1
|
Landis GN, Bell HS, Peng OK, Fan Y, Yan K, Baybutt B, Tower J. Conditional Inhibition of Eip75B Eliminates the Effects of Mating and Mifepristone on Lifespan in Female Drosophila. Cells 2024; 13:1123. [PMID: 38994975 PMCID: PMC11240670 DOI: 10.3390/cells13131123] [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: 05/11/2024] [Revised: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024] Open
Abstract
Mating in female Drosophila melanogaster causes midgut hypertrophy and reduced lifespan, and these effects are blocked by the drug mifepristone. Eip75B is a transcription factor previously reported to have pleiotropic effects on Drosophila lifespan. Because Eip75B null mutations are lethal, conditional systems and/or partial knock-down are needed to study Eip75B effects in adults. Previous studies showed that Eip75B is required for adult midgut cell proliferation in response to mating. To test the possible role of Eip75B in mediating the lifespan effects of mating and mifepristone, a tripartite FLP-recombinase-based conditional system was employed that provides controls for genetic background. Expression of a Hsp70-FLP transgene was induced in third instar larvae by a brief heat pulse. The FLP recombinase catalyzed the recombination and activation of an Actin5C-GAL4 transgene. The GAL4 transcription factor in turn activated expression of a UAS-Eip75B-RNAi transgene. Inhibition of Eip75B activity was confirmed by loss of midgut hypertrophy upon mating, and the lifespan effects of both mating and mifepristone were eliminated. In addition, the negative effects of mifepristone on egg production were eliminated. The data indicate that Eip75B mediates the effects of mating and mifepristone on female midgut hypertrophy, egg production, and lifespan.
Collapse
Affiliation(s)
| | | | | | | | | | | | - John Tower
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089-2910, USA
| |
Collapse
|
2
|
Ma Y, Zhang L, Wei R, Dai W, Zeng R, Luo D, Jiang R, Zhuo Z, Yang Q, Li J, Leung FW, Duan C, Sha W, Chen H. Risks of digestive diseases in long COVID: evidence from a population-based cohort study. BMC Med 2024; 22:14. [PMID: 38195495 PMCID: PMC10777515 DOI: 10.1186/s12916-023-03236-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/19/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND In the post-pandemic era, a wide range of COVID-19 sequelae is of growing health concern. However, the risks of digestive diseases in long COVID have not been comprehensively understood. To investigate the long-term risk of digestive diseases among COVID patients. METHODS In this large-scale retrospective cohort study with up to 2.6 years follow-up (median follow-up: 0.7 years), the COVID-19 group (n = 112,311), the contemporary comparison group (n = 359,671) and the historical comparison group (n = 370,979) predated the COVID-19 outbreak were built using UK Biobank database. Each digestive outcome was defined as the diagnosis 30 days or more after the onset of COVID-19 infection or the index date. Hazard ratios (HRs) and corresponding 95% confidence intervals (CI) were computed utilizing the Cox regression models after inverse probability weighting. RESULTS Compared with the contemporary comparison group, patients with previous COVID-19 infection had higher risks of digestive diseases, including gastrointestinal (GI) dysfunction (HR 1.38 (95% CI 1.26 to 1.51)); peptic ulcer disease (HR 1.23 (1.00 to 1.52)); gastroesophageal reflux disease (GERD) (HR 1.41 (1.30 to 1.53)); gallbladder disease (HR 1.21 (1.06 to 1.38)); severe liver disease (HR 1.35 (1.03 to 1.76)); non-alcoholic liver disease (HR 1.27 (1.09 to 1.47)); and pancreatic disease (HR 1.36 (1.11 to 1.66)). The risks of GERD were increased stepwise with the severity of the acute phase of COVID-19 infection. Even after 1-year follow-up, GERD (HR 1.64 (1.30 to 2.07)) and GI dysfunction (HR 1.35 (1.04 to 1.75)) continued to pose risks to COVID-19 patients. Compared to those with one SARS-CoV-2 infection, reinfected patients were at a higher risk of pancreatic diseases (HR 2.57 (1.23 to 5.38)). The results were consistent when the historical cohort was used as the comparison group. CONCLUSIONS Our study provides insights into the association between COVID-19 and the long-term risk of digestive system disorders. COVID-19 patients are at a higher risk of developing digestive diseases. The risks exhibited a stepwise escalation with the severity of COVID-19, were noted in cases of reinfection, and persisted even after 1-year follow-up. This highlights the need to understand the varying risks of digestive outcomes in COVID-19 patients over time, particularly those who experienced reinfection, and develop appropriate follow-up strategies.
Collapse
Affiliation(s)
- Yuying Ma
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Lijun Zhang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Rui Wei
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Weiyu Dai
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Ruijie Zeng
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Shantou University Medical College, Shantou, 515000, Guangdong, China
| | - Dongling Luo
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, 510080, China
| | - Rui Jiang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Zewei Zhuo
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Qi Yang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Jingwei Li
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Felix W Leung
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90024, USA.
