe the rising 'new era' of immunotherapies (e.g., immune checkpoint inhibitors, adoptive T-cell transfer) and discuss for the potential incorporation of gut microbiota targeted therapeutics (e.g., probiotics, fecal microbiota transplantation) to alleviate HCC. Altogether, this chapter invigorates for continuous research to decipher the role of gut microbiome in HCC from its influence on immunometabolism and metabolic reprogramming.
Collapse
|
611
|
Mishima Y, Ishihara S. Molecular Mechanisms of Microbiota-Mediated Pathology in Irritable Bowel Syndrome. Int J Mol Sci 2020; 21:ijms21228664. [PMID: 33212919 PMCID: PMC7698457 DOI: 10.3390/ijms21228664] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
Irritable bowel syndrome (IBS) is one of the most prevalent functional gastrointestinal disorders, and accumulating evidence gained in both preclinical and clinical studies indicate the involvement of enteric microbiota in its pathogenesis. Gut resident microbiota appear to influence brain activity through the enteric nervous system, while their composition and function are affected by the central nervous system. Based on these results, the term “brain–gut–microbiome axis” has been proposed and enteric microbiota have become a potential therapeutic target in IBS cases. However, details regarding the microbe-related pathophysiology of IBS remain elusive. This review summarizes the existing knowledge of molecular mechanisms in the pathogenesis of IBS as well as recent progress related to microbiome-derived neurotransmitters, compounds, metabolites, neuroendocrine factors, and enzymes.
Collapse
|
612
|
Zhan K, Zheng H, Li J, Wu H, Qin S, Luo L, Huang S. Gut Microbiota-Bile Acid Crosstalk in Diarrhea-Irritable Bowel Syndrome. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3828249. [PMID: 33274207 PMCID: PMC7676935 DOI: 10.1155/2020/3828249] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/14/2020] [Indexed: 02/06/2023]
Abstract
The occurrence of diarrhea-predominant irritable bowel syndrome (IBS-D) is the result of multiple factors, and its pathogenesis has not yet been clarified. Emerging evidence indicates abnormal changes in gut microbiota and bile acid (BA) metabolism have a close relationship with IBS-D. Gut microbiota is involved in the secondary BA production via deconjugation, 7α-dehydroxylation, oxidation, epimerization, desulfation, and esterification reactions respectively. Changes in the composition and quantity of gut microbiota have an important impact on the metabolism of BAs, which can lead to the occurrence of gastrointestinal diseases. BAs, synthesized in the hepatocytes, play an important role in maintaining the homeostasis of gut microbiota and the balance of glucose and lipid metabolism. In consideration of the complex biological functional connections among gut microbiota, BAs, and IBS-D, it is urgent to review the latest research progress in this field. In this review, we summarized the alterations of gut microbiota in IBS-D and discussed the mechanistic connections between gut microbiota and BA metabolism in IBS-D, which may be involved in activating two important bile acid receptors, G-protein coupled bile acid receptor 1 (TGR5) and farnesoid X receptor (FXR). We also highlight the strategies of prevention and treatment of IBS-D via regulating gut microbiota-bile acid axis, including probiotics, fecal microbiota transplantation (FMT), cholestyramine, and the cutting-edge technology about bacteria genetic engineering.
Collapse
Affiliation(s)
- Kai Zhan
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Huan Zheng
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Jianqing Li
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Haomeng Wu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Shumin Qin
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Lei Luo
- Department of Gastroenterology, The Second People's Hospital of China Three Gorges University, Yichang 443000, China
| | - Shaogang Huang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| |
Collapse
|
613
|
Moon J, Yoon CH, Choi SH, Kim MK. Can Gut Microbiota Affect Dry Eye Syndrome? Int J Mol Sci 2020; 21:E8443. [PMID: 33182758 PMCID: PMC7697210 DOI: 10.3390/ijms21228443] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 02/07/2023] Open
Abstract
Using metagenomics, continuing evidence has elicited how intestinal microbiota trigger distant autoimmunity. Sjögren's syndrome (SS) is an autoimmune disease that affects the ocular surface, with frequently unmet therapeutic needs requiring new interventions for dry eye management. Current studies also suggest the possible relation of autoimmune dry eye with gut microbiota. Herein, we review the current knowledge of how the gut microbiota interact with the immune system in homeostasis as well as its influence on rheumatic and ocular autoimmune diseases, and compare their characteristics with SS. Both rodent and human studies regarding gut microbiota in SS and environmental dry eye are explored, and the effects of prebiotics and probiotics on dry eye are discussed. Recent clinical studies have commonly observed a correlation between gut dysbiosis and clinical manifestations of SS, while environmental dry eye portrays characteristics in between normal and autoimmune. Moreover, a decrease in both the Firmicutes/Bacteroidetes ratio and genus Faecalibacterium have most commonly been observed in SS subjects. The presumable pathways forming the "gut dysbiosis-ocular surface-lacrimal gland axis" are introduced. This review may provide perspectives into the link between the gut microbiome and dry eye, enhance our understanding of the pathogenesis in autoimmune dry eye, and be useful in the development of future interventions.
Collapse
Affiliation(s)
- Jayoon Moon
- Department of Ophthalmology, College of Medicine, Seoul National University, Seoul 03080, Korea; (J.M.); (C.H.Y.)
- Seoul Artificial Eye Center, Laboratory of Ocular Regenerative Medicine and Immunology, Seoul National University Hospital Biomedical Research Institute, Seoul 03082, Korea;
| | - Chang Ho Yoon
- Department of Ophthalmology, College of Medicine, Seoul National University, Seoul 03080, Korea; (J.M.); (C.H.Y.)
- Seoul Artificial Eye Center, Laboratory of Ocular Regenerative Medicine and Immunology, Seoul National University Hospital Biomedical Research Institute, Seoul 03082, Korea;
| | - Se Hyun Choi
- Seoul Artificial Eye Center, Laboratory of Ocular Regenerative Medicine and Immunology, Seoul National University Hospital Biomedical Research Institute, Seoul 03082, Korea;
- Department of Ophthalmology, Hallym University Sacred Heart Hospital, Anyang-si 14068, Korea
| | - Mee Kum Kim
- Department of Ophthalmology, College of Medicine, Seoul National University, Seoul 03080, Korea; (J.M.); (C.H.Y.)
- Seoul Artificial Eye Center, Laboratory of Ocular Regenerative Medicine and Immunology, Seoul National University Hospital Biomedical Research Institute, Seoul 03082, Korea;
| |
Collapse
|
614
|
Sharma L, Riva A. Intestinal Barrier Function in Health and Disease-Any role of SARS-CoV-2? Microorganisms 2020; 8:E1744. [PMID: 33172188 PMCID: PMC7694956 DOI: 10.3390/microorganisms8111744] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022] Open
Abstract
Alterations in the structure and function of the intestinal barrier play a role in the pathogenesis of a multitude of diseases. During the recent and ongoing coronavirus disease (COVID-19) pandemic, it has become clear that the gastrointestinal system and the gut barrier may be affected by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, and disruption of barrier functions or intestinal microbial dysbiosis may have an impact on the progression and severity of this new disease. In this review, we aim to provide an overview of current evidence on the involvement of gut alterations in human disease including COVID-19, with a prospective outlook on supportive therapeutic strategies that may be investigated to rescue intestinal barrier functions and possibly facilitate clinical improvement in these patients.
Collapse
Affiliation(s)
- Lakshya Sharma
- Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, UK;
| | - Antonio Riva
- Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, UK;
- Foundation for Liver Research, Institute of Hepatology, London SE5 9NT, UK
| |
Collapse
|
615
|
Montgomery TL, Künstner A, Kennedy JJ, Fang Q, Asarian L, Culp-Hill R, D'Alessandro A, Teuscher C, Busch H, Krementsov DN. Interactions between host genetics and gut microbiota determine susceptibility to CNS autoimmunity. Proc Natl Acad Sci U S A 2020; 117:27516-27527. [PMID: 33077601 PMCID: PMC7959502 DOI: 10.1073/pnas.2002817117] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system. The etiology of MS is multifactorial, with disease risk determined by genetics and environmental factors. An emerging risk factor for immune-mediated diseases is an imbalance in the gut microbiome. However, the identity of gut microbes associated with disease risk, their mechanisms of action, and the interactions with host genetics remain obscure. To address these questions, we utilized the principal autoimmune model of MS, experimental autoimmune encephalomyelitis (EAE), together with a genetically diverse mouse model representing 29 unique host genotypes, interrogated by microbiome sequencing and targeted microbiome manipulation. We identified specific gut bacteria and their metabolic functions associated with EAE susceptibility, implicating short-chain fatty acid metabolism as a key element conserved across multiple host genotypes. In parallel, we used a reductionist approach focused on two of the most disparate phenotypes identified in our screen. Manipulation of the gut microbiome by transplantation and cohousing demonstrated that transfer of these microbiomes into genetically identical hosts was sufficient to modulate EAE susceptibility and systemic metabolite profiles. Parallel bioinformatic approaches identified Lactobacillus reuteri as a commensal species unexpectedly associated with exacerbation of EAE in a genetically susceptible host, which was functionally confirmed by bacterial isolation and commensal colonization studies. These results reveal complex interactions between host genetics and gut microbiota modulating susceptibility to CNS autoimmunity, providing insights into microbiome-directed strategies aimed at lowering the risk for autoimmune disease and underscoring the need to consider host genetics and baseline gut microbiome composition.
