1
|
Li M, Fu F, Wang T. Escin alleviates cerebral ischemia-induced intestinal pyroptosis via the GR-dependent p38 MAPK/NF-κB signaling and NLRP3 inflammasome activation. Int Immunopharmacol 2024; 138:112592. [PMID: 38955024 DOI: 10.1016/j.intimp.2024.112592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024]
Abstract
Cerebral ischemia-induced systemic inflammation and inflammasome-dependent pyroptotic cell death in ileum, causing serious intestinal injury. Glucocorticoid receptor (GR) mediates the effects of glucocorticoids and participates in inflammation. Escin has corticosteroid-like, neuroprotective, and anti-intestinal dysfunction effects. This study aimed to investigate the effect of Escin on the intestinal barrier injury in rats subjected to middle cerebral artery occlusion (MCAO) and on Caco-2 cells exposed to lipopolysaccharides. The MCAO-caused brain injury was evaluated by assessing neurological function, cerebral infarct volume, and plasma corticosterone (Cort) levels. Intestinal injury was evaluated by observing the histopathological changes, assessing the intestinal barrier function, and determining blood FD4, endotoxin and IL-1β levels. The levels of the tight-junction proteins such as claudin-1, occludin, and ZO-1, and proteins involved in the GR/p38 MAPK/NF-κB pathway and NLRP3-inflammasome activation were evaluated using western blotting or immunofluorescence. Administration of Escin suppressed the cerebral ischemia-induced increases in Garcia-test scores and infarct volume, alleviated the injury to the intestinal barrier, and decreased the levels of Cort, endotoxin, and IL-1β. Additionally, Escin upregulated GR and downregulated phospho(p)-p65, p-p38MAPK, NLRP3, GSDMD-N, and cleaved-caspase-1 in the intestine. The effects of Escin could be suppressed by the GR antagonist RU486 or enhanced by the p38 MAPK antagonist SB203580. We revealed details how Escin improves cerebral ischemia-induced intestinal barrier injury by upregulating GR and thereby inhibiting the pyroptosis induced by NF-κB-mediated NLRP3 activation. This study will provide a experimental foundation for the features of glucocorticoid-like activity and the discovery of new clinical application for Escin.
Collapse
Affiliation(s)
- Min Li
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China; School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China.
| | - Fenghua Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Tian Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China.
| |
Collapse
|
2
|
Simpson JB, Walker ME, Sekela JJ, Ivey SM, Jariwala PB, Storch CM, Kowalewski ME, Graboski AL, Lietzan AD, Walton WG, Davis KA, Cloer EW, Borlandelli V, Hsiao YC, Roberts LR, Perlman DH, Liang X, Overkleeft HS, Bhatt AP, Lu K, Redinbo MR. Gut microbial β-glucuronidases influence endobiotic homeostasis and are modulated by diverse therapeutics. Cell Host Microbe 2024; 32:925-944.e10. [PMID: 38754417 PMCID: PMC11176022 DOI: 10.1016/j.chom.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/18/2024] [Accepted: 04/24/2024] [Indexed: 05/18/2024]
Abstract
Hormones and neurotransmitters are essential to homeostasis, and their disruptions are connected to diseases ranging from cancer to anxiety. The differential reactivation of endobiotic glucuronides by gut microbial β-glucuronidase (GUS) enzymes may influence interindividual differences in the onset and treatment of disease. Using multi-omic, in vitro, and in vivo approaches, we show that germ-free mice have reduced levels of active endobiotics and that distinct gut microbial Loop 1 and FMN GUS enzymes drive hormone and neurotransmitter reactivation. We demonstrate that a range of FDA-approved drugs prevent this reactivation by intercepting the catalytic cycle of the enzymes in a conserved fashion. Finally, we find that inhibiting GUS in conventional mice reduces free serotonin and increases its inactive glucuronide in the serum and intestines. Our results illuminate the indispensability of gut microbial enzymes in sustaining endobiotic homeostasis and indicate that therapeutic disruptions of this metabolism promote interindividual response variabilities.
Collapse
Affiliation(s)
- Joshua B Simpson
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Morgan E Walker
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Joshua J Sekela
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Samantha M Ivey
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Parth B Jariwala
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Cameron M Storch
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Mark E Kowalewski
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA
| | - Amanda L Graboski
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Adam D Lietzan
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William G Walton
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Kacey A Davis
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA
| | - Erica W Cloer
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Valentina Borlandelli
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Yun-Chung Hsiao
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lee R Roberts
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA
| | - David H Perlman
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA
| | - Xue Liang
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA 02141, USA
| | - Hermen S Overkleeft
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Aadra P Bhatt
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kun Lu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew R Redinbo
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA; Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Brenner DM, Ladewski AM, Kinsinger SW. Development and Current State of Digital Therapeutics for Irritable Bowel Syndrome. Clin Gastroenterol Hepatol 2024; 22:222-234. [PMID: 37743035 DOI: 10.1016/j.cgh.2023.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND & AIMS Irritable bowel syndrome (IBS) is a common, debilitating disorder characterized by abdominal pain and disordered bowel habits. Current pharmacologic treatments often provide incomplete symptom relief and may be poorly tolerated. Furthermore, alleviation of gastrointestinal symptoms does not always translate into improved quality of life for IBS patients. Current treatment guidelines recommend brain-gut behavior therapy (BGBT) in conjunction with other IBS therapies, and, in randomized controlled trials, BGBT has been shown to improve symptoms, patient satisfaction, functioning, and quality of life. Access to BGBT is limited by lack of adequately trained gastrointestinal psychologists, patient time constraints, and cost. Furthermore, clinician knowledge that BGBT is specific, and different from psychotherapy approaches for common mental health disorders, may limit referrals even where available. This review provides an overview of the pathophysiology of IBS, disease burden, unmet therapeutic needs, evidence base of novel digital therapeutics for IBS, and guidance on the introduction and appropriateness of these interventions for patients. METHODS We searched the literature for available published data relating to the use of novel digital therapeutics to provide cognitive behavioral therapy and gut-directed hypnotherapy in the treatment of irritable bowel syndrome. RESULTS Clinical trial data support the development and utility of digital therapeutics designed to deliver self-guided cognitive behavioral therapy and hypnotherapy for the treatment of IBS. CONCLUSIONS BGBTs are effective, guideline-recommended treatments for IBS. Digital therapeutic devices offer accessible, cost-effective treatment options for delivery of adjunctive BGBT for the treatment of IBS. The decision to recommend digital BGBTs should be guided by careful patient assessment that includes mental health screening and risk assessment.
Collapse
Affiliation(s)
- Darren M Brenner
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| | - Amy M Ladewski
- Department of Digestive Health, Digestive Health Center, Northwestern Memorial Hospital, Chicago, Illinois
| | - Sarah Wimberly Kinsinger
- Division of Gastroenterology and Nutrition, Department of Medicine, Loyola University Medical Center, Maywood, Illinois
| |
Collapse
|
4
|
Zhu H, Wang W, Li Y. The interplay between microbiota and brain-gut axis in epilepsy treatment. Front Pharmacol 2024; 15:1276551. [PMID: 38344171 PMCID: PMC10853364 DOI: 10.3389/fphar.2024.1276551] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 01/12/2024] [Indexed: 08/12/2024] Open
Abstract
The brain-gut axis plays a vital role in connecting the cognitive and emotional centers of the brain with the intricate workings of the intestines. An imbalance in the microbiota-mediated brain-gut axis extends far beyond conditions like Irritable Bowel Syndrome (IBS) and obesity, playing a critical role in the development and progression of various neurological disorders, including epilepsy, depression, Alzheimer's disease (AD), and Parkinson's disease (PD). Epilepsy, a brain disorder characterized by unprovoked seizures, affects approximately 50 million people worldwide. Accumulating evidence suggests that rebuilding the gut microbiota through interventions such as fecal microbiota transplantation, probiotics, and ketogenic diets (KD) can benefit drug-resistant epilepsy. The disturbances in the gut microbiota could contribute to the toxic side effects of antiepileptic drugs and the development of drug resistance in epilepsy patients. These findings imply the potential impact of the gut microbiota on epilepsy and suggest that interventions targeting the microbiota, such as the KD, hold promise for managing and treating epilepsy. However, the full extent of the importance of microbiota in epilepsy treatment is not yet fully understood, and many aspects of this field remain unclear. Therefore, this article aims to provide an overview of the clinical and animal evidence supporting the regulatory role of gut microbiota in epilepsy, and of potential pathways within the brain-gut axis that may be influenced by the gut microbiota in epilepsy. Furthermore, we will discuss the recent advancements in epilepsy treatment, including the KD, fecal microbiota transplantation, and antiseizure drugs, all from the perspective of the gut microbiota.
Collapse
Affiliation(s)
- Hanxiao Zhu
- Department of Neurology, The First Affiliated Hospital of Dali University, Dali, China
- Clinical Medical School, Dali University, Dali, China
| | - Wei Wang
- Neurobiology Laboratory, China Agricultural University, Beijing, China
| | - Yun Li
- Department of Neurology, The First Affiliated Hospital of Dali University, Dali, China
- Clinical Medical School, Dali University, Dali, China
| |
Collapse
|
5
|
Xu X, Hu H, Zeng H, Li B, Yin Q, Jiang Y, Zang L, Zhao C, Qian G. Sinisan ameliorates colonic injury induced by water immersion restraint stress by enhancing intestinal barrier function and the gut microbiota structure. PHARMACEUTICAL BIOLOGY 2023; 61:598-609. [PMID: 37013944 PMCID: PMC10075512 DOI: 10.1080/13880209.2023.2191643] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/21/2023] [Accepted: 03/12/2023] [Indexed: 06/19/2023]
Abstract
CONTEXT Sinisan (SNS) has been used to treat psychosomatic diseases of the digestive system. But little is known about how SNS affects water immersion restraint stress (WIRS). OBJECTIVE To study the effects of SNS on colonic tissue injury in the WIRS model. MATERIALS AND METHODS Forty-eight Kunming (KM) mice were randomized into 6 groups (n = 8): The control and WIRS groups receiving deionized water; the SNS low-dose (SL, 3.12 g/kg/d), SNS middle-dose (SM, 6.24 g/kg/d), SNS high-dose (SH, 12.48 g/kg/d), and diazepam (DZ, 5 mg/kg/d) groups; each with two daily administrations for 5 consecutive days. The 5 treatment groups were subjected to WIRS for 24 h on day 6. The effects of SNS on colon tissue injury caused by WIRS were assessed by changes in colon histology, inflammatory cytokines, brain-gut peptides, and tight junction (TJ) proteins levels. 16S rRNA gene sequencing was used to detect the regulation of the gut microbiota. RESULTS SNS pretreatment significantly reduced TNF-α (0.75- to 0.81-fold), IL-6 (0.77-fold), and IFN-γ (0.69-fold) levels; and increased TJ proteins levels, such as ZO-1 (4.06- to 5.27-fold), claudin-1 (3.33- to 5.14-fold), and occludin (6.46- to 11.82-fold). However, there was no significant difference between the levels of substance P (SP) and vasoactive intestinal peptide (VIP) in the control and WIRS groups. SNS regulated the composition of gut microbiota in WIRS mice. CONCLUSION The positive effects of SNS on WIRS could provide a theoretical basis to treat stress-related gastrointestinal disorders.