- Sepulveda Ambulatory Care Center, Veterans Affairs Greater Los Angeles Healthcare System, North Hills, CA, 91343, USA.
| | - Chongyang Duan
- Department of Biostatistics, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| | - Weihong Sha
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
- School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Shantou University Medical College, Shantou, 515000, Guangdong, China.
| | - Hao Chen
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
- School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Shantou University Medical College, Shantou, 515000, Guangdong, China.
| |
Collapse
|
3
|
Giatti S, Diviccaro S, Cioffi L, Cosimo Melcangi R. Post-Finasteride Syndrome And Post-Ssri Sexual Dysfunction: Two Clinical Conditions Apparently Distant, But Very Close. Front Neuroendocrinol 2024; 72:101114. [PMID: 37993021 DOI: 10.1016/j.yfrne.2023.101114] [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: 09/28/2023] [Revised: 10/31/2023] [Accepted: 11/19/2023] [Indexed: 11/24/2023]
Abstract
Post-finasteride syndrome and post-SSRI sexual dysfunction, are two poorly explored clinical conditions in which men treated for androgenetic alopecia with finasteride or for depression with SSRI antidepressants show persistent side effects despite drug suspension (e.g., sexual dysfunction, psychological complaints, sleep disorders). Because of some similarities in the symptoms, common pathological mechanisms are proposed here. Indeed, as discussed, clinical studies and preclinical data obtained so far suggest an important role for brain modulators (i.e., neuroactive steroids), neurotransmitters (i.e., serotonin, and cathecolamines), and gut microbiota in the context of the gut-brain axis. In particular, the observed interconnections of these signals in these two clinical conditions may suggest similar etiopathogenetic mechanisms, such as the involvement of the enzyme converting norepinephrine into epinephrine (i.e., phenylethanolamine N-methyltransferase). However, despite the current efforts, more work is still needed to advance the understanding of these clinical conditions in terms of diagnostic markers and therapeutic strategies.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Lucia Cioffi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
4
|
Landis GN, Bell HS, Peng O, Bognar B, Tong A, Manea TD, Bao H, Han X, Tower J. Dhr96[1] mutation and maternal tudor[1] mutation increase life span and reduce the beneficial effects of mifepristone in mated female Drosophila. PLoS One 2023; 18:e0292820. [PMID: 38127988 PMCID: PMC10735022 DOI: 10.1371/journal.pone.0292820] [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] [Received: 08/08/2023] [Accepted: 09/28/2023] [Indexed: 12/23/2023] Open
Abstract
Mating and receipt of male Sex Peptide hormone cause increased egg laying, increased midgut size and decreased life span in female Drosophila. Feeding mated females with the synthetic steroid mifepristone decreases egg production, reduces midgut size, and increases life span. Here, several gene mutations were assayed to investigate possible mechanisms for mifepristone action. Drosophila Dhr96 is a hormone receptor, and a key positive regulator of midgut lipid uptake and metabolism. Dhr96[1] null mutation increased female life span, and reduced the effects of mifepristone on life span, suggesting that Dhr96[1] mutation and mifepristone may act in part through the same mechanism. Consistent with this idea, lipidomics analysis revealed that mating increases whole-body levels of triglycerides and fatty-acids in triglycerides, and these changes are reversed by mifepristone. Maternal tudor[1] mutation results in females that lack the germ-line and produce no eggs. Maternal tudor[1] mutation increased mated female life span, and reduced but did not eliminate the effects of mating and mifepristone on life span. This indicates that decreased egg production may be related to the life span benefits of mifepristone, but is not essential. Mifepristone increases life span in w[1118] mutant mated females, but did not increase life span in w[1118] mutant virgin females. Mifepristone decreased egg production in w[1118] mutant virgin females, indicating that decreased egg production is not sufficient for mifepristone to increase life span. Mifepristone increases life span in virgin females of some, but not all, white[+] and mini-white[+] strains. Backcrossing of mini-white[+] transgenes into the w[1118] background was not sufficient to confer a life span response to mifepristone in virgin females. Taken together, the data support the hypothesis that mechanisms for mifepristone life span increase involve reduced lipid uptake and/or metabolism, and suggest that mifepristone may increase life span in mated females and virgin females through partly different mechanisms.
Collapse
Affiliation(s)
- Gary N. Landis
- Department of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Hans S. Bell
- Department of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Oscar Peng
- Department of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Brett Bognar
- Department of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Andy Tong
- Department of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Tomás D. Manea
- Department of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Hanmei Bao
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - John Tower
- Department of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
5
|
Chen BN, Humenick A, Yew WP, Peterson RA, Wiklendt L, Dinning PG, Spencer NJ, Wattchow DA, Costa M, Brookes SJH. Types of Neurons in the Human Colonic Myenteric Plexus Identified by Multilayer Immunohistochemical Coding. Cell Mol Gastroenterol Hepatol 2023; 16:573-605. [PMID: 37355216 PMCID: PMC10469081 DOI: 10.1016/j.jcmgh.2023.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND AND AIMS Gut functions including motility, secretion, and blood flow are largely controlled by the enteric nervous system. Characterizing the different classes of enteric neurons in the human gut is an important step to understand how its circuitry is organized and how it is affected by disease. METHODS Using multiplexed immunohistochemistry, 12 discriminating antisera were applied to distinguish different classes of myenteric neurons in the human colon (2596 neurons, 12 patients) according to their chemical coding. All antisera were applied to every neuron, in multiple layers, separated by elutions. RESULTS A total of 164 combinations of immunohistochemical markers were present among the 2596 neurons, which could be divided into 20 classes, with statistical validation. Putative functions were ascribed for 4 classes of putative excitatory motor neurons (EMN1-4), 4 inhibitory motor neurons (IMN1-4), 3 ascending interneurons (AIN1-3), 6 descending interneurons (DIN1-6), 2 classes of multiaxonal sensory neurons (SN1-2), and a small, miscellaneous group (1.8% of total). Soma-dendritic morphology was analyzed, revealing 5 common shapes distributed differentially between the 20 classes. Distinctive baskets of axonal varicosities surrounded 45% of myenteric nerve cell bodies and were associated with close appositions, suggesting possible connectivity. Baskets of cholinergic terminals and several other types of baskets selectively targeted ascending interneurons and excitatory motor neurons but were significantly sparser around inhibitory motor neurons. CONCLUSIONS Using a simple immunohistochemical method, human myenteric neurons were shown to comprise multiple classes based on chemical coding and morphology and dense clusters of axonal varicosities were selectively associated with some classes.