Collapse
Affiliation(s)
- Theresa L Montgomery
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05401
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany
| | - Josephine J Kennedy
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05401
| | - Qian Fang
- Department of Medicine, Immunobiology Division, University of Vermont, Burlington, VT 05401
| | - Lori Asarian
- Department of Medicine, Immunobiology Division, University of Vermont, Burlington, VT 05401
| | - Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado, Aurora, CO 80045
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado, Aurora, CO 80045
| | - Cory Teuscher
- Department of Medicine, Immunobiology Division, University of Vermont, Burlington, VT 05401
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany
| | - Dimitry N Krementsov
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05401;
| |
Collapse
|
616
|
Wang Y, Huang YQ, Zhu SL, Zhang CR, Chen XL, Hou QK, Liu FB. Efficacy of Tong-Xie-Yao-Fang granule and its impact on whole transcriptome profiling in diarrhea-predominant irritable bowel syndrome patients: study protocol for a randomized controlled trial. Trials 2020; 21:908. [PMID: 33143731 PMCID: PMC7607547 DOI: 10.1186/s13063-020-04833-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is one kind of common functional bowel disease with obscure pathogenesis, and exploration about whole transcriptome profiling in IBS-D is still negligible. Conventional medications have limited effects, which makes focus shifted to traditional Chinese medicine (TCM). Tong-Xie-Yao-Fang, as a classic herbal formula in TCM, is pretty effective and safe for the treatment of diarrhea-predominant irritable bowel syndrome (IBS-D), but the underlying therapeutic mechanism remains unknown. We aim to verify the efficacy and safety of TXYF granule (the formula particles mixed together) in IBS-D and elucidate the gene-level mechanism of IBS-D and therapeutic targets of TXYF granule based on whole transcriptome analysis. METHODS/DESIGN This is a randomized, double-blind, and placebo-controlled clinical trial consisting of 2 weeks of run-in period, 12 weeks of treatment period, and 8 weeks of follow-up period. We will enroll 120 participants with IBS-D, who will be randomly assigned to the TXYF granule group and the placebo group, and recruit additional 10 healthy individuals as controls for mechanistic outcome. The two groups respectively take TXYF granule or placebo orally for treatment. The primary outcome is the response rate of IBS-Symptom Severity Score (IBS-SSS). The secondary outcomes include adequate relief (AR), IBS-Quality of Life Questionnaire (IBS-QOL), and long-term efficacy. Mechanistic outcome is the whole transcriptome profiling of the intestinal mucosae from IBS participants before and after the treatment and healthy individuals. DISCUSSION This trial will prove the effectiveness and safety of TXYF granule with high-quality evidence and provide a penetrating and comprehensive perspective on the molecular mechanism of IBS-D by whole transcriptome analysis, which makes us pinpoint specific biomarkers of IBS-D and therapeutic targets of TXYF. TRIAL REGISTRATION Chinese Clinical Trial Registry ChiCTR-IOR-1900021785 . Registered on 9 March 2019.
Collapse
Affiliation(s)
- Yan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong-Quan Huang
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shui-Lian Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chang-Rong Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin-Lin Chen
- Department of Preventive Medicine and Health Statistics, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiu-Ke Hou
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Feng-Bin Liu
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
617
|
Osadchiy V, Mayer EA, Gao K, Labus JS, Naliboff B, Tillisch K, Chang L, Jacobs JP, Hsiao EY, Gupta A. Analysis of brain networks and fecal metabolites reveals brain-gut alterations in premenopausal females with irritable bowel syndrome. Transl Psychiatry 2020; 10:367. [PMID: 33139708 PMCID: PMC7608552 DOI: 10.1038/s41398-020-01071-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/11/2020] [Accepted: 10/05/2020] [Indexed: 01/16/2023] Open
Abstract
Alterations in brain-gut-microbiome (BGM) interactions have been implicated in the pathogenesis of irritable bowel syndrome (IBS). Here, we apply a systems biology approach, leveraging neuroimaging and fecal metabolite data, to characterize BGM interactions that are driving IBS pathophysiology. Fecal samples and resting state fMRI images were obtained from 138 female subjects (99 IBS, 39 healthy controls (HCs)). Partial least-squares discriminant analysis (PLS-DA) was conducted to explore group differences, and partial correlation analysis explored significantly changed metabolites and neuroimaging data. All correlational tests were performed controlling for age, body mass index, and diet; results are reported after FDR correction, with q < 0.05 as significant. Compared to HCs, IBS showed increased connectivity of the putamen with regions of the default mode and somatosensory networks. Metabolite pathways involved in nucleic acid and amino acid metabolism differentiated the two groups. Only a subset of metabolites, primarily amino acids, were associated with IBS-specific brain changes, including tryptophan, glutamate, and histidine. Histidine was the only metabolite positively associated with both IBS-specific alterations in brain connectivity. Our findings suggest a role for several amino acid metabolites in modulating brain function in IBS. These metabolites may alter brain connectivity directly, by crossing the blood-brain-barrier, or indirectly through peripheral mechanisms. This is the first study to integrate both neuroimaging and fecal metabolite data supporting the BGM model of IBS, building the foundation for future mechanistic studies on the influence of gut microbial metabolites on brain function in IBS.
Collapse
Affiliation(s)
- Vadim Osadchiy
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Emeran A Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- UCLA Microbiome Center, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Kan Gao
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jennifer S Labus
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Bruce Naliboff
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Kirsten Tillisch
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- UCLA Microbiome Center, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Lin Chang
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Jonathan P Jacobs
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- UCLA Microbiome Center, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Elaine Y Hsiao
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Arpana Gupta
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA.
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Vatche and Tamar Manoukian Division of Digestive Diseases, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA.
| |
Collapse
|
618
|
Chaves Andrade M, Souza de Faria R, Avelino Mota Nobre S. COVID-19: Can the symptomatic SARS-CoV-2 infection affect the homeostasis of the gut-brain-microbiota axis? Med Hypotheses 2020; 144:110206. [PMID: 33254513 PMCID: PMC7444650 DOI: 10.1016/j.mehy.2020.110206] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/06/2020] [Accepted: 08/18/2020] [Indexed: 12/21/2022]
Abstract
COVID-19 is associated with acute and lethal pneumonia, causing the severe acute respiratory syndrome (SARS), which is not confined to the respiratory tract, as demonstrated by clinical evidence of the involvement of multiple organs, including the central nervous system (CNS). In this context, we hypothesized that both oligosymptomatic and symptomatic patients present an imbalance in the microbiota-gut (immune system) and nervous system axis, worsening the clinical picture. The brain constantly receives a direct and indirect influence from the intestine, more specifically from the immune system and intestinal microbiota. The presence of SARS-CoV-2 in the intestine and CNS, can contribute to both neurological disorders and gut immune system imbalance, events potentialized by an intestinal microbiota dysbiosis, aggravating the patient's condition and causing more prolonged harmful effects.
Collapse
Affiliation(s)
- Marileia Chaves Andrade
- Universidade Estadual de Montes Claros, UNIMONTES, Montes Claros, Minas Gerais, Brazil; Faculdade de Medicina de Itajubá, FMIT, Itajubá, Minas Gerais, Brazil.
| | | | | |
Collapse
|
619
|
Verduci E, Carbone MT, Borghi E, Ottaviano E, Burlina A, Biasucci G. Nutrition, Microbiota and Role of Gut-Brain Axis in Subjects with Phenylketonuria (PKU): A Review. Nutrients 2020; 12:E3319. [PMID: 33138040 PMCID: PMC7692600 DOI: 10.3390/nu12113319] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022] Open
Abstract
The composition and functioning of the gut microbiota, the complex population of microorganisms residing in the intestine, is strongly affected by endogenous and exogenous factors, among which diet is key. Important perturbations of the microbiota have been observed to contribute to disease risk, as in the case of neurological disorders, inflammatory bowel disease, obesity, diabetes, cardiovascular disease, among others. Although mechanisms are not fully clarified, nutrients interacting with the microbiota are thought to affect host metabolism, immune response or disrupt the protective functions of the intestinal barrier. Similarly, key intermediaries, whose presence may be strongly influenced by dietary habits, sustain the communication along the gut-brain-axis, influencing brain functions in the same way as the brain influences gut activity. Due to the role of diet in the modulation of the microbiota, its composition is of high interest in inherited errors of metabolism (IEMs) and may reveal an appealing therapeutic target. In IEMs, for example in phenylketonuria (PKU), since part of the therapeutic intervention is based on chronic or life-long tailored dietetic regimens, important variations of the microbial diversity or relative abundance have been observed. A holistic approach, including a healthy composition of the microbiota, is recommended to modulate host metabolism and affected neurological functions.
Collapse
Affiliation(s)
- Elvira Verduci
- Department of Paediatrics, Vittore Buzzi Children’s Hospital-University of Milan, Via Lodovico Castelvetro, 32, 20154 Milan, Italy
- Department of Health Science, University of Milan, via di Rudinì 8, 20142 Milan, Italy; (E.B.); (E.O.)
| | - Maria Teresa Carbone
- UOS Metabolic and Rare Diseases, AORN Santobono, Via Mario Fiore 6, 80122 Naples, Italy;
| | - Elisa Borghi
- Department of Health Science, University of Milan, via di Rudinì 8, 20142 Milan, Italy; (E.B.); (E.O.)
| | - Emerenziana Ottaviano
- Department of Health Science, University of Milan, via di Rudinì 8, 20142 Milan, Italy; (E.B.); (E.O.)
| | - Alberto Burlina
- Division of Inborn Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, Via Orus 2B, 35129 Padua, Italy;
| | - Giacomo Biasucci
- Department of Paediatrics & Neonatology, Guglielmo da Saliceto Hospital, Via Taverna Giuseppe, 49, 29121 Piacenza, Italy;
| |
Collapse
|
620
|
Redweik GAJ, Jochum J, Mellata M. Live Bacterial Prophylactics in Modern Poultry. Front Vet Sci 2020; 7:592312. [PMID: 33195630 PMCID: PMC7655978 DOI: 10.3389/fvets.2020.592312] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/21/2020] [Indexed: 11/25/2022] Open
Abstract
Commercial poultry farms frequently use live bacterial prophylactics like vaccines and probiotics to prevent bacterial infections. Due to the emergence of antibiotic-resistant bacteria in poultry animals, a closer examination into the health benefits and limitations of commercial, live prophylactics as an alternative to antibiotics is urgently needed. In this review, we summarize the peer-reviewed literature of several commercial live bacterial vaccines and probiotics. Per our estimation, there is a paucity of peer-reviewed published research regarding these products, making repeatability, product-comparison, and understanding biological mechanisms difficult. Furthermore, we briefly-outline significant issues such as probiotic-label accuracy, lack of commercially available live bacterial vaccines for major poultry-related bacteria such as Campylobacter and Clostridium perfringens, as well research gaps (i.e., probiotic-mediated vaccine adjuvancy, gut-brain-microbiota axis). Increased emphasis on these areas would open several avenues for research, ranging from improving protection against bacterial pathogens to using these prophylactics to modulate animal behavior.