Collapse
Affiliation(s)
- Xiaoying Xu
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, P.R. China
| | - Huimei Hu
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, P.R. China
| | - Haizhou Zeng
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, P.R. China
| | - Boyi Li
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, P.R. China
| | - Qiuxiong Yin
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, P.R. China
| | - Yupeng Jiang
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, P.R. China
| | - Linquan Zang
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, P.R. China
| | - Changlin Zhao
- Health Science College, Guangdong Pharmaceutical University, Guangzhou, P.R. China
| | - Guoqiang Qian
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, P.R. China
| |
Collapse
|
6
|
Fongang B, Satizabal C, Kautz TF, Wadop YN, Muhammad JAS, Vasquez E, Mathews J, Gireud-Goss M, Saklad AR, Himali J, Beiser A, Cavazos JE, Mahaney MC, Maestre G, DeCarli C, Shipp EL, Vasan RS, Seshadri S. Cerebral small vessel disease burden is associated with decreased abundance of gut Barnesiella intestinihominis bacterium in the Framingham Heart Study. Sci Rep 2023; 13:13622. [PMID: 37604954 PMCID: PMC10442369 DOI: 10.1038/s41598-023-40872-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 08/17/2023] [Indexed: 08/23/2023] Open
Abstract
A bidirectional communication exists between the brain and the gut, in which the gut microbiota influences cognitive function and vice-versa. Gut dysbiosis has been linked to several diseases, including Alzheimer's disease and related dementias (ADRD). However, the relationship between gut dysbiosis and markers of cerebral small vessel disease (cSVD), a major contributor to ADRD, is unknown. In this cross-sectional study, we examined the connection between the gut microbiome, cognitive, and neuroimaging markers of cSVD in the Framingham Heart Study (FHS). Markers of cSVD included white matter hyperintensities (WMH), peak width of skeletonized mean diffusivity (PSMD), and executive function (EF), estimated as the difference between the trail-making tests B and A. We included 972 FHS participants with MRI scans, neurocognitive measures, and stool samples and quantified the gut microbiota composition using 16S rRNA sequencing. We used multivariable association and differential abundance analyses adjusting for age, sex, BMI, and education level to estimate the association between gut microbiota and WMH, PSMD, and EF measures. Our results suggest an increased abundance of Pseudobutyrivibrio and Ruminococcus genera was associated with lower WMH and PSMD (p values < 0.001), as well as better executive function (p values < 0.01). In addition, in both differential and multivariable analyses, we found that the gram-negative bacterium Barnesiella intestinihominis was strongly associated with markers indicating a higher cSVD burden. Finally, functional analyses using PICRUSt implicated various KEGG pathways, including microbial quorum sensing, AMP/GMP-activated protein kinase, phenylpyruvate, and β-hydroxybutyrate production previously associated with cognitive performance and dementia. Our study provides important insights into the association between the gut microbiome and cSVD, but further studies are needed to replicate the findings.
Collapse
Affiliation(s)
- Bernard Fongang
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| | - Claudia Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Tiffany F Kautz
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yannick N Wadop
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jazmyn A S Muhammad
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Erin Vasquez
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Julia Mathews
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Monica Gireud-Goss
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Amy R Saklad
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jayandra Himali
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Alexa Beiser
- Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Jose E Cavazos
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Neurology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Michael C Mahaney
- Department of Human Genetics, South Texas Diabetes and Obesity Institute, The University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Gladys Maestre
- Department of Neurosciences and Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Charles DeCarli
- Department of Neurology, Alzheimer's Disease Center, University of California, Davis, Sacramento, CA, USA
| | - Eric L Shipp
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ramachandran S Vasan
- Framingham Heart Study, Framingham, MA, USA
- Department of Medicine, Section of Cardiovascular Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
- Department of Medicine, Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Boston University's Center for Computing and Data Sciences, Boston, MA, USA
- The University of Texas School of Public Health in San Antonio, San Antonio, TX, USA
- The Long School of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Department of Neurology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
7
|
MUHAMMAD M, MUCHIMAPURA S, WATTANATHORN J. Microbiota-gut-brain axis impairment in the pathogenesis of stroke: implication as a potent therapeutic target. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2023; 42:143-151. [PMID: 37404572 PMCID: PMC10315190 DOI: 10.12938/bmfh.2022-067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 03/09/2023] [Indexed: 07/06/2023]
Abstract
The human microbiota-gut-brain axis has an enormous role in the maintenance of homeostasis and health. Over the last two decades, it has received concerted research attention and focus due to a rapidly emerging volume of evidence that has established that impairment within the microbiota-gut-brain axis contributes to the development and progression of various diseases. Stroke is one of the entities identified to be associated with microbiota-gut-brain axis impairment. Currently, there are still limitations in the clinical treatment of stroke, and the presence of a non-nervous factor from gut microbiota that can alter the course of stroke presents a novel strategy towards the search for a therapeutic silver bullet against stroke. Hence, the aim herein, was to focus on the involvement of microbiota-gut-brain axis impairment in the pathogenesis stroke as well as elucidate its implications as a potent therapeutic target against stroke. The findings of studies to date have revealed and extended the role microbiota-gut-brain axis impairment in the pathogenesis of stroke, and studies have identified from both clinical and pre-clinical perspectives targets within the microbiota-gut-brain axis and successfully modulated the outcome of stroke. It was concluded that the microbiota-gut-brain axis stands as potent target to salvage the neurons in the ischemic penumbra for the treatment of stroke. Assessment of the microbiota profile and its metabolites status holds enormous clinical potentials as a non-invasive indicator for the early diagnosis and prognosis of stroke.
Collapse
Affiliation(s)
- Mubarak MUHAMMAD
- Graduate School (Neuroscience Program), Faculty of Medicine,
Khon Kaen University, 123 Moo 16 Mittraphap Rd., Nai-Muang, Muang District, Khon Kaen
40002, Thailand
| | - Supaporn MUCHIMAPURA
- Department of Physiology, Faculty of Medicine, Khon Kaen
University, 123 Moo 16 Mittraphap Rd., Nai-Muang, Muang District, Khon Kaen 40002,
Thailand
- Integrative Complementary Alternative Medicine Research and
Development Center in the Research Institute for Human High Performance and Health
Promotion, Khon Kaen University, 123 Moo 16 Mittraphap Rd., Nai-Muang, Muang District,
Khon Kaen 40002, Thailand
| | - Jintanaporn WATTANATHORN
- Department of Physiology, Faculty of Medicine, Khon Kaen
University, 123 Moo 16 Mittraphap Rd., Nai-Muang, Muang District, Khon Kaen 40002,
Thailand
- Integrative Complementary Alternative Medicine Research and
Development Center in the Research Institute for Human High Performance and Health
Promotion, Khon Kaen University, 123 Moo 16 Mittraphap Rd., Nai-Muang, Muang District,
Khon Kaen 40002, Thailand
| |
Collapse
|
8
|
Mady EA, Doghish AS, El-Dakroury WA, Elkhawaga SY, Ismail A, El-Mahdy HA, Elsakka EGE, El-Husseiny HM. Impact of the mother's gut microbiota on infant microbiome and brain development. Neurosci Biobehav Rev 2023; 150:105195. [PMID: 37100161 DOI: 10.1016/j.neubiorev.2023.105195] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 04/28/2023]
Abstract
The link between the gut microbiome and health has recently garnered considerable interest in its employment for medicinal purposes. Since the early microbiota exhibits more flexibility compared to that of adults, there is a considerable possibility that altering it will have significant consequences on human development. Like genetics, the human microbiota can be passed from mother to child. This provides information on early microbiota acquisition, future development, and prospective chances for intervention. The succession and acquisition of early-life microbiota, modifications of the maternal microbiota during pregnancy, delivery, and infancy, and new efforts to understand maternal-infant microbiota transmission are discussed in this article. We also examine the shaping of mother-to-infant microbial transmission, and we then explore possible paths for future research to advance our knowledge in this area.
Collapse
Affiliation(s)
- Eman A Mady
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan; Department of Animal Hygiene, Behavior and Management, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya,13736, Egypt.
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and industrial pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Samy Y Elkhawaga
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Hussein M El-Husseiny
- Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan.
| |
Collapse
|
9
|
Sun Z, Wang X, Feng S, Xie C, Xing Y, Guo L, Zhao J, Ji C. A review of neuroendocrine immune system abnormalities in IBS based on the brain–gut axis and research progress of acupuncture intervention. Front Neurosci 2023; 17:934341. [PMID: 36968497 PMCID: PMC10034060 DOI: 10.3389/fnins.2023.934341] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 02/07/2023] [Indexed: 03/11/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a common digestive disorder observed in clinics. Current studies suggest that the pathogenesis of the disease is closely related to abnormal brain–gut interactions, hypokinesia, visceral sensory hypersensitivity in the gastrointestinal tract, and alterations in the intestinal microenvironment. However, it is difficult for a single factor to explain the heterogeneity of symptoms. The Rome IV criteria emphasized the holistic biologic-psycho-social model of IBS, suggesting that symptoms of the disease are closely related to neurogastroenterology and various abnormalities in brain–gut interaction. This study comprehensively reviewed the relationship between the brain–gut axis and IBS, the structure of the brain–gut axis, and the relationship between the brain–gut axis and intestinal microenvironment, and discussed the relationship between the abnormal regulation of the nervous system, endocrine system, and immune system and the incidence of IBS on the basis of brain–gut axis. In terms of treatment, acupuncture therapy can regulate the neuroendocrine-immune system of the body and improve the intestinal microenvironment, and it has the advantages of safety, economy, and effectiveness. We study the pathogenesis of IBS from local to global and micro to macro, and review the use of acupuncture to treat the disease as a whole so as to provide new ideas for the treatment of the disease.