Collapse
Affiliation(s)
- Bao Nan Chen
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Adam Humenick
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Wai Ping Yew
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Rochelle A Peterson
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Lukasz Wiklendt
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Phil G Dinning
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia; Colorectal Surgical Unit, Division of Surgery, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Nick J Spencer
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - David A Wattchow
- Colorectal Surgical Unit, Division of Surgery, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Marcello Costa
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Simon J H Brookes
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.
| |
Collapse
|
6
|
Sharkey KA, Mawe GM. The enteric nervous system. Physiol Rev 2023; 103:1487-1564. [PMID: 36521049 PMCID: PMC9970663 DOI: 10.1152/physrev.00018.2022] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Of all the organ systems in the body, the gastrointestinal tract is the most complicated in terms of the numbers of structures involved, each with different functions, and the numbers and types of signaling molecules utilized. The digestion of food and absorption of nutrients, electrolytes, and water occurs in a hostile luminal environment that contains a large and diverse microbiota. At the core of regulatory control of the digestive and defensive functions of the gastrointestinal tract is the enteric nervous system (ENS), a complex system of neurons and glia in the gut wall. In this review, we discuss 1) the intrinsic neural control of gut functions involved in digestion and 2) how the ENS interacts with the immune system, gut microbiota, and epithelium to maintain mucosal defense and barrier function. We highlight developments that have revolutionized our understanding of the physiology and pathophysiology of enteric neural control. These include a new understanding of the molecular architecture of the ENS, the organization and function of enteric motor circuits, and the roles of enteric glia. We explore the transduction of luminal stimuli by enteroendocrine cells, the regulation of intestinal barrier function by enteric neurons and glia, local immune control by the ENS, and the role of the gut microbiota in regulating the structure and function of the ENS. Multifunctional enteric neurons work together with enteric glial cells, macrophages, interstitial cells, and enteroendocrine cells integrating an array of signals to initiate outputs that are precisely regulated in space and time to control digestion and intestinal homeostasis.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gary M Mawe
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
7
|
Mulak A, Freud T, Waluga M, Bangdiwala SI, Palsson OS, Sperber AD. Sex- and gender-related differences in the prevalence and burden of disorders of gut-brain interaction in Poland. Neurogastroenterol Motil 2023; 35:e14568. [PMID: 36989186 DOI: 10.1111/nmo.14568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/02/2023] [Accepted: 03/12/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND The aim of the study was to assess sex- and gender-related differences in the epidemiology and impact of disorders of gut-brain interaction (DGBI) in Poland. METHODS Data used for the current analysis were derived from the Polish population sample of 2057 subjects (1030 F, 1027 M) collected via the Internet survey that included the Rome IV diagnostic questionnaire and 80 supplementary questions. KEY RESULTS The overall prevalence of DGBI in Poland was 46.0% (51.7% in women and 40.3% in men, p < 0.001). Comparing women versus men, esophageal disorders were observed in 6.3% vs. 6.0%, respectively (p > 0.05), gastroduodenal disorders in 14.0% vs. 7.8% (p < 0.001), bowel disorders in 44.3% vs. 33.9% (p < 0.001), and anorectal disorders in 9.9% vs. 7.7% (p > 0.05). The six most common DGBI included functional constipation 14.2%, functional dyspepsia 8.3%, proctalgia fugax 6.6%, functional bloating 4.8%, functional diarrhea 4.5%, and irritable bowel syndrome (IBS) 4.4%. All these disorders, except for functional diarrhea, were more common in women. The DGBI overlap was significantly higher in women than in men (16.7% vs. 11.2%, p < 0.001). A higher number of overlapping DGBI correlated positively with IBS severity, higher level of somatization, anxiety and depression, poorer quality of life (QoL), and increased healthcare utilization. CONCLUSIONS AND INFERENCES This is the first comprehensive report on significant sex/gender-related differences in the prevalence and burden of DGBI in Poland. The revealed differences between women and men with DGBI in the clinical profile, psychosocial variables, and healthcare utilization may have important diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Agata Mulak
- Department of Gastroenterology and Hepatology, Wroclaw Medical University, Wroclaw, Poland
| | - Tamar Freud
- Siaal Research Center for Family Medicine and Primary Care, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Marek Waluga
- Department of Gastroenterology and Hepatology, Medical University of Silesia, Katowice, Poland
| | - Shrikant I Bangdiwala
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Olafur S Palsson
- Center for Functional GI and Motility Disorders, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ami D Sperber
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
8
|
Holscher HD, Chumpitazi BP, Dahl WJ, Fahey GC, Liska DJ, Slavin JL, Verbeke K. Perspective: Assessing Tolerance to Nondigestible Carbohydrate Consumption. Adv Nutr 2022; 13:2084-2097. [PMID: 36041178 PMCID: PMC9776727 DOI: 10.1093/advances/nmac091] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/02/2022] [Accepted: 08/25/2022] [Indexed: 01/28/2023] Open
Abstract
Human intestinal enzymes do not hydrolyze nondigestible carbohydrates (NDCs), and thus, they are not digested and absorbed in the small intestine. Instead, NDCs are partially to completely fermented by the intestinal microbiota. Select NDCs are associated with health benefits such as laxation and lowering of blood cholesterol and glucose. NDCs provide functional attributes to processed foods, including sugar or fat replacers, thickening agents, and bulking agents. Additionally, NDCs are incorporated into processed foods to increase their fiber content. Although consumption of NDCs can benefit health and contribute functional characteristics to foods, they can cause gastrointestinal symptoms, such as flatulence and bloating. As gastrointestinal symptoms negatively affect consumer well-being and their acceptance of foods containing NDC ingredients, it is crucial to consider tolerance when designing food products and testing their physiological health benefits in clinical trials. This perspective provides recommendations for the approach to assess gastrointestinal tolerance to NDCs, with a focus on study design, population criteria, intervention, comparator, and outcome. Special issues related to studies in children and implications for stakeholders are also discussed. It is recommended that the evaluation of gastrointestinal tolerance to NDCs be conducted in randomized, blinded, controlled crossover studies using standard gastrointestinal questionnaires, with attention to study participant background diets, health status, lifestyle, and medications.