Collapse
Affiliation(s)
- Graham A. J. Redweik
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States
| | - Jared Jochum
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States
| | - Melha Mellata
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States
| |
Collapse
|
621
|
Abstract
In a striking display of trans-kingdom symbiosis, gut bacteria cooperate with their animal hosts to regulate the development and function of the immune, metabolic and nervous systems through dynamic bidirectional communication along the 'gut-brain axis'. These processes may affect human health, as certain animal behaviours appear to correlate with the composition of gut bacteria, and disruptions in microbial communities have been implicated in several neurological disorders. Most insights about host-microbiota interactions come from animal models, which represent crucial tools for studying the various pathways linking the gut and the brain. However, there are complexities and manifest limitations inherent in translating complex human disease to reductionist animal models. In this Review, we discuss emerging and exciting evidence of intricate and crucial connections between the gut microbiota and the brain involving multiple biological systems, and possible contributions by the gut microbiota to neurological disorders. Continued advances from this frontier of biomedicine may lead to tangible impacts on human health.
Collapse
|
622
|
Liu YCG, Lan SJ, Hirano H, Lin LM, Hori K, Lin CS, Zwetchkenbaum S, Minakuchi S, Teng AYT. Update and review of the gerodontology prospective for 2020's: Linking the interactions of oral (hypo)-functions to health vs. systemic diseases. J Dent Sci 2020; 16:757-773. [PMID: 33854730 PMCID: PMC8025188 DOI: 10.1016/j.jds.2020.09.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/03/2020] [Indexed: 02/07/2023] Open
Abstract
New lines of evidence suggest that the oral-systemic medical links and oral hypo-function are progressively transcending beyond the traditional clinical signs and symptoms of oral diseases. Research into the dysbiotic microbiome, host immune/inflammatory regulations and patho-physiologic changes and subsequent adaptations through the oral-systemic measures under ageism points to pathways leading to mastication deficiency, dysphagia, signature brain activities for (neuro)-cognition circuitries, dementia and certain cancers of the digestive system as well. Therefore, the coming era of oral health-linked systemic disorders will likely reshape the future of diagnostics in oral geriatrics, treatment modalities and professional therapies in clinical disciplines. In parallel to these highlights, a recent international symposium was jointly held by the International Association of Gerontology and Geriatrics (IAGG), Japanese Society of Gerodontology (JSG), the representative of USA and Taiwan Academy of Geriatric Dentistry (TAGD) on Oct 25th, 2019. Herein, specific notes are briefly addressed and updated for a summative prospective from this symposium and the recent literature.
Collapse
Affiliation(s)
- Yen Chun G. Liu
- Center for Osteoimmunology & Biotechnology Research (COBR) and Dept. of Oral Hygiene, College of Dental Medicine, Kaohsiung Medical University (KMU), Kaohsiung, Taiwan
- Corresponding author. Dept. of Oral Hygiene & COBR, College of Dental Medicine, Kaohsiung Medical University; No. 100, Shih-Chun 1st Rd, Kaohsiung 807, Taiwan. Fax: +886 07 3223141.
| | - Shou-Jen Lan
- Dept. of Healthcare Administration, Asia University, Tai-Chung, Taiwan
| | - Hirohiko Hirano
- Research Team for Promoting Independence & Mental Health, and Dentistry & Oral Surgery, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Li-min Lin
- Div. of Oral Pathology & Oral Maxillo-facial Radiology, School of Dentistry, Kaohsiung Medical University & KMU-Hospital, Kaohsiung, Taiwan
| | - Kazuhiro Hori
- Div. of Comprehensive Prosthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Chia-shu Lin
- Dept. of Dentistry, School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Samuel Zwetchkenbaum
- Rhode Island Dept. of Health, Rhode Island, USA
- School of Public Health, Brown University, Providence, RI, USA
| | - Shunsuke Minakuchi
- Gerodontology & Oral Rehabilitation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Andy Yen-Tung Teng
- Center for Osteoimmunology & Biotechnology Research (COBR) and Dept. of Oral Hygiene, College of Dental Medicine, Kaohsiung Medical University (KMU), Kaohsiung, Taiwan
- Center for Osteoimmunology & Biotechnology Research (COBR) and School of Dentistry, College of Dental Medicine, Kaohsiung Medical University and KMU-Hospital, Kaohsiung, Taiwan
- Corresponding author. Center for Osteoimmunology and Biotechnology Research (COBR), College of Dental Medicine, Kaohsiung Medical University (KMU) & KMU-Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
623
|
Alessi MG, Bennett JM. Mental health is the health of the whole body: How psychoneuroimmunology & health psychology can inform & improve treatment. J Eval Clin Pract 2020; 26:1539-1547. [PMID: 32171052 DOI: 10.1111/jep.13386] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/12/2020] [Accepted: 02/20/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Converging and accumulating evidence for the cross-communication among the nervous, immune, and endocrine systems, a field of study known as psychoneuroimmunology, implicates immunological dysfunction as a shared and common mechanism of both mental and physical illness. For example, psychiatric disorders like schizophrenia, bipolar disorder, major depression, and anxiety disorders have higher prevalence rates across a spectrum of autoimmune conditions compared to the general population. Additionally, subclinical immunological abnormalities are observed in a variety of psychiatric conditions, with chronic inflammation most extensively studied in the pathophysiology of depression. These observations blur the historical distinctions between mental and physical illness, yet clinical practice remains fragmented and primarily focused on differentially treating individual symptoms. PROPOSED THESIS Therapeutically targeting inflammation offers translational opportunities for integrating mental and physical healthcare, a key niche of the interdisciplinary field of health psychology. CONCLUSION Utilizing a psychoneuroimmunological lens, health psychologists and clinicians can reconceptualize healthcare through integrative treatment approaches and advocacy for comprehensive policy-level reform at both the individual-level of care as well as community-wide prevention approaches.
Collapse
Affiliation(s)
- Maria G Alessi
- Health Psychology PhD Program, UNC Charlotte, Charlotte, North Carolina, USA
| | - Jeanette M Bennett
- Health Psychology PhD Program, UNC Charlotte, Charlotte, North Carolina, USA.,Department of Psychological Science, UNC Charlotte, Charlotte, North Carolina, USA
| |
Collapse
|
624
|
Cross-talk between gut and brain elicited by physical exercise. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165877. [PMID: 32544430 DOI: 10.1016/j.bbadis.2020.165877] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/26/2020] [Accepted: 06/08/2020] [Indexed: 12/21/2022]
Abstract
In recent years, a paradigm shift in the bidirectional interactions within the gut-brain axis in normal and pathologic conditions has been evidenced. Although the causal relationship is not completely known, the application of new therapeutic tools such as physical exercise has been described in several studies. However, there are caveats to consider when interpreting the effect of exercise training on the axis. Therefore, an integrative perspective of the gut and the brain's communication pathway is discussed and the role of exercise on influencing this communication highway is explained in this review.
Collapse
|
625
|
Beumer J, Gehart H, Clevers H. Enteroendocrine Dynamics - New Tools Reveal Hormonal Plasticity in the Gut. Endocr Rev 2020; 41:5856764. [PMID: 32531023 PMCID: PMC7320824 DOI: 10.1210/endrev/bnaa018] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 06/09/2020] [Indexed: 12/16/2022]
Abstract
The recent intersection of enteroendocrine cell biology with single-cell technologies and novel in vitro model systems has generated a tremendous amount of new data. Here we highlight these recent developments and explore how these findings contribute to the understanding of endocrine lineages in the gut. In particular, the concept of hormonal plasticity, the ability of endocrine cells to produce different hormones over the course of their lifetime, challenges the classic notion of cell types. Enteroendocrine cells travel in the course of their life through different signaling environments that directly influence their hormonal repertoire. In this context, we examine how enteroendocrine cell fate is determined and modulated by signaling molecules such as bone morphogenetic proteins (BMPs) or location along the gastrointestinal tract. We analyze advantages and disadvantages of novel in vitro tools, adult stem cell or iPS-derived intestinal organoids, that have been crucial for recent findings on enteroendocrine development and plasticity. Finally, we illuminate the future perspectives of the field and discuss how understanding enteroendocrine plasticity can lead to new therapeutic approaches.
Collapse
Affiliation(s)
- Joep Beumer
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, CT Utrecht, The Netherlands
| | - Helmuth Gehart
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, CT Utrecht, The Netherlands.,Institute for Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, CT Utrecht, The Netherlands.,Oncode Institute, Hubrecht Institute, CT Utrecht, The Netherlands
| |
Collapse
|
626
|
Smith NM, Maloney NG, Shaw S, Horgan GW, Fyfe C, Martin JC, Suter A, Scott KP, Johnstone AM. Daily Fermented Whey Consumption Alters the Fecal Short-Chain Fatty Acid Profile in Healthy Adults. Front Nutr 2020; 7:165. [PMID: 33102510 PMCID: PMC7556162 DOI: 10.3389/fnut.2020.00165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
Gut microbiota influences many aspects of host health including immune, metabolic, and gut health. We examined the effect of a fermented whey concentrate (FWC) drink rich in L-(+)-Lactic acid, consumed daily, in 18 healthy men (n = 5) and women (n = 13) in free-living conditions. Objective: The aims of this 6-weeks pilot trial were to (i) identify changes in the gut microbiota composition and fecal short chain fatty acid (SCFA) profile, and (ii) to monitor changes in glucose homeostasis. Results: Total fecal SCFA (mM) concentration remained constant throughout the intervention. Proportionally, there was a significant change in the composition of different SCFAs compared to baseline. Acetate levels were significantly reduced (−6.5%; p < 0.01), coupled to a significant increase in the relative amounts of propionate (+2.2%; p < 0.01) and butyrate (+4.2%; p < 0.01), respectively. No changes in the relative abundance of any specific bacteria were detected. No significant changes were observed in glucose homeostasis in response to an oral glucose tolerance test. Conclusion: Daily consumption of a fermented whey product led to significant changes in fecal SCFA metabolite profile, indicating some potential prebiotic activity. These changes did not result in any detectable differences in microbiota composition. Post-hoc analysis indicated that baseline microbiota composition might be indicative of participants likely to see changes in SCFA levels. However, due to the lack of a control group these findings would need to be verified in a rigorously controlled trial. Future work is also required to identify the biological mechanisms underlying the observed changes in microbiota activity and to explore if these processes can be harnessed to favorably impact host health. Clinical Trial Registration: www.clinicaltrials.gov, identifier NCT03615339; retrospectively registered on 03/08/2018.