Collapse
Affiliation(s)
- Zhangyin Sun
- College of Acupuncture and Moxibustion, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
- Department of Acupuncture and Moxibustion, Shaanxi Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Xuejiao Wang
- College of Acupuncture and Moxibustion, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
- Department of Acupuncture and Moxibustion, Shaanxi Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Shangsheng Feng
- MOE Key Laboratory of Biomedical Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Chaoju Xie
- College of Acupuncture and Moxibustion, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
- Department of Acupuncture and Moxibustion, Shaanxi Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Yu Xing
- College of Acupuncture and Moxibustion, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
- Department of Acupuncture and Moxibustion, Shaanxi Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Liang Guo
- College of Acupuncture and Moxibustion, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
- Department of Acupuncture and Moxibustion, Shaanxi Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Jingyu Zhao
- Department of Acupuncture and Moxibustion, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, China
- *Correspondence: Jingyu Zhao
| | - Changchun Ji
- Department of Acupuncture and Moxibustion, Shaanxi Hospital of Traditional Chinese Medicine, Xi'an, China
- Department of Acupuncture and Moxibustion, Shaanxi Provincial Institute of Traditional Chinese Medicine, Xi'an, China
- Changchun Ji
| |
Collapse
|
10
|
Zhang X, Wang X, Zhao H, Cao R, Dang Y, Yu B. Imbalance of Microbacterial Diversity Is Associated with Functional Prognosis of Stroke. Neural Plast 2023; 2023:6297653. [PMID: 37197229 PMCID: PMC10185427 DOI: 10.1155/2023/6297653] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/25/2022] [Accepted: 04/11/2023] [Indexed: 05/19/2023] Open
Abstract
Objectives There is mounting evidence to suggest that the pathophysiology of stroke is greatly influenced by the microbiota of the gut and its metabolites, in particular short-chain fatty acids (SCFAs). The primary purpose of the study was to evaluate whether the levels of SCFAs and the gut microbiota are altered in poststroke patients and to examine the relationship between these alterations and the physical condition, intestinal health, pain, or nutritional status of patients. Methods Twenty stroke patients and twenty healthy controls were enrolled in the current study, and their demographics were matched. Gas chromatography was used to determine the fecal SCFAs, and 16S rRNA gene sequencing was used to evaluate their fecal microbiota. Microbial diversity and richness were examined using the diversity indices alpha and beta, and taxonomic analysis was utilized to determine group differences. The relationships between the gut microbiome and fecal SCFAs, discriminant bacteria, and poststroke clinical outcomes were analyzed. Results Less community richness (ACE and Chao) was observed in the poststroke patients (P < 0.05), but the differences between the poststroke group and the healthy control group in terms of species diversity (Shannon and Simpson) were not statistically significant. The makeup of the poststroke gut microbiota was distinct from that of the control group, as evidenced by beta diversity. Then, the relative abundances of the taxa in the poststroke and control groups were compared in order to identify the specific microbiota changes. At the level of phylum, the poststroke subjects showed a significant increase in the relative abundances of Akkermansiaceae, Fusobacteriota, Desulfobacterota, Ruminococcaceae, and Oscillospirales and a particularly noticeable decrease in the relative abundance of Acidobacteriota compared to the control subjects (P < 0.05). In regard to SCFA concentrations, lower levels of fecal acetic acid (P = 0.001) and propionic acid (P = 0.049) were found in poststroke subjects. Agathobacter was highly correlated with acetic acid level (r = 0.473, P = 0.002), whereas Fusobacteria (r = -0.371, P = 0.018), Flavonifractor (r = -0.334, P = 0.034), Desulfovibrio (r = -0.362, P = 0.018), and Akkermansia (r = -0.321, P = 0.043) were negatively related to acetic acid levels. Additionally, the findings of the correlation analysis revealed that Akkermansia (r = -0.356, P = 0.024), Desulfovibrio (r = -0.316, P = 0.047), and Alloprevotella (r = -0.366, P = 0.020) were significantly negatively correlated with high-density lipoprotein cholesterol. In addition, the Neurogenic Bowel Dysfunction score (r = 0.495, P = 0.026), Barthel index (r = -0.531, P = 0.015), Fugl-Meyer Assessment score (r = -0.565, P = 0.009), Visual Analogue Scale score (r = 0.605, P = 0.005), and Brief Pain Inventory score (r = 0.507, P = 0.023) were significantly associated with alterations of distinctive gut microbiota. Conclusions Stroke generates extensive and substantial alterations in the gut microbiota and SCFAs, according to our findings. The differences of intestinal flora and lower fecal SCFA levels are closely related to the physical function, intestinal function, pain, or nutritional status of poststroke patients. Treatment strategies aimed at modulating the gut microbiota and SCFAs may have the potential to enhance the clinical results of patients.
Collapse
Affiliation(s)
- Xintong Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, China
| | - Xiangyu Wang
- Department of Rehabilitation Medicine, The Affiliated Lianyungang Oriental Hospital of Kangda College of Nanjing Medical University, Jiangsu, China
| | - Hong Zhao
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, China
| | - Risheng Cao
- Department of Science and Technology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, China
| | - Yini Dang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, China
| | - Binbin Yu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, China
| |
Collapse
|
11
|
Awuti K, Wang X, Sha L, Leng X. Exploring the regulatory mechanism of osteoporosis based on intestinal flora: A review. Medicine (Baltimore) 2022; 101:e32499. [PMID: 36596003 PMCID: PMC9803483 DOI: 10.1097/md.0000000000032499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Osteoporosis is 1 of the common diseases of bone metabolism in clinic. With the aging of the population in China, osteoporosis is becoming more and more serious, and it has become 1 of the major public health problems. However, traditional therapies, such as calcium therapy and estrogen therapy, can cause serious adverse effects and damage to the body when ingested over a long period of time. Therefore, there is an urgent need to explore alternative therapies with less side effects in clinical practice. Intestinal flora is a hot topic of research in recent years. It has been studied in inflammatory bowel disease, diabetes, depression and so on. Recently, intestinal flora has received increasing attention in the pathways regulating bone metabolism. This paper contains a review of recent studies related to osteoporosis and gut flora in terms of its metabolites, immune, endocrine, and brain-gut axis pathways. The strong association between intestinal flora and bone metabolism suggests, to some extent, that intestinal flora can be a potential target for osteoporosis prevention and treatment, providing new ideas and therapies for the prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Kasimu Awuti
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Xukai Wang
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Liquan Sha
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
- * Liquan Sha, The Third Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun 130117, China ()
| | - Xiangyang Leng
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| |
Collapse
|
12
|
Simpson JB, Redinbo MR. Multi-omic analysis of host-microbial interactions central to the gut-brain axis. Mol Omics 2022; 18:896-907. [PMID: 36169030 DOI: 10.1039/d2mo00205a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The gut microbiota impact numerous aspects of human physiology, including the central nervous system (CNS). Emerging work is now focusing on the microbial factors underlying the bi-directional communication network linking host and microbial systems within the gastrointestinal tract to the CNS, the "gut-brain axis". Neurotransmitters are key coordinators of this network, and their dysregulation has been linked to numerous neurological disease states. As the bioavailability of neurotransmitters is modified by gut microbes, it is critical to unravel the influence of the microbiota on neurotransmitters in the context of the gut-brain axis. Here we review foundational studies that defined molecular relationships between the microbiota, neurotransmitters, and the gut-brain axis. We examine links between the gut microbiome, behavior, and neurological diseases, as well as microbial influences on neurotransmitter bioavailability and physiology. Finally, we review multi-omics technologies uniquely applicable to this area, including high-throughput genetics, modern metabolomics, structure-guided metagenomics, targeted proteomics, and chemogenetics. Interdisciplinary studies will continue to drive the discovery of molecular mechanisms linking the gut microbiota to clinical manifestations of neurobiology.
Collapse
Affiliation(s)
- Joshua B Simpson
- Department of Chemistry, University of North Carolina at Chapel Hill, USA
| | - Matthew R Redinbo
- Department of Chemistry, University of North Carolina at Chapel Hill, USA
- Department of Biochemistry & Biophysics, Department of Microbiology & Immunology, and the Integrated Program in Biological & Genome Sciences, University of North Carolina at Chapel Hill, USA.
| |
Collapse
|
13
|
Bilister Egilmez C, Koyu A, Azak Pazarlar B, Keselik E, Oz Oyar E, Kurus M. Effects of intracerebroventricular and intravenous administration of Kisspeptin-54 and Gonadotropin-releasing hormone agonist in rats with ovarian hyperstimulation. Neuropeptides 2022; 96:102298. [PMID: 36367482 DOI: 10.1016/j.npep.2022.102298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/15/2022] [Accepted: 10/16/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE We aim to determine the effect of local and systemic administration of kisspeptin-54 on ovarian hyperstimulation. METHODS Immature female rats were used. In order to generate the ovarian hyperstimulation model, 50 IU PMSG was administered for 4 consecutive days and a single dose of 25 IU hCG was administered to all groups except for the sham group. To synchronize the sham group, a single dose of 10 IU PMSG followed by 10 IU hCG (48 h later) was applied. Kisspeptin-54 and gonadotropin-releasing hormone (GnRH) agonists were administered 48 h after hCG injection. While intracerebroventricular injection is performed with stereotaxic surgery, Intravenous administration was from the tail vein. Ovarian weights were measured. FSH, LH, estrogen and progesterone hormones were detected in serum by ELISA. VEGFa, IL-1β, TNF-α, MCP-1 immunohistochemical staining was performed on the ovaries and hypothalamus and their optical densities were determined with Image J. Kiss1R mRNA expression was determined by qRT-PCR. RESULTS Ovarian weights increased significantly in the OHSS group and the systemic GnRH agonist group. The optical densities of VEGFa, IL-1β, TNF- α and MCP-1 immunoreactivity showed us that both local and systemic applied kisspeptin-54 attenuates the level of investigated inflammation parameters in the ovaries. Moreover, local administration of kisspeptin-54 has been shown to enhance the level of Kiss1R mRNA in both the ovaries and the hypothalamus. CONCLUSION(S) Local and systemic administration of Kisspeptin-54 as a post-treatment reduces inflammation parameters in the ovaries. These findings promote the potential use of kisspeptin-54 on OHSS.
Collapse
Affiliation(s)
- Cansu Bilister Egilmez
- Department of Physiology, Faculty of Medicine, Izmir Katip Celebi University, Izmir, Turkey.
| | - Ahmet Koyu
- Department of Physiology, Faculty of Medicine, Izmir Katip Celebi University, Izmir, Turkey
| | - Burcu Azak Pazarlar
- Department of Physiology, Faculty of Medicine, Izmir Katip Celebi University, Izmir, Turkey
| | - Erdi Keselik
- Department of Histology and Embryology, Faculty of Medicine, Izmir Katip Celebi University, Izmir, Turkey
| | - Eser Oz Oyar
- Department of Physiology, Faculty of Medicine, Izmir Katip Celebi University, Izmir, Turkey
| | - Meltem Kurus
- Department of Histology and Embryology, Faculty of Medicine, Izmir Katip Celebi University, Izmir, Turkey
| |
Collapse
|
14
|
Glynn H, Mikocka-Walus A, Knowles SR. Editorial: Hospitalization Experiences and Post-traumatic Stress in Inflammatory Bowel Disease: Opportunities for Change. Inflamm Bowel Dis 2022; 29:839-841. [PMID: 35894682 DOI: 10.1093/ibd/izac166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Indexed: 12/09/2022]
Affiliation(s)
- Heidi Glynn
- Department of Psychology, Swinburne University of Technology, Melbourne, VIC, Australia
| | | | - Simon R Knowles
- Department of Psychology, Swinburne University of Technology, Melbourne, VIC, Australia
| |
Collapse
|
15
|
Fernández-Serrano AB, Moya-Faz FJ, Giner Alegría CA, Fernández Rodríguez JC. Negative correlation between IL-1β, IL-12 and TNF-γ, and cortisol levels in patients with panic disorder. Brain Behav 2022; 12:e2624. [PMID: 35588458 PMCID: PMC9226804 DOI: 10.1002/brb3.2624] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 03/26/2022] [Accepted: 04/07/2022] [Indexed: 11/06/2022] Open
Abstract
INTRODUCTION Chronic exposure to stress is a major risk factor in anxiety disorders (ADs) and can be accompanied by an altered microbiome-gut-brain axis and a compromised immune system. In recent years, the study of inflammatory processes in AD has gained special attention. Continued stress causes the reactivity of the hypothalamic-pituitary-adrenal (HPA) axis, the alteration of the intestinal microbiota and the consequent release of pro-inflammatory cytokines, affecting the sensitivity to stress and the similar behavior of anxiety. METHOD The aim of the present study was to evaluate the interrelationships between measures of proinflammatory cytokines and cortisol in patients with panic disorder (PD). RESULTS The main results of the correlation analysis revealed that the levels of pro-inflammatory cytokines interleukin (IL)-1β, IL-12, and tumor necrosis factor gamma were negatively correlated with cortisol scores (area under the curve with respect to the ground). CONCLUSIONS These results suggest that the inflammatory response is associated with the reactivity of the HPA axis in patients with PD and may influence the maintenance of anxiety behavior.