Collapse
Affiliation(s)
- Hannah D Holscher
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, USA
| | - Bruno P Chumpitazi
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Children's Nutrition Research Center, United States Department of Agriculture, Houston, TX, USA
| | - Wendy J Dahl
- Department of Food Science and Human Nutrition, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, USA
| | - George C Fahey
- Department of Animal Sciences, University of Illinois, Urbana, IL USA
| | | | - Joanne L Slavin
- Department of Food Science and Nutrition, University of Minnesota, Twin Cities, MN USA
| | - Kristin Verbeke
- Translational Research in Gastrointestinal Disorders, KU Leuven, Targid, Leuven, Belgium; and Leuven Food Science and Nutrition Research Centre, Leuven, Belgium
| |
Collapse
|
9
|
Gálvez-Robleño C, López-Tofiño Y, López-Gómez L, Bagüés A, Soto-Montenegro ML, Abalo R. Radiographic assessment of the impact of sex and the circadian rhythm-dependent behaviour on gastrointestinal transit in the rat. Lab Anim 2022:236772221124381. [DOI: 10.1177/00236772221124381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Relatively little is known about the influence of sex and the circadian rhythm on gastrointestinal transit. However, these factors could have an important impact on aspects such as digestion, oral absorption of drugs or the clinical manifestation of gastrointestinal diseases, among others. Remarkably, preclinical models have scarcely taken these factors into consideration. In this study, we assessed the gastrointestinal transit of young adult Wistar Han rats of both sexes, under normal and inverted light cycle. To do this, serial radiographs were taken for 24 h (T0–T24) after intragastric barium administration and subsequently analysed to construct transit curves for each gastrointestinal region. Under a normal light cycle, transit curves were similar, except for a slower transit in females compared with males from T8 to T24. Under the inverted cycle, there was a significant acceleration in stomach emptying (similar in both sexes), emptying of the small intestine (even faster in females) and filling of the caecum and colon (which was also even faster in females). This study confirms, using X-ray non-invasive methods for the first time, that both sex and circadian rhythm (probably through its effect on behaviour) influence gastrointestinal transit in laboratory animals.
Collapse
Affiliation(s)
- Carlos Gálvez-Robleño
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), Spain
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut), University Rey Juan Carlos (URJC), Spain
| | - Yolanda López-Tofiño
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), Spain
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut), University Rey Juan Carlos (URJC), Spain
| | - Laura López-Gómez
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), Spain
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut), University Rey Juan Carlos (URJC), Spain
| | - Ana Bagüés
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), Spain
- Unidad Asociada I+D+i al Instituto de Química Médica, IQM (CSIC), Spain
- High Performance Research Group in Experimental Pharmacology (PHARMAKOM), University Rey Juan Carlos (URJC), Spain
| | - María Luisa Soto-Montenegro
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut), University Rey Juan Carlos (URJC), Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Spain
- CIBER de Salud Mental (CIBERSAM), Spain
| | - Raquel Abalo
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), Spain
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut), University Rey Juan Carlos (URJC), Spain
- Unidad Asociada I+D+i al Instituto de Química Médica, IQM (CSIC), Spain
- Grupo de Trabajo de Ciencias Básicas en Dolor y Analgesia de la Sociedad Española del Dolor, Spain
- Grupo de Trabajo de Cannabinoides de la Sociedad Española del Dolor, Spain
| |
Collapse
|
10
|
Landis GN, Hilsabeck TAU, Bell HS, Ronnen-Oron T, Wang L, Doherty DV, Tejawinata FI, Erickson K, Vu W, Promislow DEL, Kapahi P, Tower J. Mifepristone Increases Life Span of Virgin Female Drosophila on Regular and High-fat Diet Without Reducing Food Intake. Front Genet 2021; 12:751647. [PMID: 34659367 PMCID: PMC8511958 DOI: 10.3389/fgene.2021.751647] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 09/13/2021] [Indexed: 12/14/2022] Open
Abstract
Background: The synthetic steroid mifepristone is reported to have anti-obesity and anti-diabetic effects in mammals on normal and high-fat diets (HFD). We previously reported that mifepristone blocks the negative effect on life span caused by mating in female Drosophila melanogaster. Methods: Here we asked if mifepristone could protect virgin females from the life span-shortening effect of HFD. Mifepristone was assayed for effects on life span in virgin females, in repeated assays, on regular media and on media supplemented with coconut oil (HFD). The excrement quantification (EX-Q) assay was used to measure food intake of the flies after 12 days mifepristone treatment. In addition, experiments were conducted to compare the effects of mifepristone in virgin and mated females, and to identify candidate mifepristone targets and mechanisms. Results: Mifepristone increased life span of virgin females on regular media, as well as on media supplemented with either 2.5 or 5% coconut oil. Food intake was not reduced in any assay, and was significantly increased by mifepristone in half of the assays. To ask if mifepristone might rescue virgin females from all life span-shortening stresses, the oxidative stressor paraquat was tested, and mifepristone produced little to no rescue. Analysis of extant metabolomics and transcriptomics data suggested similarities between effects of mifepristone in virgin and mated females, including reduced tryptophan breakdown and similarities to dietary restriction. Bioinformatics analysis identified candidate mifepristone targets, including transcription factors Paired and Extra-extra. In addition to shortening life span, mating also causes midgut hypertrophy and activation of the lipid metabolism regulatory factor SREBP. Mifepristone blocked the increase in midgut size caused by mating, but did not detectably affect midgut size in virgins. Finally, mating increased activity of a SREBP reporter in abdominal tissues, as expected, but reporter activity was not detectably reduced by mifepristone in either mated or virgin females. Conclusion: Mifepristone increases life span of virgin females on regular and HFD without reducing food intake. Metabolomics and transcriptomics analyses suggest some similar effects of mifepristone between virgin and mated females, however reduced midgut size was observed only in mated females. The results are discussed regarding possible mifepristone mechanisms and targets.