Collapse
Affiliation(s)
- Nicola M Smith
- School of Medicine, Medical Sciences and Nutrition, The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| | - Niamh G Maloney
- School of Medicine, Medical Sciences and Nutrition, The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| | - Sophie Shaw
- Centre for Genome Enabled Biology and Medicine, University of Aberdeen, Aberdeen, United Kingdom
| | - Graham W Horgan
- Biomathematics & Statistics Scotland, University of Aberdeen, Aberdeen, United Kingdom
| | - Claire Fyfe
- School of Medicine, Medical Sciences and Nutrition, The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| | - Jennifer C Martin
- School of Medicine, Medical Sciences and Nutrition, The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| | | | - Karen P Scott
- School of Medicine, Medical Sciences and Nutrition, The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| | - Alexandra M Johnstone
- School of Medicine, Medical Sciences and Nutrition, The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
627
|
Jang SH, Woo YS, Lee SY, Bahk WM. The Brain-Gut-Microbiome Axis in Psychiatry. Int J Mol Sci 2020; 21:E7122. [PMID: 32992484 PMCID: PMC7583027 DOI: 10.3390/ijms21197122] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
Beginning with the concept of the brain-gut axis, the importance of the interaction between the brain and the gastrointestinal tract has been extended to the microbiome with increasing clinical applications. With the recent development of various techniques for microbiome analysis, the number of relevant preclinical and clinical studies on animals and human subjects has rapidly increased. Various psychotic symptoms affect the intestinal microbiome through the hypothalamus-pituitary-adrenal gland axis. Conversely, the intestinal microbiome regulates the gastrointestinal tract environment and affects psychological factors by means of the microorganisms or their metabolites, either acting directly on the brain or through the synthesis of various neurotransmitters. This review discusses the clinical applicability of the brain-gut-microbiome axis and directions for improving psychological symptoms based on the studies published to date.
Collapse
Affiliation(s)
- Seung-Ho Jang
- Department of Psychiatry, School of Medicine, Wonkwang University, Iksan 54538, Korea; (S.-H.J.); (S.-Y.L.)
| | - Young Sup Woo
- Department of Psychiatry, College of Medicine, The Catholic University of Korea, Seoul 07345, Korea;
| | - Sang-Yeol Lee
- Department of Psychiatry, School of Medicine, Wonkwang University, Iksan 54538, Korea; (S.-H.J.); (S.-Y.L.)
| | - Won-Myong Bahk
- Department of Psychiatry, College of Medicine, The Catholic University of Korea, Seoul 07345, Korea;
| |
Collapse
|
628
|
Bistoletti M, Bosi A, Banfi D, Giaroni C, Baj A. The microbiota-gut-brain axis: Focus on the fundamental communication pathways. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 176:43-110. [PMID: 33814115 DOI: 10.1016/bs.pmbts.2020.08.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Annalisa Bosi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Davide Banfi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy.
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
629
|
Dong TS, Gupta A, Jacobs JP, Lagishetty V, Gallagher E, Bhatt RR, Vora P, Osadchiy V, Stains J, Balioukova A, Chen Y, Dutson E, Mayer EA, Sanmiguel C. Improvement in Uncontrolled Eating Behavior after Laparoscopic Sleeve Gastrectomy Is Associated with Alterations in the Brain-Gut-Microbiome Axis in Obese Women. Nutrients 2020; 12:E2924. [PMID: 32987837 PMCID: PMC7599899 DOI: 10.3390/nu12102924] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bariatric surgery is proven to change eating behavior and cause sustained weight loss, yet the exact mechanisms underlying these changes are not clearly understood. We explore this in a novel way by examining how bariatric surgery affects the brain-gut-microbiome (BGM) axis. METHODS Patient demographics, serum, stool, eating behavior questionnaires, and brain magnetic resonance imaging (MRI) were collected before and 6 months after laparoscopic sleeve gastrectomy (LSG). Differences in eating behavior and brain morphology and resting-state functional connectivity in core reward regions were correlated with serum metabolite and 16S microbiome data. RESULTS LSG resulted in significant weight loss and improvement in maladaptive eating behaviors as measured by the Yale Food Addiction Scale (YFAS). Brain imaging showed a significant increase in brain volume of the putamen (p.adj < 0.05) and amygdala (p.adj < 0.05) after surgery. Resting-state connectivity between the precuneus and the putamen was significantly reduced after LSG (p.adj = 0.046). This change was associated with YFAS symptom count. Bacteroides, Ruminococcus, and Holdemanella were associated with reduced connectivity between these areas. Metabolomic profiles showed a positive correlation between this brain connection and a phosphatidylcholine metabolite. CONCLUSION Bariatric surgery modulates brain networks that affect eating behavior, potentially through effects on the gut microbiota and its metabolites.
Collapse
Affiliation(s)
- Tien S. Dong
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (A.G.); (J.P.J.); (V.L.); (E.A.M.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90025, USA
| | - Arpana Gupta
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (A.G.); (J.P.J.); (V.L.); (E.A.M.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, CA 90095, USA; (E.G.); (R.R.B.); (P.V.); (V.O.); (J.S.)
- David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (Y.C.); (E.D.)
| | - Jonathan P. Jacobs
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (A.G.); (J.P.J.); (V.L.); (E.A.M.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90025, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, CA 90095, USA; (E.G.); (R.R.B.); (P.V.); (V.O.); (J.S.)
- David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (Y.C.); (E.D.)
| | - Venu Lagishetty
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (A.G.); (J.P.J.); (V.L.); (E.A.M.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90025, USA
| | - Elizabeth Gallagher
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, CA 90095, USA; (E.G.); (R.R.B.); (P.V.); (V.O.); (J.S.)
| | - Ravi R. Bhatt
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, CA 90095, USA; (E.G.); (R.R.B.); (P.V.); (V.O.); (J.S.)
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine at USC, University of Southern California, Los Angeles, CA 90033, USA
| | - Priten Vora
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, CA 90095, USA; (E.G.); (R.R.B.); (P.V.); (V.O.); (J.S.)
| | - Vadim Osadchiy
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, CA 90095, USA; (E.G.); (R.R.B.); (P.V.); (V.O.); (J.S.)
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Jean Stains
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, CA 90095, USA; (E.G.); (R.R.B.); (P.V.); (V.O.); (J.S.)
| | - Anna Balioukova
- UCLA Center for Obesity and METabolic Health (COMET), Los Angeles, CA 90024, USA;
| | - Yijun Chen
- David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (Y.C.); (E.D.)
- UCLA Center for Obesity and METabolic Health (COMET), Los Angeles, CA 90024, USA;
| | - Erik Dutson
- David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (Y.C.); (E.D.)
- UCLA Center for Obesity and METabolic Health (COMET), Los Angeles, CA 90024, USA;
| | - Emeran A. Mayer
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (A.G.); (J.P.J.); (V.L.); (E.A.M.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, CA 90095, USA; (E.G.); (R.R.B.); (P.V.); (V.O.); (J.S.)
- David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (Y.C.); (E.D.)
| | - Claudia Sanmiguel
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (T.S.D.); (A.G.); (J.P.J.); (V.L.); (E.A.M.)
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90025, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, CA 90095, USA; (E.G.); (R.R.B.); (P.V.); (V.O.); (J.S.)
- David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (Y.C.); (E.D.)
| |
Collapse
|
630
|
Laue HE, Korrick SA, Baker ER, Karagas MR, Madan JC. Prospective associations of the infant gut microbiome and microbial function with social behaviors related to autism at age 3 years. Sci Rep 2020; 10:15515. [PMID: 32968156 PMCID: PMC7511970 DOI: 10.1038/s41598-020-72386-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/31/2020] [Indexed: 12/22/2022] Open
Abstract
The hypothesized link between gut bacteria and autism spectrum disorder (ASD) has been explored through animal models and human studies with microbiome assessment after ASD presentation. We aimed to prospectively characterize the association between the infant/toddler gut microbiome and ASD-related social behaviors at age 3 years. As part of an ongoing birth cohort gut bacterial diversity, structure, taxa, and function at 6 weeks (n = 166), 1 year (n = 158), 2 years (n = 129), and 3 years (n = 140) were quantified with 16S rRNA gene and shotgun metagenomic sequencing (n = 101 six weeks, n = 103 one year). ASD-related social behavior was assessed at age 3 years using Social Responsiveness Scale (SRS-2) T-scores. Covariate-adjusted linear and permutation-based models were implemented. Microbiome structure at 1 year was associated with SRS-2 total T-scores (p = 0.01). Several taxa at 1, 2, and 3 years were associated with SRS-2 performance, including many in the Lachnospiraceae family. Higher relative abundance of Adlercreutzia equolifaciens and Ruminococcus torques at 1 year related to poorer SRS-2 performance. Two functional pathways, L-ornithine and vitamin B6 biosynthesis, were associated with better social skills at 3 years. Our results support potential associations between early-childhood gut microbiome and social behaviors. Future mechanistic studies are warranted to pinpoint sensitive targets for intervention.