Collapse
Affiliation(s)
| | - Francisco José Moya-Faz
- Chair of Psychogeriatrics, Department of Health Sciences, Universidad Católica de Murcia UCAM, Murcia, Spain
| | | | | |
Collapse
|
16
|
Nazarova L, Liu H, Xie H, Wang L, Ding H, An H, Huang D. Targeting gut-brain axis through Scalp-Abdominal electroacupuncture in Parkinson’s disease. Brain Res 2022; 1790:147956. [DOI: 10.1016/j.brainres.2022.147956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/03/2022] [Accepted: 05/25/2022] [Indexed: 11/02/2022]
|
17
|
Frostad S, Bentz M. Anorexia nervosa: Outpatient treatment and medical management. World J Psychiatry 2022; 12:558-579. [PMID: 35582333 PMCID: PMC9048449 DOI: 10.5498/wjp.v12.i4.558] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 08/20/2021] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
Anorexia nervosa (AN) is a disabling, costly and potentially deadly illness. Treatment failure and relapse are common after completing treatment, and a substantial proportion of patients develop severe and enduring AN. The time from AN debut to the treatment initiation is normally unreasonably long. Over the past 20 years there has been empirical support for the efficacy of several treatments for AN. Moreover, outpatient treatment with family-based therapy or individual psychotherapy is associated with good outcomes for a substantial proportion of patients. Early intervention improves outcomes and should be a priority for all patients. Outpatient treatment is usually the best format for early intervention, and it has been demonstrated that even patients with severe or extreme AN can be treated as outpatients if they are medically stable. Inpatient care is more disruptive, more costly, and usually has a longer waiting list than does outpatient care. The decision as to whether to proceed with outpatient treatment or to transfer the patient for inpatient therapy may be difficult. The core aim of this opinion review is to provide the knowledge base needed for performing safe outpatient treatment of AN. The scientific essentials for outpatient treatment are described, including how to assess and manage the medical risks of AN and how to decide when transition to inpatient care is indicated. The following aspects are discussed: early intervention, outpatient treatment of AN, including outpatient psychotherapy for severe and extreme AN, how to determine when outpatient treatment is safe, and when transfer to inpatient healthcare is indicated. Emerging treatments, ethical issues and outstanding research questions are also addressed.
Collapse
Affiliation(s)
- Stein Frostad
- Department of Mental Health Research, Division of Psychiatry, Haukeland University Hospital, Bergen 5021, Norway
| | - Mette Bentz
- Child and Adolescent Mental Health Centre, Capital Region of Denmark, University of Copenhagen, Copenhagen 2400, Denmark
| |
Collapse
|
18
|
Collier CA, Mendiondo C, Raghavan S. Tissue engineering of the gastrointestinal tract: the historic path to translation. J Biol Eng 2022; 16:9. [PMID: 35379299 PMCID: PMC8981633 DOI: 10.1186/s13036-022-00289-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/08/2022] [Indexed: 11/15/2022] Open
Abstract
The gastrointestinal (GI) tract is imperative for multiple functions including digestion, nutrient absorption, and timely waste disposal. The central feature of the gut is peristalsis, intestinal motility, which facilitates all of its functions. Disruptions in GI motility lead to sub-optimal GI function, resulting in a lower quality of life in many functional GI disorders. Over the last two decades, tissue engineering research directed towards the intestine has progressed rapidly due to advances in cell and stem-cell biology, integrative physiology, bioengineering and biomaterials. Newer biomedical tools (including optical tools, machine learning, and nuanced regenerative engineering approaches) have expanded our understanding of the complex cellular communication within the GI tract that lead to its orchestrated physiological function. Bioengineering therefore can be utilized towards several translational aspects: (i) regenerative medicine to remedy/restore GI physiological function; (ii) in vitro model building to mimic the complex physiology for drug and pharmacology testing; (iii) tool development to continue to unravel multi-cell communication networks to integrate cell and organ-level physiology. Despite the significant strides made historically in GI tissue engineering, fundamental challenges remain including the quest for identifying autologous human cell sources, enhanced scaffolding biomaterials to increase biocompatibility while matching viscoelastic properties of the underlying tissue, and overall biomanufacturing. This review provides historic perspectives for how bioengineering has advanced over time, highlights newer advances in bioengineering strategies, and provides a realistic perspective on the path to translation.
Collapse
Affiliation(s)
- Claudia A Collier
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, 3120 TAMU, College Station, TX, 77843, USA
| | - Christian Mendiondo
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, 3120 TAMU, College Station, TX, 77843, USA
| | - Shreya Raghavan
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, 3120 TAMU, College Station, TX, 77843, USA.
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
19
|
De R, Dutta S. Role of the Microbiome in the Pathogenesis of COVID-19. Front Cell Infect Microbiol 2022; 12:736397. [PMID: 35433495 PMCID: PMC9009446 DOI: 10.3389/fcimb.2022.736397] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 01/28/2022] [Indexed: 12/12/2022] Open
Abstract
The ongoing pandemic coronavirus disease COVID-19 is caused by the highly contagious single-stranded RNA virus, SARS-coronavirus 2 (SARS-CoV-2), which has a high rate of evolution like other RNA viruses. The first genome sequences of SARS-CoV-2 were available in early 2020. Subsequent whole-genome sequencing revealed that the virus had accumulated several mutations in genes associated with viral replication and pathogenesis. These variants showed enhanced transmissibility and infectivity. Soon after the first outbreak due to the wild-type strain in December 2019, a genetic variant D614G emerged in late January to early February 2020 and became the dominant genotype worldwide. Thereafter, several variants emerged, which were found to harbor mutations in essential viral genes encoding proteins that could act as drug and vaccine targets. Numerous vaccines have been successfully developed to assuage the burden of COVID-19. These have different rates of efficacy, including, although rarely, a number of vaccinated individuals exhibiting side effects like thrombosis. However, the recent emergence of the Britain strain with 70% more transmissibility and South African variants with higher resistance to vaccines at a time when several countries have approved these for mass immunization has raised tremendous concern regarding the long-lasting impact of currently available prophylaxis. Apart from studies addressing the pathophysiology, pathogenesis, and therapeutic targets of SARS-CoV-2, analysis of the gut, oral, nasopharyngeal, and lung microbiome dysbiosis has also been undertaken to find a link between the microbiome and the pathogenesis of COVID-19. Therefore, in the current scenario of skepticism regarding vaccine efficacy and challenges over the direct effects of currently available drugs looming large, investigation of alternative therapeutic avenues based on the microbiome can be a rewarding finding. This review presents the currently available understanding of microbiome dysbiosis and its association with cause and consequence of COVID-19. Taking cues from other inflammatory diseases, we propose a hypothesis of how the microbiome may be influencing homeostasis, pro-inflammatory condition, and the onset of inflammation. This accentuates the importance of a healthy microbiome as a protective element to prevent the onset of COVID-19. Finally, the review attempts to identify areas where the application of microbiome research can help in reducing the burden of the disease.
Collapse
Affiliation(s)
- Rituparna De
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases, Kolkota, India
- Division of Immunology, National Institute of Cholera and Enteric Diseases, Kolkota, India
| | - Shanta Dutta
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases, Kolkota, India
| |
Collapse
|
20
|
Hu Q, Wu C, Yu J, Luo J, Peng X. Angelica sinensis polysaccharide improves rheumatoid arthritis by modifying the expression of intestinal Cldn5, Slit3 and Rgs18 through gut microbiota. Int J Biol Macromol 2022; 209:153-161. [PMID: 35318077 DOI: 10.1016/j.ijbiomac.2022.03.090] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/18/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease with a high incidence. Recent studies have demonstrated that diet can contribute to the development and progression of RA. Indeed, non-starch polysaccharides (NSPs) were known to be related to the improvement of RA. In this study, the collagen-induced rats were administrated with Angelica sinensis polysaccharide (ASP) at 200 mg/kg (L), 400 mg/kg (M), or 800 mg/kg (H). Results showed that ASP could reduce joint swelling and significantly inhibit anti-CII-antibodies and pro-inflammatory factors in RA, H group showed the best treatment among them. Further analysis using 16S rDNA sequencing suggested that ASP could shape the gut microbiota composition. Several key bacteria, including norank_f__norank_o__Clostridia_UCG-014, Lactobacillus, norank_f__Oscillospiraceae, and norank_f__Desulfovibrionaceae, were found to be related to the development of RA. The colonic transcriptome showed that ASP could restore RA-induced intestinal dysfunction, such as tight junction disarrangement, by upregulating Cldn5. The balance between osteoblasts and osteoclasts might be modified by regulating the expression of Slit3 and Rgs18 to alleviate RA, which may be correlated with gut microbiota. Our results suggested that ASP improved RA by regulating gut microbiota and gene expression, revealing a positive relationship between dietary patterns and RA.