Collapse
Affiliation(s)
- Gary N. Landis
- Molecular and Computational Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
| | - Tyler A. U. Hilsabeck
- Buck Institute for Research on Aging, Novato, CA, United States
- Davis School of Gerontology, University of Southern California, University Park, Los Angeles, CA, United States
| | - Hans S. Bell
- Molecular and Computational Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
| | - Tal Ronnen-Oron
- Buck Institute for Research on Aging, Novato, CA, United States
| | - Lu Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| | - Devon V. Doherty
- Molecular and Computational Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
| | - Felicia I. Tejawinata
- Molecular and Computational Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
| | - Katherine Erickson
- Molecular and Computational Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
| | - William Vu
- Molecular and Computational Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
| | - Daniel E. L. Promislow
- Department of Biology, University of Washington, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, United States
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, Novato, CA, United States
| | - John Tower
- Molecular and Computational Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
11
|
Shrestha A, Samuelsson LM, Sharma P, Day L, Cameron-Smith D, Milan AM. Comparing Response of Sheep and Cow Milk on Acute Digestive Comfort and Lactose Malabsorption: A Randomized Controlled Trial in Female Dairy Avoiders. Front Nutr 2021; 8:603816. [PMID: 33659266 PMCID: PMC7917135 DOI: 10.3389/fnut.2021.603816] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/22/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Sheep milk (SM) is a possible alternate dairy source for those who experience digestive symptoms with cow milk (CM). While both the milks contain lactose, one of the causes for self-reported intolerance to CM, the composition of SM and CM also differs across proteins and fats, which have been shown to impact digestive processes. Objective: To compare the acute digestive comfort and lactose malabsorption of SM to CM in female dairy avoiders. Method: In a double-blinded, randomized cross over trial, 30 dairy-avoiding females (aged 20-30 years) drank 650 mL of SM or CM (each reconstituted from spray dried powder) following an overnight fast, on two separate occasions at least 1 week apart. Blood samples were collected for glucose and insulin assessment, and single nucleotide polymorphisms of the lactase (LCT) gene (C/T13910 and G/A22018). Breath H2 and visual analog scale (VAS) digestive symptom scores were recorded at fasting and regular intervals over 4 h after ingestion. Results: Eighty percentage of study participants were lactase non-persistent (LNP; CC13910 and GG22018 genotype). Digestive symptoms, including abdominal cramps, distension, rumbling, bloating, belching, diarrhea, flatulence, vomiting, and nausea, were similar in response to SM and CM ingestion (milk × time, P > 0.05). Breath H2 was greater after CM than SM (72 ± 10 vs. 43 ± 6 ppm at 240 min, P < 0.001), which may be due to greater lactose content in CM (33 vs. 25 g). Accordingly, when corrected for the lactose content breath H2 did not differ between the two milks. The response remained similar when analyzed in the LNP subset alone (n = 20). Conclusions: Despite a higher energy and nutrient content, SM did not increase adverse digestive symptoms after ingestion, relative to CM, although there was a reduced breath H2 response, which could be attributed to the lower lactose content in SM. The tolerability of SM should be explored in populations without lactose intolerance for whom underlying trigger for intolerance is unknown.
Collapse
Affiliation(s)
- Aahana Shrestha
- The Liggins Institute, The University of Auckland, Auckland, New Zealand.,Riddet Institute, Palmerston North, New Zealand
| | | | - Pankaja Sharma
- The Liggins Institute, The University of Auckland, Auckland, New Zealand.,Riddet Institute, Palmerston North, New Zealand
| | - Li Day
- AgResearch Ltd., Te Ohu Rangahau Kai, Palmerston North, New Zealand
| | - David Cameron-Smith
- The Liggins Institute, The University of Auckland, Auckland, New Zealand.,Riddet Institute, Palmerston North, New Zealand.,Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Amber M Milan
- The Liggins Institute, The University of Auckland, Auckland, New Zealand.,AgResearch Ltd., Te Ohu Rangahau Kai, Palmerston North, New Zealand
| |
Collapse
|
12
|
May-Zhang AA, Tycksen E, Southard-Smith AN, Deal KK, Benthal JT, Buehler DP, Adam M, Simmons AJ, Monaghan JR, Matlock BK, Flaherty DK, Potter SS, Lau KS, Southard-Smith EM. Combinatorial Transcriptional Profiling of Mouse and Human Enteric Neurons Identifies Shared and Disparate Subtypes In Situ. Gastroenterology 2021; 160:755-770.e26. [PMID: 33010250 PMCID: PMC7878294 DOI: 10.1053/j.gastro.2020.09.032] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/24/2020] [Accepted: 09/17/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND & AIMS The enteric nervous system (ENS) coordinates essential intestinal functions through the concerted action of diverse enteric neurons (ENs). However, integrated molecular knowledge of EN subtypes is lacking. To compare human and mouse ENs, we transcriptionally profiled healthy ENS from adult humans and mice. We aimed to identify transcripts marking discrete neuron subtypes and visualize conserved EN subtypes for humans and mice in multiple bowel regions. METHODS Human myenteric ganglia and adjacent smooth muscle were isolated by laser-capture microdissection for RNA sequencing. Ganglia-specific transcriptional profiles were identified by computationally subtracting muscle gene signatures. Nuclei from mouse myenteric neurons were isolated and subjected to single-nucleus RNA sequencing, totaling more than 4 billion reads and 25,208 neurons. Neuronal subtypes were defined using mouse single-nucleus RNA sequencing data. Comparative informatics between human and mouse data sets identified shared EN subtype markers, which were visualized in situ using hybridization chain reaction. RESULTS Several EN subtypes in the duodenum, ileum, and colon are conserved between humans and mice based on orthologous gene expression. However, some EN subtype-specific genes from mice are expressed in completely distinct morphologically defined subtypes in humans. In mice, we identified several neuronal subtypes that stably express gene modules across all intestinal segments, with graded, regional expression of 1 or more marker genes. CONCLUSIONS Our combined transcriptional profiling of human myenteric ganglia and mouse EN provides a rich foundation for developing novel intestinal therapeutics. There is congruency among some EN subtypes, but we note multiple species differences that should be carefully considered when relating findings from mouse ENS research to human gastrointestinal studies.