Collapse
Affiliation(s)
- Hannah E Laue
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH, USA.
| | - Susan A Korrick
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Emily R Baker
- Department of Obstetrics and Gynecology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Margaret R Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH, USA
| | - Juliette C Madan
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH, USA
- Department of Pediatrics, Children's Hospital at Dartmouth, Lebanon, NH, USA
- Department of Psychiatry, Children's Hospital at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
631
|
Berentsen B, Nagaraja BH, Teige EP, Lied GA, Lundervold AJ, Lundervold K, Steinsvik EK, Hillestad ER, Valeur J, Brønstad I, Gilja OH, Osnes B, Hatlebakk JG, Haász J, Labus J, Gupta A, Mayer EA, Benitez-Páez A, Sanz Y, Lundervold A, Hausken T. Study protocol of the Bergen brain-gut-microbiota-axis study: A prospective case-report characterization and dietary intervention study to evaluate the effects of microbiota alterations on cognition and anatomical and functional brain connectivity in patients with irritable bowel syndrome. Medicine (Baltimore) 2020; 99:e21950. [PMID: 32925728 PMCID: PMC7489588 DOI: 10.1097/md.0000000000021950] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Irritable bowel syndrome (IBS) is a common clinical label for medically unexplained gastrointestinal (GI) symptoms, recently described as a disturbance of the brain-gut-microbiota (BGM) axis. To gain a better understanding of the mechanisms underlying the poorly understood etiology of IBS, we have designed a multifaceted study that aim to stratify the complex interaction and dysfunction between the brain, the gut, and the microbiota in patients with IBS. METHODS Deep phenotyping data from patients with IBS (n = 100) and healthy age- (between 18 and 65) and gender-matched controls (n = 40) will be collected between May 2019 and December 2021. Psychometric tests, questionnaires, human biological tissue/samples (blood, faeces, saliva, and GI biopsies from antrum, duodenum, and sigmoid colon), assessment of gastric accommodation and emptying using transabdominal ultrasound, vagal activity, and functional and structural magnetic resonance imaging (MRI) of the brain, are included in the investigation of each participant. A subgroup of 60 patients with IBS-D will be further included in a 12-week low FODMAP dietary intervention-study to determine short and long-term effects of diet on GI symptoms, microbiota composition and functions, molecular GI signatures, cognitive, emotional and social functions, and structural and functional brain signatures. Deep machine learning, prediction tools, and big data analyses will be used for multivariate analyses allowing disease stratification and diagnostic biomarker detection. DISCUSSION To our knowledge, this is the first study to employ unsupervised machine learning techniques and incorporate systems-based interactions between the central and the peripheral components of the brain-gut-microbiota axis at the levels of the multiomics, microbiota profiles, and brain connectome of a cohort of 100 patients with IBS and matched controls; study long-term safety and efficacy of the low-FODMAP diet on changes in nutritional status, gut microbiota composition, and metabolites; and to investigate changes in the brain and gut connectome after 12 weeks strict low-FODMAP-diet in patients with IBS. However, there are also limitations to the study. As a restrictive diet, the low-FODMAP diet carries risks of nutritional inadequacy and may foster disordered eating patterns. Strict FODMAP restriction induces a potentially unfavourable gut microbiota, although the health effects are unknown. TRIAL REGISTRATION NUMBER NCT04296552 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Birgitte Berentsen
- National Center for Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
- Center for Nutrition, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | | | - Erica Pearson Teige
- National Center for Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
- Center for Nutrition, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Gülen Arslan Lied
- National Center for Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
- Center for Nutrition, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Astri J. Lundervold
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Katarina Lundervold
- National Center for Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
- National Center for Ultrasound in Gastroenterology, Medical Department, Haukeland University Hospital, Bergen, Norway
| | - Elisabeth Kjelsvik Steinsvik
- National Center for Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
- National Center for Ultrasound in Gastroenterology, Medical Department, Haukeland University Hospital, Bergen, Norway
| | - Eline Randulff Hillestad
- National Center for Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
| | - Jørgen Valeur
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
| | - Ingeborg Brønstad
- National Center for Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
- National Center for Ultrasound in Gastroenterology, Medical Department, Haukeland University Hospital, Bergen, Norway
| | - Odd Helge Gilja
- National Center for Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
- National Center for Ultrasound in Gastroenterology, Medical Department, Haukeland University Hospital, Bergen, Norway
| | - Berge Osnes
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Jan Gunnar Hatlebakk
- National Center for Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
| | - Judit Haász
- Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Jennifer Labus
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, and UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Arpana Gupta
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, and UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Emeran A. Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, and UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Alfonso Benitez-Páez
- Microbial Ecology, Nutrition & Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Yolanda Sanz
- Microbial Ecology, Nutrition & Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Arvid Lundervold
- Mohn Medical Imaging and Visualization Center, Haukeland University Hospital, Bergen, Norway
| | - Trygve Hausken
- National Center for Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
- Center for Nutrition, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
632
|
Genome sequence of segmented filamentous bacteria present in the human intestine. Commun Biol 2020; 3:485. [PMID: 32887924 PMCID: PMC7474095 DOI: 10.1038/s42003-020-01214-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 08/06/2020] [Indexed: 12/16/2022] Open
Abstract
Segmented filamentous bacteria (SFB) are unique immune modulatory bacteria colonizing the small intestine of a variety of animals in a host-specific manner. SFB exhibit filamentous growth and attach to the host’s intestinal epithelium, offering a physical route of interaction. SFB affect functions of the host immune system, among them IgA production and T-cell maturation. Until now, no human-specific SFB genome has been reported. Here, we report the metagenomic reconstruction of an SFB genome from a human ileostomy sample. Phylogenomic analysis clusters the genome with SFB genomes from mouse, rat and turkey, but the genome is genetically distinct, displaying 65–71% average amino acid identity to the others. By screening human faecal metagenomic datasets, we identified individuals carrying sequences identical to the new SFB genome. We thus conclude that a unique SFB variant exists in humans and foresee a renewed interest in the elucidation of SFB functionality in this environment. Hans Jonsson et al. report the metagenomic reconstruction of the genome of a potentially immune modulatory segmented filamentous bacteria (SFB) from a human ileostomy sample. They demonstrate that the genome clusters closely with SFB genomes from other species. They also detect the unique SFB variant in human faecal metagenomics datasets.
Collapse
|
633
|
Barraza-Ortiz DA, Pérez-López N, Medina-López VM, Minero-Alfaro JI, Zamarripa-Dorsey F, Fernández-Martínez NDC, Llorente-Ramón A, Ramos-Aguilar GA. Combination of a Probiotic and an Antispasmodic Increases Quality of Life and Reduces Symptoms in Patients with Irritable Bowel Syndrome: A Pilot Study. Dig Dis 2020; 39:294-300. [PMID: 32810850 DOI: 10.1159/000510950] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/17/2020] [Indexed: 02/02/2023]
Abstract
BACKGROUND Probiotics and antispasmodics have been tested extensively in the management of symptoms of irritable bowel syndrome (IBS), but they have rarely been evaluated in combination. The objective of this pilot study was to assess the efficacy of treatment with the probiotic formulation i3.1 (Lactobacillus plantarum CECT7484 and CECT7485 and Pediococcus acidilactici CECT7483), with or without the addition of the antispasmodic alverine/simethicone, in improving IBS-related quality of life (QoL) and reducing abdominal pain and diarrhea in patients with IBS. METHODS This was a randomized, placebo-controlled clinical trial with 3 parallel arms (probiotic, probiotic plus antispasmodic, and placebo). Patients with IBS (N = 55) were recruited at the Gastroenterology Department of the Juárez Hospital (México City). QoL was assessed with the IBS-QoL questionnaire, abdominal pain with a visual analog scale, and stool consistency with the Bristol scale. RESULTS The IBS-QoL rate of response (ITT analysis) was 50.0% for patients in the group with probiotic alone, 68.4% in the group with probiotic plus antispasmodic, and 16.7% in the group with placebo after 6 weeks of treatment (p = 0.005). Response to abdominal pain was reported by 38.9% of patients treated with probiotic, 57.9% with probiotic plus antispasmodic, and 16.7% with placebo (p = 0.035). Regarding stool consistency, a response to treatment was reported by 44.4% of patients treated with probiotic, 57.9% with probiotic plus antispasmodic, and 16.7% with placebo (p = 0.032). CONCLUSION The results are consistent with previous studies on the use of the i3.1 probiotic formulation for the management of symptoms in IBS patients, and the addition of an antispasmodic improves its observed effects.
Collapse
Affiliation(s)
- Diego A Barraza-Ortiz
- Department of Gastroenterology, Hospital Juárez de Mexico, CDMX, Ciudad de Mexico, Mexico,
| | - Nuria Pérez-López
- Department of Gastroenterology, Hospital Juárez de Mexico, CDMX, Ciudad de Mexico, Mexico
| | - Víctor M Medina-López
- Department of Gastroenterology, Hospital Juárez de Mexico, CDMX, Ciudad de Mexico, Mexico
| | - José I Minero-Alfaro
- Department of Gastroenterology, Hospital Español de Mexico, CDMX, Ciudad de Mexico, Mexico
| | | | | | - Alberto Llorente-Ramón
- Department of Gastroenterology, Hospital Juárez de Mexico, CDMX, Ciudad de Mexico, Mexico
| | | |
Collapse
|
634
|
Abstract
To investigate the effects of probiotics on liver function, glucose and lipids metabolism, and hepatic fatty deposition in patients with non-alcoholic fatty liver disease (NAFLD).Totally 140 NAFLD cases diagnosed in our hospital from March 2017 to March 2019 were randomly divided into the observation group and control group, 70 cases in each. The control group received the diet and exercise therapy, while the observation group received oral probiotics based on the control group, and the intervention in 2 groups lasted for 3 months. The indexes of liver function, glucose and lipids metabolism, NAFLD activity score (NAS), and conditions of fecal flora in 2 groups were compared before and after the treatment.Before the treatment, there were no significant differences on alanine aminotransferase (ALT), aspartate aminotransferase (AST), glutamine transferase (GGT), total bilirubin (TBIL), total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), insulin resistance index (HOMA-IR), NAFLD activity score (NAS), and conditions of fecal flora in 2 groups (P > .05). After the treatment, ALT, AST, GGT, TC, TG, HOMA-IR, NAS, and conditions of fecal flora in the observation group were better than those in the control group, and the observation group was better after treatment than before. All these above differences were statistically significant (P < .05).Probiotics can improve some liver functions, glucose and lipids metabolism, hepatic fatty deposition in patients with NAFLD, which will enhance the therapeutic effects of NAFLD.