Collapse
Affiliation(s)
- Qing Hu
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, Guangdong, China
| | - Changyu Wu
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, Guangdong, China
| | - Juntong Yu
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, Guangdong, China
| | - Jianming Luo
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, Guangdong, China.
| | - Xichun Peng
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, Guangdong, China.
| |
Collapse
|
21
|
Knowles SR, Apputhurai P, Burgell RE, Keefer L. Development and Validation of the Gastrointestinal Unhelpful Thinking Scale (GUTs): A Brief Self-Report Measure for Clinical and Research Settings. Gastroenterol Nurs 2022; 45:E1-E12. [PMID: 35020630 DOI: 10.1097/sga.0000000000000644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 08/16/2021] [Indexed: 11/27/2022] Open
Abstract
This article describes the development and validation of the Gastrointestinal Unhelpful Thinking scale. The purpose of the research was to develop the Gastrointestinal Unhelpful Thinking scale to assess in tandem the primary cognitive-affective drivers of brain-gut dysregulation, gastrointestinal-specific visceral anxiety, and pain catastrophizing. The research involved 3 phases which included undergraduate and community samples. In the first phase, an exploratory factor analysis revealed a 15-item 2-factor (visceral sensitivity and pain catastrophizing) scale (N= 323), which then was confirmed in the second phase: N = 399, χ2(26) = 2.08, p = .001, Tucker-Lewis Index = 0.94, comparative fit index = 0.96, standardized root mean square residual = 0.05, and root mean square error of approximation = 0.07. Demonstrating convergent validity, Gastrointestinal Unhelpful Thinking scale total and subscales were strongly correlated with the modified Manitoba Index, Irritable Bowel Syndrome Symptom Severity Scale scores, Visceral Sensitivity Index, and the Pain Catastrophizing Scale. A third phase (N = 16) established test-retest reliability for the Gastrointestinal Unhelpful Thinking scale (total and subscales). The test-retest reliability correlation coefficient for the Gastrointestinal Unhelpful Thinking scale total score was .93 (p < .001) and for the subscales was .86 (p < .001) and .94 (p < .001), respectively. The Gastrointestinal Unhelpful Thinking scale is a brief psychometrically valid measure of visceral anxiety and pain catastrophizing that can be useful for both clinicians and researchers who wish to measure these thinking patterns and relate them to changes in gastrointestinal and psychological symptoms.
Collapse
Affiliation(s)
- Simon R Knowles
- Simon R. Knowles, PhD, is Associate Professor, Department of Psychological Sciences, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, Victoria, Australia; Faculty of Medicine, Dentistry, & Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia; Colorectal Medicine and Genetics, The Royal Melbourne Hospital, Melbourne, Victoria, Australia; and Department of Gastroenterology, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia
- Pragalathan Apputhurai, PhD, is Lecturer, Department of Health Sciences and Biostatistics, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, Victoria, Australia
- Rebecca E. Burgell, PhD, is Associate Professor, Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; and is Consultant Gastroenterologist, Department of Gastroenterology, Alfred Health, Melbourne, Victoria, Australia
- Laurie Keefer, PhD, is Professor, Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Pragalathan Apputhurai
- Simon R. Knowles, PhD, is Associate Professor, Department of Psychological Sciences, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, Victoria, Australia; Faculty of Medicine, Dentistry, & Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia; Colorectal Medicine and Genetics, The Royal Melbourne Hospital, Melbourne, Victoria, Australia; and Department of Gastroenterology, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia
- Pragalathan Apputhurai, PhD, is Lecturer, Department of Health Sciences and Biostatistics, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, Victoria, Australia
- Rebecca E. Burgell, PhD, is Associate Professor, Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; and is Consultant Gastroenterologist, Department of Gastroenterology, Alfred Health, Melbourne, Victoria, Australia
- Laurie Keefer, PhD, is Professor, Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Rebecca E Burgell
- Simon R. Knowles, PhD, is Associate Professor, Department of Psychological Sciences, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, Victoria, Australia; Faculty of Medicine, Dentistry, & Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia; Colorectal Medicine and Genetics, The Royal Melbourne Hospital, Melbourne, Victoria, Australia; and Department of Gastroenterology, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia
- Pragalathan Apputhurai, PhD, is Lecturer, Department of Health Sciences and Biostatistics, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, Victoria, Australia
- Rebecca E. Burgell, PhD, is Associate Professor, Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; and is Consultant Gastroenterologist, Department of Gastroenterology, Alfred Health, Melbourne, Victoria, Australia
- Laurie Keefer, PhD, is Professor, Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Laurie Keefer
- Simon R. Knowles, PhD, is Associate Professor, Department of Psychological Sciences, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, Victoria, Australia; Faculty of Medicine, Dentistry, & Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia; Colorectal Medicine and Genetics, The Royal Melbourne Hospital, Melbourne, Victoria, Australia; and Department of Gastroenterology, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia
- Pragalathan Apputhurai, PhD, is Lecturer, Department of Health Sciences and Biostatistics, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, Victoria, Australia
- Rebecca E. Burgell, PhD, is Associate Professor, Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; and is Consultant Gastroenterologist, Department of Gastroenterology, Alfred Health, Melbourne, Victoria, Australia
- Laurie Keefer, PhD, is Professor, Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, New York
| |
Collapse
|
22
|
van Tilburg MAL, Drossman DA, Knowles SR. Psychogastroenterology: The brain-gut axis and its psychological applications. J Psychosom Res 2021; 152:110684. [PMID: 34847487 DOI: 10.1016/j.jpsychores.2021.110684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 11/20/2021] [Indexed: 12/19/2022]
Affiliation(s)
- Miranda A L van Tilburg
- Department of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, NC, United States; Department of Internal Medicine, Joan C Edwards School of Medicine, Marshall University, Huntington, WV, United States; School of Social Work, University of Washington, Seattle, WA, United States
| | - Douglas A Drossman
- Professor Emeritus of Medicine and Psychiatry, University of North Carolina, Chapel Hill, NC, United States; Center for Education and Practice for Biopsychosocial Care and Drossman Gastroenterology, Chapel Hill, NC, United States; President Emeritus and COO, Rome Foundation, Raleigh, NC, United States
| | - Simon R Knowles
- Department of Psychological Sciences, Swinburne University of Technology, Melbourne, Australia; Department of Gastroenterology and Hepatology, Royal Melbourne Hospital, Melbourne, Australia.
| |
Collapse
|
23
|
|
24
|
Person H, Keefer L. Psychological comorbidity in gastrointestinal diseases: Update on the brain-gut-microbiome axis. Prog Neuropsychopharmacol Biol Psychiatry 2021; 107:110209. [PMID: 33326819 PMCID: PMC8382262 DOI: 10.1016/j.pnpbp.2020.110209] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/20/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022]
Abstract
The high comorbidity of psychological disorders in both functional and organic gastrointestinal diseases suggests the intimate and complex link between the brain and the gut. Termed the brain-gut axis, this bidirectional communication between the central nervous system and enteric nervous system relies on immune, endocrine, neural, and metabolic pathways. There is increasing evidence that the gut microbiome is a key part of this system, and dysregulation of the brain-gut-microbiome axis (BGMA) has been implicated in disorders of brain-gut interaction, including irritable bowel syndrome, and in neuropsychiatric disorders, including depression, Alzheimer's disease, and autism spectrum disorder. Further, alterations in the gut microbiome have been implicated in the pathogenesis of organic gastrointestinal diseases, including inflammatory bowel disease. The BGMA is an attractive therapeutic target, as using prebiotics, probiotics, or postbiotics to modify the gut microbiome or mimic gut microbial signals could provide novel treatment options to address these debilitating diseases. However, despite significant advancements in our understanding of the BGMA, clinical data is lacking. In this article, we will review current understanding of the comorbidity of gastrointestinal diseases and psychological disorders. We will also review the current evidence supporting the key role of the BGMA in this pathology. Finally, we will discuss the clinical implications of the BGMA in the evaluation and management of psychological and gastrointestinal disorders.
Collapse
Affiliation(s)
- Hannibal Person
- Division of Pediatric Gastroenterology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Laurie Keefer
- Division of Pediatric Gastroenterology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
25
|
Banerjee A, Pradhan LK, Sahoo PK, Jena KK, Chauhan NR, Chauhan S, Das SK. Unravelling the potential of gut microbiota in sustaining brain health and their current prospective towards development of neurotherapeutics. Arch Microbiol 2021; 203:2895-2910. [PMID: 33763767 DOI: 10.1007/s00203-021-02276-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/18/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022]
Abstract
Increasing incidences of neurological disorders, such as Parkinson's disease (PD), multiple sclerosis (MS), Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS) are being reported, but an insight into their pathology remains elusive. Findings have suggested that gut microbiota play a major role in regulating brain functions through the gut-brain axis. A unique bidirectional communication between gut microbiota and maintenance of brain health could play a pivotal role in regulating incidences of neurodegenerative diseases. Contrarily, the present life style with changing food habits and disturbed circadian rhythm may contribute to gut homeostatic imbalance and dysbiosis leading to progression of several neurological disorders. Therefore, dysbiosis, as a primary factor behind intestinal disorders, may also augment inflammation, intestinal and blood-brain barrier permeability through microbiota-gut-brain axis. This review primarily focuses on the gut-brain axis functions, specific gut microbial population, metabolites produced by gut microbiota, their role in regulating various metabolic processes and role of gut microbiota towards development of neurodegenerative diseases. However, several studies have reported a decrease in abundance of a specific gut microbial population and a corresponding increase in other microbial family, with few findings revealing some contradictions. Reports also showed that colonization of gut microbiota isolated from patients suffering from neurodegenerative disease leads to the development of enhance pathological outcomes in animal models. Hence, a systematic understanding of the dominant role of specific gut microbiome towards development of different neurodegenerative diseases could possibly provide novel insight into the use of probiotics and microbial transplantation as a substitute approach for treating/preventing such health maladies.
Collapse
Affiliation(s)
- Ankita Banerjee
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, 751003, Odisha, India
| | - Lilesh Kumar Pradhan
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, 751003, Odisha, India
| | - Pradyumna Kumar Sahoo
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, 751003, Odisha, India
| | - Kautilya Kumar Jena
- Autophagy Laboratory, Infectious Disease Biology Division, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
| | - Nishant Ranjan Chauhan
- Autophagy Laboratory, Infectious Disease Biology Division, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
| | - Santosh Chauhan
- Autophagy Laboratory, Infectious Disease Biology Division, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
| | - Saroj Kumar Das
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, 751003, Odisha, India.
| |
Collapse
|
26
|
Livovsky DM, Azpiroz F. Gastrointestinal Contributions to the Postprandial Experience. Nutrients 2021; 13:nu13030893. [PMID: 33801924 PMCID: PMC7998507 DOI: 10.3390/nu13030893] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
Food ingestion induces homeostatic sensations (satiety, fullness) with a hedonic dimension (satisfaction, changes in mood) that characterize the postprandial experience. Both types of sensation are secondary to intraluminal stimuli produced by the food itself, as well as to the activity of the digestive tract. Postprandial sensations also depend on the nutrient composition of the meal and on colonic fermentation of non-absorbed residues. Gastrointestinal function and the sensitivity of the digestive tract, i.e., perception of gut stimuli, are determined by inherent individual factors, e.g., sex, and can be modulated by different conditioning mechanisms. This narrative review examines the factors that determine perception of digestive stimuli and the postprandial experience.