Collapse
Affiliation(s)
- Aaron A May-Zhang
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Eric Tycksen
- Genome Technology Access Center, McDonnell Genome Institute, St Louis, Missouri
| | - Austin N Southard-Smith
- Epithelial Biology Center and the Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Karen K Deal
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Joseph T Benthal
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Dennis P Buehler
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Mike Adam
- University of Cincinnati Children's Medical Hospital Research Center, Cincinnati, Ohio
| | - Alan J Simmons
- Epithelial Biology Center and the Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - James R Monaghan
- Northeastern University, Department of Biology, Boston, Massachusetts
| | - Brittany K Matlock
- Office of Shared Resources, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - David K Flaherty
- Office of Shared Resources, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - S Steven Potter
- University of Cincinnati Children's Medical Hospital Research Center, Cincinnati, Ohio
| | - Ken S Lau
- Epithelial Biology Center and the Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - E Michelle Southard-Smith
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee.
| |
Collapse
|
13
|
Ferrer M, Aguilera M, Martinez V. Effects of Rifaximin on Luminal and Wall-Adhered Gut Commensal Microbiota in Mice. Int J Mol Sci 2021; 22:E500. [PMID: 33419066 PMCID: PMC7825446 DOI: 10.3390/ijms22020500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/30/2020] [Accepted: 01/01/2021] [Indexed: 12/12/2022] Open
Abstract
Rifaximin is a broad-spectrum antibiotic that ameliorates symptomatology in inflammatory/functional gastrointestinal disorders. We assessed changes in gut commensal microbiota (GCM) and Toll-like receptors (TLRs) associated to rifaximin treatment in mice. Adult C57BL/6NCrl mice were treated (7/14 days) with rifaximin (50/150 mg/mouse/day, PO). Luminal and wall-adhered ceco-colonic GCM were characterized by fluorescent in situ hybridization (FISH) and microbial profiles determined by terminal restriction fragment length polymorphism (T-RFLP). Colonic expression of TLR2/3/4/5/7 and immune-related markers was assessed (RT-qPCR). Regardless the period of treatment or the dose, rifaximin did not alter total bacterial counts or bacterial biodiversity. Only a modest increase in Bacteroides spp. (150 mg/1-week treatment) was detected. In control conditions, only Clostridium spp. and Bifidobacterium spp. were found attached to the colonic epithelium. Rifaximin showed a tendency to favour their adherence after a 1-week, but not 2-week, treatment period. Minor up-regulation in TLRs expression was observed. Only the 50 mg dose for 1-week led to a significant increase (by 3-fold) in TLR-4 expression. No changes in the expression of immune-related markers were observed. Rifaximin, although its antibacterial properties, induces minor changes in luminal and wall-adhered GCM in healthy mice. Moreover, no modulation of TLRs or local immune systems was observed. These findings, in normal conditions, do not rule out a modulatory role of rifaximin in inflammatory and or dysbiotic states of the gut.
Collapse
Affiliation(s)
- Marina Ferrer
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (M.F.); (M.A.)
- Neuroscience Institute, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Mònica Aguilera
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (M.F.); (M.A.)
- Neuroscience Institute, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Vicente Martinez
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (M.F.); (M.A.)
- Neuroscience Institute, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Centro de Investigación Biomédicaen Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
14
|
Wiley JW, Zong Y, Zheng G, Zhu S, Hong S. Histone H3K9 methylation regulates chronic stress and IL-6-induced colon epithelial permeability and visceral pain. Neurogastroenterol Motil 2020; 32:e13941. [PMID: 32743845 PMCID: PMC8007084 DOI: 10.1111/nmo.13941] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/08/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Chronic stress is associated with activation of the HPA axis, elevation in pro-inflammatory cytokines, decrease in intestinal epithelial cell tight junction (TJ) proteins, and enhanced visceral pain. It is unknown whether epigenetic regulatory pathways play a role in chronic stress-induced intestinal barrier dysfunction and visceral hyperalgesia. METHODS Young adult male rats were subjected to water avoidance stress ± H3K9 methylation inhibitors or siRNAs. Visceral pain response was assessed. Differentiated Caco-2/BBE cells and human colonoids were treated with cortisol or IL-6 ± antagonists. Expression of TJ, IL-6, and H3K9 methylation status at gene promoters was measured. Transepithelial electrical resistance and FITC-dextran permeability were evaluated. KEY RESULTS Chronic stress induced IL-6 up-regulation prior to a decrease in TJ proteins in the rat colon. The IL-6 level inversely correlated with occludin expression. Treatment with IL-6 decreased occludin and induced visceral hyperalgesia. Chronic stress and IL-6 increased H3K9 methylation and decreased transcriptional GR binding to the occludin gene promoter, leading to down-regulation of protein expression and increase in paracellular permeability. Intrarectal administration of a H3K9 methylation antagonist prevented chronic stress-induced visceral hyperalgesia in the rat. In a human colonoid model, cortisol decreased occludin expression, which was prevented by the GR antagonist RU486, and IL-6 increased H3K9 methylation and decreased TJ protein levels, which were prevented by inhibitors of H3K9 methylation. CONCLUSIONS & INFERENCES Our findings support a novel role for methylation of the repressive histone H3K9 to regulate chronic stress, pro-inflammatory cytokine-mediated reduction in colon TJ protein levels, and increase in paracellular permeability and visceral hyperalgesia.