Collapse
Affiliation(s)
| | - Hui Su
- Department of Gastroenterology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | | |
Collapse
|
635
|
Artifon M, Schestatsky P, Griebler N, Tossi GM, Beraldo LM, Pietta-Dias C. Effects of transcranial direct current stimulation on the gut microbiome: A case report. Brain Stimul 2020; 13:1451-1452. [PMID: 32771419 DOI: 10.1016/j.brs.2020.07.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/15/2022] Open
Affiliation(s)
- Milena Artifon
- Exercise Research Laboratory, Universidade Federal Do Rio Grande Do Sul - UFRGS, Brazil.
| | - Pedro Schestatsky
- Medical School, Universidade Federal Do Rio Grande Do Sul - UFRGS, Brazil
| | - Nathália Griebler
- Exercise Research Laboratory, Universidade Federal Do Rio Grande Do Sul - UFRGS, Brazil
| | - Gabriel Mayer Tossi
- Exercise Research Laboratory, Universidade Federal Do Rio Grande Do Sul - UFRGS, Brazil
| | - Lucas M Beraldo
- Mathematic and Statistics Institute, Universidade Federal Do Rio Grande Do Sul - UFRGS, Brazil
| | - Caroline Pietta-Dias
- Exercise Research Laboratory, Universidade Federal Do Rio Grande Do Sul - UFRGS, Brazil
| |
Collapse
|
636
|
Jena A, Montoya CA, Mullaney JA, Dilger RN, Young W, McNabb WC, Roy NC. Gut-Brain Axis in the Early Postnatal Years of Life: A Developmental Perspective. Front Integr Neurosci 2020; 14:44. [PMID: 32848651 PMCID: PMC7419604 DOI: 10.3389/fnint.2020.00044] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence suggests that alterations in the development of the gastrointestinal (GI) tract during the early postnatal period can influence brain development and vice-versa. It is increasingly recognized that communication between the GI tract and brain is mainly driven by neural, endocrine, immune, and metabolic mediators, collectively called the gut-brain axis (GBA). Changes in the GBA mediators occur in response to the developmental changes in the body during this period. This review provides an overview of major developmental events in the GI tract and brain in the early postnatal period and their parallel developmental trajectories under physiological conditions. Current knowledge of GBA mediators in context to brain function and behavioral outcomes and their synthesis and metabolism (site, timing, etc.) is discussed. This review also presents hypotheses on the role of the GBA mediators in response to the parallel development of the GI tract and brain in infants.
Collapse
Affiliation(s)
- Ankita Jena
- School of Food & Advanced Technology, College of Sciences, Massey University, Palmerston North, New Zealand.,The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand
| | - Carlos A Montoya
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand
| | - Jane A Mullaney
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Ryan N Dilger
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Wayne Young
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Warren C McNabb
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Nicole C Roy
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand.,Liggins Institute, The University of Auckland, Auckland, New Zealand.,Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| |
Collapse
|
637
|
Xie W, Strong JA, Zhang JM. Localized sympathectomy reduces peripheral nerve regeneration and pain behaviors in 2 rat neuropathic pain models. Pain 2020; 161:1925-1936. [PMID: 32701850 PMCID: PMC7572566 DOI: 10.1097/j.pain.0000000000001887] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Previous studies have shown that the peripheral nerve regeneration process is linked to pain in several neuropathic pain models. Other studies show that sympathetic blockade may relieve pain in some pain models and clinical conditions. This study examined reduction in peripheral nerve regeneration as one possible mechanism for relief of neuropathic pain by sympathetic blockade. A "microsympathectomy," consisting of cutting the gray rami containing sympathetic postganglionic axons where they enter the L4 and L5 spinal nerves, reduced mechanical hypersensitivity in 2 different rat neuropathic pain models. In the spinal nerve ligation model, in which some functional regeneration and reinnervation of the ligated spinal nerve can be observed, microsympathectomy reduced functional and anatomical measures of regeneration as well as expression of growth-associated protein 43 (GAP43), a regeneration-related protein. In the spared nerve injury model, in which functional reinnervation is not possible and the futile regeneration process results in formation of a neuroma, microsympathectomy reduced neuroma formation and GAP43 expression. In both models, microsympathectomy reduced macrophage density in the sensory ganglia and peripheral nerve. This corroborates previous work showing that sympathetic nerves may locally affect immune function. The results further highlight the challenge of improving pain in neuropathic conditions without inhibiting peripheral nerve regeneration that might otherwise be possible and desired.
Collapse
Affiliation(s)
- Wenrui Xie
- Department of Anesthesiology, Pain Research Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | | | | |
Collapse
|
638
|
Dong TS, Mayer EA, Osadchiy V, Chang C, Katzka W, Lagishetty V, Gonzalez K, Kalani A, Stains J, Jacobs JP, Longo VD, Gupta A. A Distinct Brain-Gut-Microbiome Profile Exists for Females with Obesity and Food Addiction. Obesity (Silver Spring) 2020; 28:1477-1486. [PMID: 32935533 PMCID: PMC7494955 DOI: 10.1002/oby.22870] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/07/2020] [Accepted: 04/21/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Alterations in brain-gut-microbiome interactions have been implicated as an important factor in obesity. This study aimed to explore the relationship between food addiction (FA) and the brain-gut-microbiome axis, using a multi-omics approach involving microbiome data, metabolomics, and brain imaging. METHODS Brain magnetic resonance imaging was obtained in 105 females. FA was defined by using the Yale Food Addiction Scale. Fecal samples were collected for sequencing and metabolomics. Statistical analysis was done by using multivariate analyses and machine learning algorithms. RESULTS Of the females with obesity, 33.3% exhibited FA as compared with 5.3% and 0.0% of females with overweight and normal BMI, respectively (P = 0.0001). Based on a multilevel sparse partial least square discriminant analysis, there was a difference in the gut microbiome of females with FA versus those without. Differential abundance testing showed Bacteroides, Megamonas, Eubacterium, and Akkermansia were statistically associated with FA (q < 0.05). Metabolomics showed that indolepropionic acid was inversely correlated with FA. FA was also correlated with increased connectivity within the brain's reward network, specifically between the intraparietal sulcus, brain stem, and putamen. CONCLUSIONS This is the first study to examine FA along the brain-gut-microbiome axis and it supports the idea of targeting the brain-gut-microbiome axis for the treatment of FA and obesity.
Collapse
Affiliation(s)
- Tien S. Dong
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- David Geffen School of Medicine, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- UCLA Microbiome Center, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Emeran A. Mayer
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- David Geffen School of Medicine, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- UCLA Microbiome Center, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Vadim Osadchiy
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Candace Chang
- UCLA Microbiome Center, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - William Katzka
- UCLA Microbiome Center, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Venu Lagishetty
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- UCLA Microbiome Center, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Kimberly Gonzalez
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Amir Kalani
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Jean Stains
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Jonathan P. Jacobs
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- David Geffen School of Medicine, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- UCLA Microbiome Center, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Valter D. Longo
- USC Longevity Institute, University of Southern California, Los Angeles
| | - Arpana Gupta
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- David Geffen School of Medicine, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- UCLA Microbiome Center, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| |
Collapse
|
639
|
Liu RT, Rowan-Nash AD, Sheehan AE, Walsh RFL, Sanzari CM, Korry BJ, Belenky P. Reductions in anti-inflammatory gut bacteria are associated with depression in a sample of young adults. Brain Behav Immun 2020; 88:308-324. [PMID: 32229219 PMCID: PMC7415740 DOI: 10.1016/j.bbi.2020.03.026] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/07/2020] [Accepted: 03/25/2020] [Indexed: 12/15/2022] Open
Abstract
We assessed the gut microbiota of 90 American young adults, comparing 43 participants with major depressive disorder (MDD) and 47 healthy controls, and found that the MDD subjects had significantly different gut microbiota compared to the healthy controls at multiple taxonomic levels. At the phylum level, participants with MDD had lower levels of Firmicutes and higher levels of Bacteroidetes, with similar trends in the at the class (Clostridia and Bacteroidia) and order (Clostridiales and Bacteroidales) levels. At the genus level, the MDD group had lower levels of Faecalibacterium and other related members of the family Ruminococcaceae, which was also reduced relative to healthy controls. Additionally, the class Gammaproteobacteria and genus Flavonifractor were enriched in participants with MDD. Accordingly, predicted functional differences between the two groups include a reduced abundance of short-chain fatty acid production pathways in the MDD group. We also demonstrated that the magnitude of taxonomic changes was associated with the severity of depressive symptoms in many cases, and that most changes were present regardless of whether depressed participants were taking psychotropic medications. Overall, our results support a link between MDD and lower levels of anti-inflammatory, butyrate-producing bacteria, and may support a connection between the gut microbiota and the chronic, low-grade inflammation often observed in MDD patients.
Collapse
Affiliation(s)
- Richard T Liu
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA.
| | - Aislinn D Rowan-Nash
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Ana E Sheehan
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, USA
| | - Rachel F L Walsh
- Department of Psychology, Temple University, Philadelphia, PA, USA
| | - Christina M Sanzari
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
| | - Benjamin J Korry
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| |
Collapse
|
640
|
Steinway SN, Saleh J, Koo BK, Delacour D, Kim DH. Human Microphysiological Models of Intestinal Tissue and Gut Microbiome. Front Bioeng Biotechnol 2020; 8:725. [PMID: 32850690 PMCID: PMC7411353 DOI: 10.3389/fbioe.2020.00725] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal (GI) tract is a complex system responsible for nutrient absorption, digestion, secretion, and elimination of waste products that also hosts immune surveillance, the intestinal microbiome, and interfaces with the nervous system. Traditional in vitro systems cannot harness the architectural and functional complexity of the GI tract. Recent advances in organoid engineering, microfluidic organs-on-a-chip technology, and microfabrication allows us to create better in vitro models of human organs/tissues. These micro-physiological systems could integrate the numerous cell types involved in GI development and physiology, including intestinal epithelium, endothelium (vascular), nerve cells, immune cells, and their interplay/cooperativity with the microbiome. In this review, we report recent progress in developing micro-physiological models of the GI systems. We also discuss how these models could be used to study normal intestinal physiology such as nutrient absorption, digestion, and secretion as well as GI infection, inflammation, cancer, and metabolism.