Collapse
Affiliation(s)
- Dan M. Livovsky
- Digestive System Research Unit, University Hospital Vall d’Hebron, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Departament de Medicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain;
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9103102, Israel
- ShaareZedek Medical Center, Digestive Diseases Institute, Jerusalem 9103102, Israel
| | - Fernando Azpiroz
- Digestive System Research Unit, University Hospital Vall d’Hebron, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Departament de Medicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain;
- Correspondence: ; Tel.: +34-93-2746259
| |
Collapse
|
27
|
Han QQ, Fu Y, Le JM, Pilot A, Cheng S, Chen PQ, Wu H, Wan GQ, Gu XF. Electroacupuncture may alleviate behavioral defects via modulation of gut microbiota in a mouse model of Parkinson's disease. Acupunct Med 2021; 39:501-511. [PMID: 33557583 DOI: 10.1177/0964528421990658] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Parkinson's disease (PD) is a chronic neurodegenerative disease involving non-motor symptoms, of which gastrointestinal disorders are the most common. In light of recent results, intestinal dysfunction may be involved in the pathogenesis of PD. Electroacupuncture (EA) has shown potential effects, although the underlying mechanism remains mostly unknown. We speculated that EA could relieve the behavioral defects of PD, and that this effect would be associated with modulation of the gut microbiota. METHODS Mice were randomly divided into three groups: control, PD + MA (manual acupuncture), and PD + EA. MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) was used to establish the mouse model of PD. Rotarod performance tests, open field tests, and pole tests were carried out to assess motor deficiencies. Immunohistochemistry was conducted to examine the survival of dopaminergic neurons. 16S ribosomal RNA (rRNA) gene sequencing was applied to investigate the alterations of the gut microbiome. Quantitative real-time polymerase chain reaction (PCR) was performed to characterize the messenger RNA (mRNA) levels of pro-inflammatory and anti-inflammatory cytokines. RESULTS We found that EA was able to alleviate the behavioral defects in the rotarod performance test and pole test, and partially rescue the significant loss of dopaminergic neurons in the substantia nigra (SN) chemically induced by MPTP in mice. Moreover, the PD + MA mice showed a tendency toward decreased intestinal microbial alpha diversity, while EA significantly reversed it. The abundance of Erysipelotrichaceae was significantly increased in PD + MA mice, and the alteration was also reversed by EA. In addition, the pro-inflammatory cytokines interleukin (IL)-6 and tumor necrosis factor (TNF)-α were substantially increased in the SN of PD + MA mice, an effect that was reversed by EA. CONCLUSION These results suggest that EA may alleviate behavioral defects via modulation of gut microbiota and suppression of inflammation in the SN of mice with PD, which provides new insights into the pathogenesis of PD and its treatment.
Collapse
Affiliation(s)
- Qiu-Qin Han
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yi Fu
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Jia-Mei Le
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Adam Pilot
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
| | - Si Cheng
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Pei-Qing Chen
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Hailong Wu
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Guo-Qing Wan
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Xue-Feng Gu
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
28
|
Barbuti RC, Schiavon LL, Oliveira CP, Alvares-DA-Silva MR, Sassaki LY, Passos MDCF, Farias AQ, Barros LL, Barreto BP, Albuquerque GBDMLD, Alves AM, Navarro-Rodriguez T, Bittencourt PL. GUT MICROBIOTA, PREBIOTICS, PROBIOTICS, AND SYNBIOTICS IN GASTROINTESTINAL AND LIVER DISEASES: PROCEEDINGS OF A JOINT MEETING OF THE BRAZILIAN SOCIETY OF HEPATOLOGY (SBH), BRAZILIAN NUCLEUS FOR THE STUDY OF HELICOBACTER PYLORI AND MICROBIOTA (NBEHPM), AND BRAZILIAN FEDERATION OF GASTROENTEROLOGY (FBG). ARQUIVOS DE GASTROENTEROLOGIA 2021; 57:381-398. [PMID: 33331485 DOI: 10.1590/s0004-2803.202000000-72] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/06/2020] [Indexed: 02/07/2023]
Abstract
Over the last years, there is growing evidence that microorganisms are involved in the maintenance of our health and are related to various diseases, both intestinal and extraintestinal. Changes in the gut microbiota appears to be a key element in the pathogenesis of hepatic and gastrointestinal disorders, including non-alcoholic fatty liver disease, alcoholic liver disease, liver cirrhosis, inflammatory bowel disease, irritable bowel syndrome, and Clostridium difficile - associated diarrhea. In 2019, the Brazilian Society of Hepatology (SBH) in cooperation with the Brazilian Nucleus for the Study of Helicobacter Pylori and Microbiota (NBEHPM), and Brazilian Federation of Gastroenterology (FBG) sponsored a joint meeting on gut microbiota and the use of prebiotics, probiotics, and synbiotics in gastrointestinal and liver diseases. This paper summarizes the proceedings of the aforementioned meeting. It is intended to provide practical information about this topic, addressing the latest discoveries and indicating areas for future studies.
Collapse
Affiliation(s)
- Ricardo Correa Barbuti
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Leonardo Lucca Schiavon
- Universidade Federal de Santa Catarina, Faculdade de Medicina, Departamento de Clínica Médica, Florianópolis, SC, Brasil
| | - Cláudia P Oliveira
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Mário Reis Alvares-DA-Silva
- Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Medicina Interna, Porto Alegre, RS, Brasil
| | | | | | - Alberto Queiroz Farias
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Luisa Leite Barros
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Bruno Paes Barreto
- Universidade do Estado do Pará, Centro de Ciências Biológicas e da Saúde, Belém, PA, Brasil.,Centro Universitário do Estado do Pará (CESUPA), Belém, PA, Brasil
| | | | - Amanda Mandarino Alves
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Tomás Navarro-Rodriguez
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | | |
Collapse
|
29
|
Metabolomics analysis of microbiota-gut-brain axis in neurodegenerative and psychiatric diseases. J Pharm Biomed Anal 2020; 194:113681. [PMID: 33279302 DOI: 10.1016/j.jpba.2020.113681] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/30/2020] [Accepted: 10/06/2020] [Indexed: 12/19/2022]
Abstract
Gut microbiota represents a complex physiological ecosystem that influences the host health. Alterations in the microbiome metabolism affect the body homeostasis and they have been associated with the development of different human neurodegenerative and neuropsychiatric disorders, such as Alzheimer's disease, autism spectrum disorder, bipolar disorder, depression, Huntington's disease, Parkinson's disease, posttraumatic stress disorder and schizophrenia. The development of these complex diseases is influenced by various factors, including genetic predisposition and environmental triggers. Gut microbiota has recently emerged as an important actor in their physiopathology that has been shown to play a role in inflammation, oxidative stress, and gut permeability. Therefore, targeting the metabolites that are produced by or associated with the gut microbiota may help us understand how imbalance in the gut-brain axis affects human health. This review offers a comprehensive overview of the literature on this matter, offering the readers an insight in the state-of-art metabolic measurements of the gut-brain axis in various brain-related diseases.
Collapse
|
30
|
Pinheiro da Silva F, Vinicius Macarini Bruzaferro E, Olsen Saraiva Câmara N. Antimicrobial peptides in the gut-brain axis: A straightforward review to unravel some missing links. J Neurosci Res 2020; 98:2384-2389. [PMID: 32945561 DOI: 10.1002/jnr.24729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/26/2020] [Accepted: 08/30/2020] [Indexed: 11/11/2022]
Abstract
Antimicrobial peptides (AMPs) are intriguing molecules, able to directly kill several microorganisms and to regulate multiple aspects of the immune response. Despite the extensive studies on the role of AMPs in the epithelial barrier, placing them as a pivotal line of defense against pathogen invasion, little attention has been directed to their role in the maintenance and modulation of the gut microbiota and, by consequence, of the homeostasis of extra intestinal tissues. Here, we review the recent literature about the microbiome-gut-brain axis, focusing on the role of AMPs in this scenario. We provide a straightforward revision of current data in order to provide an overview of the subject, discussing more in depth some points that, in our opinion, are crucial and have received little attention.
Collapse
Affiliation(s)
- Fabiano Pinheiro da Silva
- Laboratório de Emergências Clínicas, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | | | - Niels Olsen Saraiva Câmara
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Instituto de Ciências Biológicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
31
|
Heenan P, Creemers RH, Sharma S, Keenan J, Bayer S, Young W, Cooney J, Armstrong K, Fraser K, Skidmore PM, Talley NJ, Roy N, Gearry RB. Cohort Profile: The Christchurch IBS cOhort to investigate Mechanisms FOr gut Relief and improved Transit (COMFORT). Inflamm Intest Dis 2020; 5:132-143. [PMID: 32999886 PMCID: PMC7506285 DOI: 10.1159/000508160] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 04/23/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND AIMS This cross-sectional observational case-control study was initiated in July 2016 with the aim of increasing an understanding of the underlying disease mechanisms in functional gastrointestinal disorders (FGIDs) including irritable bowel syndrome (IBS), functional diarrhoea (FD), and functional constipation (FC). Specific areas of interest include the effect of food, microbiome, host and microbial genetics, metabolome, and psychological variables on unexplained chronic gastrointestinal (GI) symptoms. METHODS This study recruited consecutive patients who were attending one of two endoscopy centres in Christchurch, New Zealand, for colonoscopy and a subgroup of participants from the general public who did not undergo colonoscopy. Participants with known GI disease other than an FGID were excluded. Those with symptoms were recruited as cases, whilst those without symptoms were recruited as controls. In the days prior to preparation for colonoscopy, or an agreeable time for those not undergoing colonoscopy, demographic, symptom, psychological, dietary, and health data were collected in addition to biological samples (breath, faeces, blood, and urine). Colonic biopsies were taken at the time of colonoscopy from participants in the colonoscopy subgroup. RESULTS Between July 2016 and December 2018, 349 participants were recruited, 315 of whom completed the study, 220 participants were from the colonoscopy subgroup, and 95 from the non-colonoscopy subgroup. This included 129 controls and 186 cases (57 IBS-diarrhoea predominant, 30 IBS-constipation predominant, 41 IBS-mixed, 42 FC, and 16 FD). The mean age of FGID cases was 53.4 years and controls 54.4 years. Cases (149/186, 80.1%) and controls (57/72, 55.8%) were predominantly female. Education levels were similar across the cohort. Smoking and alcohol rates were also similar. Biological samples were collected as planned from participants. CONCLUSIONS The COMFORT cohort is a unique clinical cohort of FGID cases and controls with a wide range of demographic, dietary, clinical, psychological, and health data in addition to biological samples. Future research will aim to use a systems biology approach to establish the potential role of diet, host-microbiome interactions, and other factors in the pathogenesis of FGIDs.