Collapse
Affiliation(s)
- John W Wiley
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, 48109 USA
| | - Ye Zong
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Gen Zheng
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, 48109 USA
| | - Shengtao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shuangsong Hong
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, 48109 USA
| |
Collapse
|
15
|
Diviccaro S, Giatti S, Borgo F, Falvo E, Caruso D, Garcia-Segura LM, Melcangi RC. Steroidogenic machinery in the adult rat colon. J Steroid Biochem Mol Biol 2020; 203:105732. [PMID: 32777355 DOI: 10.1016/j.jsbmb.2020.105732] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022]
Abstract
Gastrointestinal function is known to be regulated by steroid molecules produced by the gonads, the adrenal glands and the gut microbiota. However, we have a limited knowledge on the functional significance of local steroid production by gastrointestinal tract tissue. On this basis, we have here evaluated, as a first methodological approach, the expression of steroidogenic molecules and the local levels of key steroids in the male rat colon. Our findings indicate that the colon tissue expresses molecules involved in the early steps of steroidogenesis and in the consecutive synthesis and metabolism of steroid hormones, such as progesterone, testosterone and 17β-estradiol. In addition, the levels of the steroid hormone precursor pregnenolone and the levels of active metabolites of progesterone and testosterone, such as dihydroprogesterone, tetrahydroprogesterone, dihydrotestosterone and 17β-estradiol, were higher in colon than in plasma. Higher levels of the androgen metabolite 3α-diol were detected in the colon in comparison with another non-classical steroidogenic tissue, such as the cerebral cortex. These findings suggest the existence of local steroid synthesis and metabolism in the colon, with the production of active steroid metabolites that may impact on the activity of the enteric nervous system and on the composition of the gut microbiota.
Collapse
Affiliation(s)
- S Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - S Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - F Borgo
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - E Falvo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - D Caruso
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - L M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - R C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy,.
| |
Collapse
|
16
|
Sørup FKH, Eriksson R, Westergaard D, Hallas J, Brunak S, Ejdrup Andersen S. Sex differences in text-mined possible adverse drug events associated with drugs for psychosis. J Psychopharmacol 2020; 34:532-539. [PMID: 32048538 DOI: 10.1177/0269881120903466] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Understanding sex differences in adverse drug reactions to drugs for psychosis could potentially guide clinicians in optimal drug choices. AIMS By applying a text-mining approach, this study aimed to investigate the relationship between drugs for psychosis and biological sex differences in frequencies and co-occurrences of potential adverse drug events (ADEs). METHODS Electronic patient records of a psychiatric population (1427 men and 727 women) were text mined for potential ADEs. The relative risk of experiencing specific ADEs and co-occurrence of ADEs were calculated for each sex. RESULTS Findings included 55 potential ADEs with significantly different frequencies between the two sexes. Of these, 20 were more frequent in men, with relative risks of 1.10-7.64, and 35 were more frequent in women, with relative risks of 1.19-21.58. Frequent potential ADEs were psychiatric symptoms, including sexual dysfunction and disturbances in men, and gastrointestinal symptoms, suicidal and self-injurious behaviour and hyperprolactinemia-related events in women. Mention of different hyperprolactinemia-related ADEs often co-occurred in female patients but not in male patients. CONCLUSION Several known sex-related ADEs were identified, as well as some previously not reported. When considering the risk-benefit profile of drugs for psychosis, the patient's sex should be considered.
Collapse
Affiliation(s)
- Freja Karuna Hemmingsen Sørup
- Clinical Pharmacology Unit, Zealand University Hospital, Roskilde, Denmark.,Disease Systems Biology, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Robert Eriksson
- Disease Systems Biology, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - David Westergaard
- Disease Systems Biology, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Hallas
- Clinical Pharmacology and Pharmacy, University of Southern Denmark, Odense, Denmark
| | - Søren Brunak
- Disease Systems Biology, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
17
|
Corticotropin-Releasing Factor Family: A Stress Hormone-Receptor System's Emerging Role in Mediating Sex-Specific Signaling. Cells 2020; 9:cells9040839. [PMID: 32244319 PMCID: PMC7226788 DOI: 10.3390/cells9040839] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/25/2020] [Accepted: 03/28/2020] [Indexed: 12/14/2022] Open
Abstract
No organ in the body is impervious to the effects of stress, and a coordinated response from all organs is essential to deal with stressors. A dysregulated stress response that fails to bring systems back to homeostasis leads to compromised function and ultimately a diseased state. The components of the corticotropin-releasing factor (CRF) family, an ancient and evolutionarily conserved stress hormone-receptor system, helps both initiate stress responses and bring systems back to homeostasis once the stressors are removed. The mammalian CRF family comprises of four known agonists, CRF and urocortins (UCN1–3), and two known G protein-coupled receptors (GPCRs), CRF1 and CRF2. Evolutionarily, precursors of CRF- and urocortin-like peptides and their receptors were involved in osmoregulation/diuretic functions, in addition to nutrient sensing. Both CRF and UCN1 peptide hormones as well as their receptors appeared after a duplication event nearly 400 million years ago. All four agonists and both CRF receptors show sex-specific changes in expression and/or function, and single nucleotide polymorphisms are associated with a plethora of human diseases. CRF receptors harbor N-terminal cleavable peptide sequences, conferring biased ligand properties. CRF receptors have the ability to heteromerize with each other as well as with other GPCRs. Taken together, CRF receptors and their agonists due to their versatile functional adaptability mediate nuanced responses and are uniquely positioned to orchestrate sex-specific signaling and function in several tissues.