Collapse
Affiliation(s)
- Steven N. Steinway
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jad Saleh
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR 7592, Paris Diderot University, Paris, France
| | - Bon-Kyoung Koo
- Institute of Molecular Biotechnology, Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - Delphine Delacour
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR 7592, Paris Diderot University, Paris, France
| | - Deok-Ho Kim
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
641
|
Ghanemi A, Melouane A, Yoshioka M, St-Amand J. Exercise and High-Fat Diet in Obesity: Functional Genomics Perspectives of Two Energy Homeostasis Pillars. Genes (Basel) 2020; 11:genes11080875. [PMID: 32752100 PMCID: PMC7463441 DOI: 10.3390/genes11080875] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022] Open
Abstract
The heavy impact of obesity on both the population general health and the economy makes clarifying the underlying mechanisms, identifying pharmacological targets, and developing efficient therapies for obesity of high importance. The main struggle facing obesity research is that the underlying mechanistic pathways are yet to be fully revealed. This limits both our understanding of pathogenesis and therapeutic progress toward treating the obesity epidemic. The current anti-obesity approaches are mainly a controlled diet and exercise which could have limitations. For instance, the “classical” anti-obesity approach of exercise might not be practical for patients suffering from disabilities that prevent them from routine exercise. Therefore, therapeutic alternatives are urgently required. Within this context, pharmacological agents could be relatively efficient in association to an adequate diet that remains the most efficient approach in such situation. Herein, we put a spotlight on potential therapeutic targets for obesity identified following differential genes expression-based studies aiming to find genes that are differentially expressed under diverse conditions depending on physical activity and diet (mainly high-fat), two key factors influencing obesity development and prognosis. Such functional genomics approaches contribute to elucidate the molecular mechanisms that both control obesity development and switch the genetic, biochemical, and metabolic pathways toward a specific energy balance phenotype. It is important to clarify that by “gene-related pathways”, we refer to genes, the corresponding proteins and their potential receptors, the enzymes and molecules within both the cells in the intercellular space, that are related to the activation, the regulation, or the inactivation of the gene or its corresponding protein or pathways. We believe that this emerging area of functional genomics-related exploration will not only lead to novel mechanisms but also new applications and implications along with a new generation of treatments for obesity and the related metabolic disorders especially with the modern advances in pharmacological drug targeting and functional genomics techniques.
Collapse
Affiliation(s)
- Abdelaziz Ghanemi
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada; (A.G.); (A.M.)
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada;
| | - Aicha Melouane
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada; (A.G.); (A.M.)
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada;
| | - Mayumi Yoshioka
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada;
| | - Jonny St-Amand
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada; (A.G.); (A.M.)
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada;
- Correspondence: ; Tel.: +1-418-654-2296; Fax: +1-418-654-2761
| |
Collapse
|
642
|
Prinelli F, Jesuthasan N, Severgnini M, Musicco M, Adorni F, Correa Leite ML, Crespi C, Bernini S. Exploring the relationship between Nutrition, gUT microbiota, and BRain AgINg in community-dwelling seniors: the Italian NutBrain population-based cohort study protocol. BMC Geriatr 2020; 20:253. [PMID: 32703186 PMCID: PMC7376643 DOI: 10.1186/s12877-020-01652-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
Background Epidemiological evidence suggests that healthy diet is associated with a slowdown of cognitive decline leading to dementia, but the underlying mechanisms are still partially unexplored. Diet is the main determinant of gut microbiota composition, which in turn impacts on brain structures and functions, however to date no studies on this topic are available. The goal of the present paper is to describe the design and methodology of the NutBrain Study aimed at investigating the association of dietary habits with cognitive function and their role in modulating the gut microbiota composition, and brain measures as well. Methods/design This is a population-based cohort study of community-dwelling adults aged 65 years or more living in Northern Milan, Italy. At the point of presentation people are screened for cognitive functions. Socio-demographic characteristics along with lifestyles and dietary habits, medical history, drugs, functional status, and anthropometric measurements are also recorded. Individuals suspected to have cognitive impairment at the screening phase undergo a clinical evaluation including a neurological examination and a Magnetic Resonance Imaging (MRI) scanning (both structural and functional). Stool and blood samples for the gut microbiota analysis and for the evaluation of putative biological markers are also collected. For each subject with a confirmed diagnosis of Mild Cognitive Impairment (MCI), two cognitively intact controls of the same sex and age are visited. We intend to enrol at least 683 individuals for the screening phase and 240 persons for the clinical assessment. Discussion The NutBrain is an innovative study that incorporates modern and advanced technologies (i.e. microbiome and neuroimaging) into traditional epidemiologic design. The study represents a unique opportunity to address key questions about the role of modifiable risk factors on cognitive impairment, with a particular focus on dietary habits and their association with gut microbiota and markers of the brain-aging process. These findings will help to encourage and plan lifestyle interventions, for both prevention and treatment, aiming at promoting healthy cognitive ageing. Trial registration Trial registration number NCT04461951, date of registration July 7, 2020 (retrospectively registered, ClinicalTrials.gov).
Collapse
Affiliation(s)
- Federica Prinelli
- Institute of Biomedical Technologies-National Research Council, Via Fratelli Cervi, 93 20090, Segrate, MI, Italy. .,IRCCS Mondino Foundation, Neuropsychology/Alzheimer's Disease Assessment Unit, Via Mondino 2, 27100, Pavia, Italy.
| | - Nithiya Jesuthasan
- Institute of Biomedical Technologies-National Research Council, Via Fratelli Cervi, 93 20090, Segrate, MI, Italy
| | - Marco Severgnini
- Institute of Biomedical Technologies-National Research Council, Via Fratelli Cervi, 93 20090, Segrate, MI, Italy
| | - Massimo Musicco
- Institute of Biomedical Technologies-National Research Council, Via Fratelli Cervi, 93 20090, Segrate, MI, Italy
| | - Fulvio Adorni
- Institute of Biomedical Technologies-National Research Council, Via Fratelli Cervi, 93 20090, Segrate, MI, Italy
| | - Maria Lea Correa Leite
- Institute of Biomedical Technologies-National Research Council, Via Fratelli Cervi, 93 20090, Segrate, MI, Italy
| | - Chiara Crespi
- Scuola Universitaria Superiore IUSS Pavia, Nets Center, Piazza della Vittoria, 15 -, 27100, Pavia, Italy
| | - Sara Bernini
- IRCCS Mondino Foundation, Neuropsychology/Alzheimer's Disease Assessment Unit, Via Mondino 2, 27100, Pavia, Italy
| |
Collapse
|
643
|
Minireview Exploring the Biological Cycle of Vitamin B3 and Its Influence on Oxidative Stress: Further Molecular and Clinical Aspects. Molecules 2020; 25:molecules25153323. [PMID: 32707945 PMCID: PMC7436124 DOI: 10.3390/molecules25153323] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/13/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
Vitamin B3, or niacin, is one of the most important compounds of the B-vitamin complex. Recent reports have demonstrated the involvement of vitamin B3 in a number of pivotal functions which ensure that homeostasis is maintained. In addition, the intriguing nature of its synthesis and the underlying mechanism of action of vitamin B3 have encouraged further studies aimed at deepening our understanding of the close link between the exogenous supply of B3 and how it activates dependent enzymes. This crucial role can be attributed to the gut microflora and its ability to shape human behavior and development by mediating the bioavailability of metabolites. Recent studies have indicated a possible interconnection between the novel coronavirus and commensal bacteria. As such, we have attempted to explain how the gastrointestinal deficiencies displayed by SARS-CoV-2-infected patients arise. It seems that the stimulation of a proinflammatory cascade and the production of large amounts of reactive oxygen species culminates in the subsequent loss of host eubiosis. Studies of the relationhip between ROS, SARS-CoV-2, and gut flora are sparse in the current literature. As an integrated component, oxidative stress (OS) has been found to negatively influence host eubiosis, in vitro fertilization outcomes, and oocyte quality, but to act as a sentinel against infections. In conclusion, research suggests that in the future, a healthy diet may be considered a reliable tool for maintaining and optimizing our key internal parameters.
Collapse
|
644
|
Slavin M, Li HA, Frankenfeld C, Cheskin LJ. What is Needed for Evidence-Based Dietary Recommendations for Migraine: A Call to Action for Nutrition and Microbiome Research. Headache 2020; 59:1566-1581. [PMID: 31603554 DOI: 10.1111/head.13658] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The gastrointestinal symptoms of migraine attacks have invited numerous dietary hypotheses for migraine etiology through the centuries. Substantial efforts have been dedicated to identifying dietary interventions for migraine attack prevention, with limited success. Meanwhile, mounting evidence suggests that the reverse relationship may also exist - that the biological mechanisms of migraine may influence dietary intake. More likely, the truth involves some combination of both, where the disease influences food intake, and the foods eaten impact the manifestations of the disease. In addition, the gut's microbiota is increasingly suspected to influence the migraine brain via the gut-brain axis, though these hypotheses remain largely unsubstantiated. OBJECTIVE This paper presents an overview of the strength of existing evidence for food-based dietary interventions for migraine, noting that there is frequently evidence to suggest that a dietary risk factor for migraine exists but no evidence for how to best intervene; in fact, our intuitive assumptions on interventions are being challenged with new evidence. We then look to the future for promising avenues of research, notably the gut microbiome. CONCLUSION The evidence supports a call to action for high-quality dietary and microbiome research in migraine, both to substantiate hypothesized relationships and build the evidence base regarding nutrition's potential impact on migraine attack prevention and treatment.
Collapse
Affiliation(s)
- Margaret Slavin
- Department of Nutrition and Food Studies, George Mason University, Fairfax, VA, USA
| | - Huilun Amber Li
- Department of Nutrition and Food Studies, George Mason University, Fairfax, VA, USA
| | - Cara Frankenfeld
- Department of Global and Community Health, George Mason University, Fairfax, VA, USA
| | - Lawrence J Cheskin
- Department of Nutrition and Food Studies, George Mason University, Fairfax, VA, USA
| |
Collapse
|
645
|
Lim EY, Lee SY, Shin HS, Lee J, Nam YD, Lee DO, Lee JY, Yeon SH, Son RH, Park CL, Heo YH, Kim YT. The Effect of Lactobacillus acidophilus YT1 (MENOLACTO) on Improving Menopausal Symptoms: A Randomized, Double-Blinded, Placebo-Controlled Clinical Trial. J Clin Med 2020; 9:E2173. [PMID: 32660010 PMCID: PMC7408745 DOI: 10.3390/jcm9072173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/02/2020] [Accepted: 07/07/2020] [Indexed: 12/18/2022] Open
Abstract
This study evaluated the efficacy of Lactobacillus acidophilus YT1 (MENOLACTO) for alleviating menopausal symptoms. This study was a multi-center, randomized, double-blinded, placebo-controlled clinical trial involving female subjects (ages: 40-60 years) with menopausal symptoms and a Kupperman index (KMI) score ≥ 20. Subjects were administered 1 × 108 CFU/day MENOLACTO or placebo, with the primary endpoint being total KMI score, and the effect of secondary endpoints on alleviating menopausal symptoms according to individual categories of the modified KMI, as well as a quality of life questionnaire (MENQOL questionnaire). After 12 weeks, total KMI scores decreased significantly, demonstrating improved menopausal symptoms relative to placebo along with improved modified KMI scores. Additionally, quality of life, according to the MENQOL questionnaire, significantly improved in all four symptoms-physical, psychosocial, vasomotor, and sexual symptoms. Moreover, we observed no significant difference between the two groups or significant changes in blood follicle-stimulating hormone and estradiol levels or endometrial thickness. These results demonstrated that MENOLACTO alleviated menopausal symptoms without notable side effects and improved quality of life, suggesting its efficacy as an alternative supplement to alleviate menopausal symptoms in women ineligible for hormonal therapy.