Collapse
Affiliation(s)
- Phoebe Heenan
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Rob H. Creemers
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Shriya Sharma
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Jacqueline Keenan
- Department of Surgery, University of Otago, Christchurch, New Zealand
| | - Simone Bayer
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Wayne Young
- Food Nutrition and Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand
- Riddet Institute, Massey University, Palmerston North, New Zealand
- High Value Nutrition Science Challenge, University of Newcastle, Newcastle, New South Wales, Australia
| | - Janine Cooney
- High Value Nutrition Science Challenge, University of Newcastle, Newcastle, New South Wales, Australia
- Massey University, Biological Chemistry & Bioactives Group and Food Innovation, Palmerston North, New Zealand
| | - Kelly Armstrong
- Food Nutrition and Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand
| | - Karl Fraser
- Food Nutrition and Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand
- Riddet Institute, Massey University, Palmerston North, New Zealand
- High Value Nutrition Science Challenge, University of Newcastle, Newcastle, New South Wales, Australia
| | - Paula M. Skidmore
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Nicholas J. Talley
- Plant & Food Research, Research and Innovation Division, Hamilton, New Zealand
| | - Nicole Roy
- Riddet Institute, Massey University, Palmerston North, New Zealand
- High Value Nutrition Science Challenge, University of Newcastle, Newcastle, New South Wales, Australia
- Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| | - Richard B. Gearry
- Department of Medicine, University of Otago, Christchurch, New Zealand
- High Value Nutrition Science Challenge, University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
32
|
Isaiah S, Loots DT, Solomons R, van der Kuip M, Tutu Van Furth AM, Mason S. Overview of Brain-to-Gut Axis Exposed to Chronic CNS Bacterial Infection(s) and a Predictive Urinary Metabolic Profile of a Brain Infected by Mycobacterium tuberculosis. Front Neurosci 2020; 14:296. [PMID: 32372900 PMCID: PMC7186443 DOI: 10.3389/fnins.2020.00296] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/16/2020] [Indexed: 12/12/2022] Open
Abstract
A new paradigm in neuroscience has recently emerged - the brain-gut axis (BGA). The contemporary focus in this paradigm has been gut → brain ("bottom-up"), in which the gut-microbiome, and its perturbations, affects one's psychological state-of-mind and behavior, and is pivotal in neurodegenerative disorders. The emerging brain → gut ("top-down") concept, the subject of this review, proposes that dysfunctional brain health can alter the gut-microbiome. Feedback of this alternative bidirectional highway subsequently aggravates the neurological pathology. This paradigm shift, however, focuses upon non-communicable neurological diseases (progressive neuroinflammation). What of infectious diseases, in which pathogenic bacteria penetrate the blood-brain barrier and interact with the brain, and what is this effect on the BGA in bacterial infection(s) that cause chronic neuroinflammation? Persistent immune activity in the CNS due to chronic neuroinflammation can lead to irreversible neurodegeneration and neuronal death. The properties of cerebrospinal fluid (CSF), such as immunological markers, are used to diagnose brain disorders. But what of metabolic markers for such purposes? If a BGA exists, then chronic CNS bacterial infection(s) should theoretically be reflected in the urine. The premise here is that chronic CNS bacterial infection(s) will affect the gut-microbiome and that perturbed metabolism in both the CNS and gut will release metabolites into the blood that are filtered (kidneys) and excreted in the urine. Here we assess the literature on the effects of chronic neuroinflammatory diseases on the gut-microbiome caused by bacterial infection(s) of the CNS, in the context of information attained via metabolomics-based studies of urine. Furthermore, we take a severe chronic neuroinflammatory infectious disease - tuberculous meningitis (TBM), caused by Mycobacterium tuberculosis, and examine three previously validated CSF immunological biomarkers - vascular endothelial growth factor, interferon-gamma and myeloperoxidase - in terms of the expected changes in normal brain metabolism. We then model the downstream metabolic effects expected, predicting pivotal altered metabolic pathways that would be reflected in the urinary profiles of TBM subjects. Our cascading metabolic model should be adjustable to account for other types of CNS bacterial infection(s) associated with chronic neuroinflammation, typically prevalent, and difficult to distinguish from TBM, in the resource-constrained settings of poor communities.
Collapse
Affiliation(s)
- Simon Isaiah
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Du Toit Loots
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Regan Solomons
- Department of Pediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Martijn van der Kuip
- Pediatric Infectious Diseases and Immunology, Amsterdam University Medical Center, Academic Medical Center, Emma Children’s Hospital, Amsterdam, Netherlands
| | - A. Marceline Tutu Van Furth
- Pediatric Infectious Diseases and Immunology, Amsterdam University Medical Center, Academic Medical Center, Emma Children’s Hospital, Amsterdam, Netherlands
| | - Shayne Mason
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
33
|
Liu L, Yuan X, Yang L, Zhang J, Luo J, Huang G, Huo J. Effect of acupuncture on hormone level in patients with gastrointestinal dysfunction after general anesthesia: A study protocol for a randomized controlled trial. Medicine (Baltimore) 2020; 99:e19610. [PMID: 32243385 PMCID: PMC7440186 DOI: 10.1097/md.0000000000019610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Postoperative gastrointestinal dysfunction (PGD) refers to one of the common postoperative complications. Acupuncture can facilitate the recovery of PGD, whereas no therapeutic schedule of acupuncture has been internationally recognized for treating PGD. In the present study, a scientific trial protocol has been proposed to verify the feasibility of acupuncture in treating gastrointestinal dysfunction after laparoscopic cholecystectomy under general anesthesia. We conduct this protocol to investigate whether acupuncture recovery gastrointestinal dysfunction by influencing the expression of gastrointestinal hormone. METHOD The present study refers to a randomized, evaluator blinded, controlled, multi-center clinical trial; it was designed complying with the Consolidated Standards of Reporting Trials (CONSORT 2010) as well as the Standard for Reporting Interventions in Controlled Trials of Acupuncture (STRICTA). The subjects will be taken from the inpatients having undergone laparoscopic surgery of Mianyang Affiliated Hospital of Chengdu University of traditional Chinese medicine, Mianyang Third Hospital and Mianyang Anzhou Hospital. Based on the random number yielded using SPSS 25.0 software, the qualified subjects will be randomly classified to the experimental group and the control group. Therapies will be performed 30 min once after operation, the experimental group will be treated with acupuncture, while the control group will receive intravenous injection of granisetron. The major outcome will be the time to first flatus, and the secondary outcomes will include the time to first defecation, abdominal pain, dosage of analgesia pump, abdominal distention, nausea, vomiting, gastrointestinal hormone, as well as mental state. The efficacy and safety of acupuncture will be also assessed following the principle of Good Clinical Practice (GCP). DISCUSS A standardized and scientific clinical trial is conducted to assess the efficacy and safety of acupuncture for gastrointestinal dysfunction after laparoscopic cholecystectomy under general anesthesia. The aim is to objectively evidence and improves the clinical practice of acupoint prescription, as an attempt to promote the clinical application of this technology.
Collapse
Affiliation(s)
- Lisha Liu
- Mianyang Affiliated Hospital, Chengdu University of Traditional Chinese Medicine, Mianyang
| | - Xiuli Yuan
- Mianyang Affiliated Hospital, Chengdu University of Traditional Chinese Medicine, Mianyang
| | - Lei Yang
- Mianyang Affiliated Hospital, Chengdu University of Traditional Chinese Medicine, Mianyang
| | - Jingyuan Zhang
- ChengDu University of Traditional Chinese Medicine, Chengdu
| | - Jing Luo
- Mianyang Third Hospital, Mianyang
| | | | - Jian Huo
- ChengDu University of Traditional Chinese Medicine, Chengdu
| |
Collapse
|
34
|
Weaver KR, Melkus GD, Fletcher J, Henderson WA. Relevance of Sex and Subtype in Patients With IBS: An Exploratory Study of Gene Expression. Biol Res Nurs 2020; 22:13-23. [PMID: 31833409 PMCID: PMC7068753 DOI: 10.1177/1099800419889189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Psychological state, stress level, and gastrointestinal function are intricately related and relevant to symptom exacerbation in patients with irritable bowel syndrome (IBS), but genetic contributors to this brain-gut connection are not fully understood. The purpose of this exploratory study was to compare gene expression in participants with IBS to that of healthy controls (HC) and to examine patterns of expression in participants with IBS by sex and IBS subtype. METHOD Participants were recruited to an ongoing protocol at the National Institutes of Health. Differences in demographic and clinical characteristics were assessed using descriptive statistics and Mann-Whitney U tests. Expression levels of 84 genes were evaluated in peripheral whole blood using Custom RT2 Profiler polymerase chain reaction (PCR) Arrays, and data analysis was performed through GeneGlobe Data Analysis Center. RESULTS Participants with IBS (n = 27) reported greater levels of perceived stress (p = .037) and differed in expression values of ±2 for the genes ADIPOR1, ADIPOR2, CNR2, COMT, OXTR, and PPARA compared to HC (n = 43). Further analyses by sex and IBS subtype revealed differential patterns of gene expression related to the endocannabinoid system, cytokines, stress, and sex steroid hormones. CONCLUSIONS Diverse yet interconnected processes such as metabolism, inflammation, immunity, social behavior, and pain are associated with differences in gene expression between participants with IBS and HC. These findings lend support for genomic associations with the brain-gut connection in patients with IBS and highlight the relevance of sex and IBS subtype in performing such analyses.
Collapse
Affiliation(s)
- Kristen R. Weaver
- Department of Pain and Translational Symptom Science, University of Maryland
School of Nursing, Baltimore, MD, USA
- National Institute of Nursing Research, National Institutes of Health,
Bethesda, MD, USA
| | | | - Jason Fletcher
- Rory Meyers College of Nursing, New York University, NY, USA
| | - Wendy A. Henderson
- Digestive Disorders Unit, Division of Intramural Research, National
Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
35
|
Xiang L, Lou Y, Liu L, Liu Y, Zhang W, Deng J, Guan Y, She M, You X, Liu M, Li H, Xu X, Liu F, Cai X. Gut Microbiotic Features Aiding the Diagnosis of Acute Ischemic Stroke. Front Cell Infect Microbiol 2020; 10:587284. [PMID: 33409158 PMCID: PMC7779606 DOI: 10.3389/fcimb.2020.587284] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 11/18/2020] [Indexed: 12/14/2022] Open
Abstract
Increasing evidence suggests that features of the gut microbiota correlate with ischemic stroke. However, the specific characteristics of the gut microbiota in patients suffering different types of ischemic stroke, or recovering from such strokes, have rarely been studied, and potential microbiotic predictors of different types of stroke have seldom been analyzed. We subjected fecal specimens from patients with lacunar or non-lacunar acute ischemic infarctions, and those recovering from such strokes, to bacterial 16S rRNA sequencing and compared the results to those of healthy volunteers. We identified microbial markers of different types of ischemic stroke and verified that these were of diagnostic utility. Patients with two types of ischemic stroke, and those recovering from ischemic stroke, exhibited significant shifts in microbiotic diversities compared to healthy subjects. Cluster of Orthologous Groups of Proteins (COG) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses revealed reduced metabolic and transport-related pathway activities in ischemic stroke patients. We performed fivefold cross-validation using a Random Forest model to identify two optimal bacterial species (operational taxonomic units; OTUs) serving as markers of lacunar infarction; these were Lachnospiraceae (OTU_45) and Bacteroides (OTU_4), and the areas under the receiver operating characteristic curves (AUCs under the ROCs) were 0.881 and 0.872 respectively. In terms of non-lacunar acute ischemic infarction detection, the two optimal species were Bilophila (OTU_330) and Lachnospiraceae (OTU_338); the AUCs under the ROCs were 0.985 and 0.929 respectively. In post-ischemic stroke patients, the three optimal species were Pseudomonas (OTU_35), Sphingomonadaceae (OTU_303), and Akkermansia (OTU_9); the AUCs under the ROCs were 1, 0.897, and 0.846 respectively. Notably, the gut microbial markers were of considerable value for utility when diagnosing lacunar infarction, non-lacunar acute ischemic infarction, and post-ischemic stroke. This study is the first to characterize the gut microbiotic profiles of patients with lacunar or non-lacunar, acute ischemic strokes, and those recovering from stroke, and to identify microbiotic predictors of such strokes.
Collapse
Affiliation(s)
- Lei Xiang
- Department of Integrative Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanfeng Lou
- Department of Dermatology, Jinling Hospital, Southern Medical University, Nanjing, China
| | - Lingyu Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yuanling Liu
- Administrative Department, Guangdong Province Hospital for Women and Children Healthcare, Guangzhou, China
| | - Weizheng Zhang
- Clinical Laboratory, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianxin Deng
- Department of Endocrinology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
| | - Yubin Guan
- Clinical Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Miaoqin She
- Research Section, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Xinchao You
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Minqi Liu
- Clinical Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Hongwei Li
- Institute of Biotherapy, Southern Medical University, Guangzhou, China
| | - Xiaosong Xu
- Clinical Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- *Correspondence: Xiangsheng Cai, ; Fang Liu, ; Xiaosong Xu,
| | - Fang Liu
- Department of Dermatology, Jinling Hospital, Southern Medical University, Nanjing, China
- *Correspondence: Xiangsheng Cai, ; Fang Liu, ; Xiaosong Xu,
| | - Xiangsheng Cai
- Clinical Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Center for Medical Experiments, University of Chinese Academy of Science-Shenzhen Hospital, Shenzhen, China
- *Correspondence: Xiangsheng Cai, ; Fang Liu, ; Xiaosong Xu,
| |
Collapse
|
36
|
Hou Y, Yan Q, An H, Wang J, Tian M, Zhao W, Wu A, Feng Y. The use and protective effects of transcutaneous electrical acupoint stimulation during abdominal surgery: study protocol for a multicenter randomized parallel controlled trial. Trials 2019; 20:462. [PMID: 31358034 PMCID: PMC6664584 DOI: 10.1186/s13063-019-3558-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/05/2019] [Indexed: 12/31/2022] Open
Abstract
Background Acupuncture-balanced anesthesia has been found to offer protective benefits. Electrical stimulation at certain acupoints can potentially promote perioperative gastrointestinal function recovery. The purpose of this study is to explore the effects of acupuncture-balanced anesthesia on the postoperative recovery of gastrointestinal function, on anesthesia strategies for abdominal surgery, on postoperative pain treatment, and on any associated complications or alterations in immune function. We further seek to verify the protective effects of transcutaneous electrical acupoint stimulation (TEAS), to explore possible underlying neuroimmune–endocrine mechanisms, and to thereby develop an optimized acupuncture-balanced anesthesia strategy suitable for abdominal surgery. Together, these findings will provide a scientific basis for the clinical utilization of acupuncture-balanced anesthesia in the context of abdominal surgery. Methods/design This study is a multicenter, large-sample, randomized placebo-controlled trial. All subjects will be patients undergoing elective gastric or colorectal surgery. In Part 1, these patients will be stratified according to surgical site (gastric or colorectal), and randomly divided into four groups based on different perioperative interventions: Con group, which will undergo sham TEAS before, during, and after surgery; T1 group, which will receive TEAS during the preoperative and intraoperative periods, and sham TEAS during the postoperative period; T2 group, which will receive TEAS during the preoperative period, sham TEAS during the intraoperative period, and TEAS during the postoperative period; and T3 group, which will receive TEAS before, during, and after operation. Part 2 of this study will focus solely on colorectal surgery patients. All patients will receive TEAS during the preoperative and intraoperative periods, and they will be randomized into four groups according to different postoperative treatments: Con′ group, which will not receive TEAS; T1′ group, which will receive sham TEAS; T2′ group, which will receive 5-Hz TEAS; and T3′ group, which will receive 100-Hz TEAS. Venous blood (5 ml) will be used to measure immunological and inflammatory indexes both at the preoperative stage prior to TEAS and 4–5 days after operation. The primary outcome will be the time to first bowel sounds after surgery. Secondary outcomes will include gastrointestinal functional recovery, analgesic efficacy during the postoperative period, acupuncture-balanced anesthesia efficacy, postoperative nausea and vomiting, and postoperative complications. Discussion This study is designed to investigate the clinical value of TEAS during various perioperative periods in those undergoing abdominal surgery, with the overall goal of evaluating the clinical value and advantages of acupuncture-balanced anesthesia, and of providing new strategies for improving patient prognoses. Trial registration Chinese Clinical Trial Registry, ChiCTR-TRC-14004435. Registered on 26 March 2014. Electronic supplementary material The online version of this article (10.1186/s13063-019-3558-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuantao Hou
- Peking University People's Hospital, Beijing, China
| | - Qi Yan
- Peking University People's Hospital, Beijing, China
| | - Haiyan An
- Peking University People's Hospital, Beijing, China
| | - Junlu Wang
- First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Province Wencheng County People's Hospital, Wenzhou, China
| | - Ming Tian
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wensheng Zhao
- Zhejiang Integrated Traditional and Western Medicine Hospital, Hangzhou, China
| | - Anshi Wu
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yi Feng
- Peking University People's Hospital, Beijing, China.
| |
Collapse
|
37
|
Manousiouthakis E, Chen Y, Cairns DM, Pollard R, Gerlovin K, Dente MJ, Razavi Y, Kaplan DL. Bioengineered in vitro enteric nervous system. J Tissue Eng Regen Med 2019; 13:1712-1723. [PMID: 31278844 DOI: 10.1002/term.2926] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 06/06/2019] [Accepted: 06/19/2019] [Indexed: 12/13/2022]
Abstract
Bidirectional interactions between the human central nervous system and the gastrointestinal tract, via the enteric nervous system, are unmapped and central to many human conditions. There is a critical need to develop 3D human in vitro intestinal tissue models to emulate the intricate cell interactions of the human enteric nervous system within the gastrointestinal tract in order to better understand these complex interactions that cannot be studied utilizing in vivo animal models. In vitro systems, if sufficiently replicative of some in vivo conditions, may assist with the study of individual cell interactions. Here, we describe a 3D-innervated tissue model of the human intestine consisting of human-induced neural stem cells differentiated into relevant enteric nervous system neural cell types. Enterocyte-like (Caco-2) and goblet-like (HT29-MTX) cells are used to form the intestinal epithelial layer, and intestinal myofibroblasts are utilized to simulate the stromal layer. In vitro enteric nervous system cultures supported survival and function of the various cell types, with mucosal and neural transcription factors evident over 5 weeks. The human-induced neural stem cells migrated from the seeding location on the peripheral layer of the hollow scaffolds toward the luminal epithelial cells, prompted by the addition of neural growth factor. nNOS-expressing neurons and the substance P precursor gene TAC1 were expressed within the in vitro enteric nervous system to support the utility of the tissue model to recapitulate enteric nervous system phenotypes. This innervated tissue system offers a new tool to use to help in understanding neural circuits controlling the human intestine and associated communication networks.
Collapse
Affiliation(s)
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, Medford, MA
| | - Dana M Cairns
- Department of Biomedical Engineering, Tufts University, Medford, MA
| | - Rachel Pollard
- Department of Biomedical Engineering, Tufts University, Medford, MA
| | - Kaia Gerlovin
- Department of Biomedical Engineering, Tufts University, Medford, MA
| | - Michael J Dente
- Department of Biomedical Engineering, Tufts University, Medford, MA
| | - Yasmin Razavi
- Department of Biomedical Engineering, Tufts University, Medford, MA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA
| |
Collapse
|
38
|
Cong L, Duan LW, Su WP, Hao S, Li DF. Efficacy of High Specific Volume Polysaccharide - A New Type of Dietary Fiber - On Molecular Mechanism of Intestinal Water Metabolism in Rats With Constipation. Med Sci Monit 2019; 25:5028-5035. [PMID: 31280283 PMCID: PMC6636405 DOI: 10.12659/msm.916526] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background The aim of this study was to evaluate the effects of a new type of dietary fiber – high specific volume polysaccharide (HSVP) – on fecal properties, serum vasoactive intestinal peptide (VIP) concentration, intestinal flora count, and expression of the VIP-cAMP-PKA-AQP3 signaling pathway. Marerial/Methods Compound diphenoxylate was used in 48 healthy Wistar rats to establish a constipation model. Rats were divided into a normal control group, a constipation model group, an HSVP low-dose group, an HSVP medium-dose group, an HSVP high-dose group, and a fructose control group. We used colony count method, ELISA, WB, and RT-PCR to determine fecal moisture content, fecal hardness, fecal passage time, serum VIP concentration, number of intestinal bacteria, and VIP-cAMP-PKA-AQP3 signal pathway protein expression. Results The constipation model was established successfully. HSVP (the medium dose was 10% and the high dose was 15%) improved fecal moisture content, reduced hardness, shortened fecal emptying time, increased intestinal bacteria, reduced serum VIP concentration, downregulated cAMP and PKAm RNA transcription, reduced protein expression, and reduced intestinal AQP3 expression. Conclusions HSVP improved constipation, increased the number of intestinal bacteria, and elevated expression of the VIP-cAMP-PKA-AQP3 signaling pathway. The mechanism of HSVP in regulating intestinal water metabolism in constipated rats may occur through the VIP-cAMP-PKA-AQP3 signaling pathway, and be closely related to changes in intestinal bacteria. The important role of the brain-gut-microbiome axis in the pathogenesis of constipation has been confirmed in this study.
Collapse
Affiliation(s)
- Liang Cong
- Digestive System Department, The Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Li-Wei Duan
- Digestive System Department, The Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Wei-Ping Su
- Key Laboratory of Polymer Ecomaterials, Chinese Academy of Sciences, Changchun, Jilin, China (mainland)
| | - Shuhong Hao
- Department of Hematology and Oncology, The Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Dong-Fu Li
- Digestive System Department, The Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
39
|
Brain-Gut Therapies for Pediatric Functional Gastrointestinal Disorders and Inflammatory Bowel Disease. Curr Gastroenterol Rep 2019; 21:12. [PMID: 30868282 DOI: 10.1007/s11894-019-0683-8] [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] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to discuss current knowledge of brain-gut therapies (BGT) in pediatric functional gastrointestinal disorders (FGID) and inflammatory bowel disease (IBD), including their evidence base, the common psychopathology that they address, and the integration of this knowledge into medical settings. RECENT FINDINGS Cognitive behavioral therapy (CBT), hypnotherapy (HT), mindfulness-based therapy (MBT), and exposure-based therapy (EBT) have the most data supporting their use in children, particularly in FGID, more so than in IBD. This difference is most likely because of the increased role of psychological factors in FGID, though these same factors can be seen comorbidly in IBD. Integrative BGT treatment strategies with the collaboration of clinicians across disciplines may provide the most benefit to patients. This review details our current understanding of the evidence for BGT in pediatric FGID and IBD and how they may best be used in treatment strategies.
Collapse
|