Collapse
Affiliation(s)
- Eun Yeong Lim
- Division of Food Functionality Research, Korea Food Research Institute, Wanju 55365, Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.); (J.L.); (Y.-D.N.)
- Department of Food Biotechnology, Korea University of Science & Technology, Daejeon 34113, Korea
| | - So-Young Lee
- Division of Food Functionality Research, Korea Food Research Institute, Wanju 55365, Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.); (J.L.); (Y.-D.N.)
- Department of Food Biotechnology, Korea University of Science & Technology, Daejeon 34113, Korea
| | - Hee Soon Shin
- Division of Food Functionality Research, Korea Food Research Institute, Wanju 55365, Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.); (J.L.); (Y.-D.N.)
- Department of Food Biotechnology, Korea University of Science & Technology, Daejeon 34113, Korea
| | - Jaekwang Lee
- Division of Food Functionality Research, Korea Food Research Institute, Wanju 55365, Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.); (J.L.); (Y.-D.N.)
| | - Young-Do Nam
- Division of Food Functionality Research, Korea Food Research Institute, Wanju 55365, Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.); (J.L.); (Y.-D.N.)
- Department of Food Biotechnology, Korea University of Science & Technology, Daejeon 34113, Korea
| | - Dong Ock Lee
- Center for Gynecologic Cancer, National Cancer Center Korea, Goyang-si 10408, Korea;
| | - Ji Young Lee
- Department of Obstetrics and Gynecology, Konkuk University Hospital, Konkuk University School of Medicine, Seoul 05030, Korea;
| | - Sung Hum Yeon
- R&D Center, Huons Co., Ltd., Ansan 15588, Korea; (S.H.Y.); (R.H.S.); (C.L.P.)
| | - Rak Ho Son
- R&D Center, Huons Co., Ltd., Ansan 15588, Korea; (S.H.Y.); (R.H.S.); (C.L.P.)
| | - Chae Lee Park
- R&D Center, Huons Co., Ltd., Ansan 15588, Korea; (S.H.Y.); (R.H.S.); (C.L.P.)
| | - Yun Haeng Heo
- Clinical Operation Team, Huons Co., Ltd., Seongnam-si 13486, Korea;
| | - Yun Tai Kim
- Division of Food Functionality Research, Korea Food Research Institute, Wanju 55365, Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.); (J.L.); (Y.-D.N.)
- Department of Food Biotechnology, Korea University of Science & Technology, Daejeon 34113, Korea
| |
Collapse
|
646
|
Spontaneous remission of infantile spasms following rotavirus gastroenteritis. Neurol Sci 2020; 42:253-257. [PMID: 32632632 DOI: 10.1007/s10072-020-04564-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 07/02/2020] [Indexed: 10/23/2022]
Abstract
Epileptic seizures might be provoked and/or exacerbated by fever or viral infection in children with epilepsy. However, this is not true for infantile spasms; in this study, we report three cases with infantile spasms became seizure free within 2-5 days following rotavirus gastroenteritis without an exchange or addition of antiepileptic drugs, and hypsarrhythmia evolved to diffuse slow waves or localized spikes on electroencephalography. We propose that the probability regarding the mechanism of spontaneous mitigation is the suppression of immunopathological processes caused by infection, while the possibility of ketogenic effects of diarrhea and intestinal flora recombination after rotavirus gastroenteritis is unlikely. Further study may provide important information concerning the mechanism of seizure control and the applicability to treatment for infantile spasms.
Collapse
|
647
|
Bai YF, Gao C, Li WJ, Du Y, An LX. Transcutaneous electrical acupuncture stimulation (TEAS) for gastrointestinal dysfunction in adults undergoing abdominal surgery: study protocol for a prospective randomized controlled trial. Trials 2020; 21:617. [PMID: 32631387 PMCID: PMC7336398 DOI: 10.1186/s13063-020-04470-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/30/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Postoperative gastrointestinal (GI) dysfunction (PGD) is a common problem after abdominal surgery. PGD can increase the length of hospital stay and may lead to serious complications. Acupuncture and moxibustion are alternative therapies for PGD that have been used in some settings. However, the effect of preventive application of acupuncture or transcutaneous electrical acupuncture stimulation (TEAS) is still uncertain. The purpose of this study is to investigate the efficacy of the continuous application of TEAS on GI function recovery in adults undergoing abdominal surgery. At the same time, we will try to confirm the mechanism of TEAS through the brain-gut axis. METHODS/DESIGN This study is a prospective, single-center, two-arm, randomized controlled trial that will be performed in a general hospital. In total, 280 patients undergoing abdominal surgery were stratified by type of surgery (i.e. gastric or colorectal procedure) and randomized into two treatment groups. The experimental group will receive TEAS stimulation at L14 and PC6, ST36 and ST37. The sham group will receive pseudo-TEAS at sham acupoints. The primary outcome will be the time to the first bowel motion by auscultation. The recovery time of flatus, defecation, the changes in perioperative brain-intestinal peptides, postoperative pain, perioperative complications, and hospitalization duration will be the secondary outcomes. DISCUSSION The results of this study will demonstrate that continuous preventive application of TEAS can improve the GI function recovery in patients undergoing abdominal surgery and that this effect may act through brain-gut peptides. TRIAL REGISTRATION Chinese Clinical Trial Registry, ChiCTR1900023263 . Registered on 11 May 2019.
Collapse
Affiliation(s)
- Ya-Fan Bai
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing, 100050 China
| | - Chao Gao
- Department of Anesthesiology, Beijing Huimin Hospital, Beijing, China
| | - Wen-Jing Li
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing, 100050 China
| | - Yi Du
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Li-Xin An
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing, 100050 China
| |
Collapse
|
648
|
Noonan S, Zaveri M, Macaninch E, Martyn K. Food & mood: a review of supplementary prebiotic and probiotic interventions in the treatment of anxiety and depression in adults. BMJ Nutr Prev Health 2020; 3:351-362. [PMID: 33521545 PMCID: PMC7841823 DOI: 10.1136/bmjnph-2019-000053] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/19/2020] [Accepted: 02/23/2020] [Indexed: 02/06/2023] Open
Abstract
Background A bidirectional relationship exists between the brain and the gastrointestinal tract. Foods containing bacteria that positively influence the gastrointestinal microbiome are termed, probiotics; compounds that promote the flourishing of these bacteria are termed, prebiotics. Whether microbiome influencing therapies could treat psychiatric conditions, including depression and anxiety, is an area of interest. Presently, no established consensus for such treatment exists. Methods This systematic review analyses databases and grey literature sites to investigate pre and/or probiotics as treatments for depression and/or anxiety disorders. Articles included are from within 15 years. Pre-determined inclusion exclusion criteria were applied, and articles were appraised for their quality using a modified-CASP checklist. This review focuses specifically on quantitative measures from patients with clinical diagnoses of depression and/or anxiety disorders. Results 7 studies were identified. All demonstrated significant improvements in one or more of the outcomes measuring the of effect taking pre/probiotics compared with no treatment/placebo, or when compared to baseline measurements. Discussion Our review suggests utilising pre/probiotic may be a potentially useful adjunctive treatment. Furthermore, patients with certain co-morbidities, such as IBS, might experience greater benefits from such treatments, given that pre/probiotic are useful treatments for other conditions that were not the primary focus of this discourse. Our results are limited by several factors: sample sizes (adequate, though not robust); short study durations, long-term effects and propensity for remission undetermined. Conclusion Our results affirm that pre/probiotic therapy warrants further investigation. Efforts should aim to elucidate whether the perceived efficacy of pre/probiotic therapy in depression and/or anxiety disorders can be replicated in larger test populations, and whether such effects are maintained through continued treatment, or post cessation. Interventions should also be investigated in isolation, not combination, to ascertain where the observed effects are attributable to. Efforts to produce mechanistic explanations for such effect should be a priority.
Collapse
Affiliation(s)
| | | | - Elaine Macaninch
- Nutrition and dietetics, Brighton and Sussex University Hospitals NHS Trust, Brighton, East Sussex, UK
| | | |
Collapse
|
649
|
Preparation of a Unique Bioavailable Bacoside Formulation (Cognique®) Using Polar-Nonpolar-Sandwich (PNS) Technology and Its Characterization, In Vitro Release Study, and Proposed Mechanism of Action. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020. [DOI: 10.1007/s40883-020-00162-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
650
|
Everett GJ, Jafferany M, Skurya J. Recent advances in the treatment of trichotillomania (hair-pulling disorder). Dermatol Ther 2020; 33:e13818. [PMID: 32531098 DOI: 10.1111/dth.13818] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/16/2022]
Abstract
Trichotillomania (TTM) is a condition in which affected individuals pull out their hair resulting in hair loss. This disorder affects roughly 0.5% to 2.0% of the population and can have significant psychological morbidity. Behavioral therapy has been used with success in the treatment of TTM, but not all patients are willing or able to comply with this treatment strategy. There is a need for effective pharmacological treatment options. Historically, pharmacotherapy for TTM has been inadequate in most cases, but recent advances have been made in this regard. Fluoxetine, clomipramine, olanzapine, and naltrexone have all been used in the treatment of TTM, but evidence of benefit has varied, and side effect profiles can limit practical utility. Recent advances in the understanding of the pathophysiology of TTM, as well as evidence of benefit seen with some glutamate-modulating agents such as N-acetylcysteine and dronabinol, have provided newer potential pharmacotherapy options.
Collapse
Affiliation(s)
- Gregory J Everett
- Division of Psychodermatology, Central Michigan University, Saginaw, Michigan, USA
| | - Mohammad Jafferany
- Division of Psychodermatology, Central Michigan University, Saginaw, Michigan, USA
| | - Jonathon Skurya
- Division of Psychodermatology, Central Michigan University, Saginaw, Michigan, USA
| |
Collapse
|