Collapse
|
18
|
Soni KG, Halder T, Conner ME, Preidis GA. Sexual dimorphism in upper gastrointestinal motility is dependent on duration of fast, time of day, age, and strain of mice. Neurogastroenterol Motil 2019; 31:e13654. [PMID: 31157504 PMCID: PMC6693981 DOI: 10.1111/nmo.13654] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/29/2019] [Accepted: 05/24/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND An important limitation of gastrointestinal motility testing is high variability. Conditions that could contribute to variability, including the duration of pretest fasting and time of day, are rarely reported and have not been examined systematically. This study aimed to explore whether these conditions, as well as age, sex, and strain of mice, affect the results of a standard laboratory test of upper gastrointestinal motility. METHODS Male and female 8-week-old C57BL/6J mice received a gastric gavage of fluorescein isothiocyanate (FITC)-conjugated dextran. FITC-dextran distribution was measured 30 minutes later. Mean geometric centers (MGCs) were calculated to determine the effects of short versus prolonged fasting and morning versus afternoon testing. The influence of age was assessed in 2- to 10-week-old animals, and the influence of strain was determined in C57BL/6J, BALB/c, and CD-1 mice. KEY RESULTS Motility was sexually dimorphic. MGC progressed 19% further in 8-week-old C57BL/6J males versus females when tested in the morning after a short fast. Similar patterns were observed in morning or afternoon testing after overnight fasting. In males, motility was unaffected by time of day; however, MGC progressed 31% further in females tested in the afternoon versus morning after a short fast. Sex differences also were present in CD-1 but not BALB/c mice. Testing in neonates revealed strikingly low variability and no sex differences. CONCLUSIONS & INFERENCES Fasting duration, time of day, age, sex, and strain of mice all influence upper gastrointestinal motility testing. Sex differences are not present in neonatal pups, but develop soon after weaning.
Collapse
Affiliation(s)
- Krishnakant G. Soni
- Section of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Tripti Halder
- Section of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Margaret E. Conner
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Geoffrey A. Preidis
- Section of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| |
Collapse
|
19
|
Meister AL, Jiang Y, Doheny KK, Travagli RA. Correlation between the motility of the proximal antrum and the high-frequency power of heart rate variability in freely moving rats. Neurogastroenterol Motil 2019; 31:e13633. [PMID: 31119854 PMCID: PMC6639127 DOI: 10.1111/nmo.13633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/18/2019] [Accepted: 05/08/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Cardiac vagal tone can be monitored non-invasively via electrocardiogram measurements of the high-frequency power spectrum of heart rate variability (HF-HRV). Vagal inputs to the upper GI tract are cumbersome to measure non-invasively. Although cardiac and GI vagal outputs arise from distinct brainstem nuclei, the nucleus ambiguus, and the dorsal motor nucleus of the vagus, respectively, we aim to test the hypotheses that in freely moving rats HF-HRV power is correlated to proximal antral motility and can be altered by high levels of circulating estrogen and vagal-selective treatments known to affect antral motility. METHODS Male and female Sprague-Dawley rats were implanted with a miniaturized strain gauge on the proximal gastric antrum and ECG electrodes to collect simultaneous antral motility and electrocardiogram. After recovery, male rats underwent baseline recordings before and after administration of saline (N = 8), cholecystokinin (CCK; N = 7), ghrelin (N = 6), or food (N = 6). Female rats (N = 6) underwent twice-daily recordings to determine baseline correlations during estrous cycle stages. KEY RESULTS There was a significant positive correlation between HF-HRV and proximal antral motility at baseline in males and females with low, but not high, estrogen levels. In male rats, the significant positive correlation was maintained following CCK, but not ghrelin or food administration. CONCLUSIONS AND INFERENCES Our data suggest that in rodents, HF-HRV positively correlates to proximal antral motility at baseline conditions in males and low-estrogen females or following interventions, such as CCK, known to affect vagal tone. This correlation is not observed when antral motility is influenced by more complex events.
Collapse
Affiliation(s)
- Alissa L. Meister
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey PA
| | - Yanyan Jiang
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey PA
| | - Kim K. Doheny
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey PA,Division of Neonatal-Perinatal Medicine, Penn State College of Medicine, Hershey PA
| | - R. Alberto Travagli
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey PA,Corresponding author: Dr. R. Alberto Travagli, Department of Neural and Behavioral Sciences, Penn State College of Medicine, 500 University Drive, MC H109, Hershey, PA 17033,
| |
Collapse
|
20
|
Age and Sex-Dependent Differences in the Neurochemical Characterization of Calcitonin Gene-Related Peptide-Like Immunoreactive (CGRP-LI) Nervous Structures in the Porcine Descending Colon. Int J Mol Sci 2019; 20:ijms20051024. [PMID: 30818742 PMCID: PMC6429317 DOI: 10.3390/ijms20051024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/21/2019] [Accepted: 02/23/2019] [Indexed: 12/26/2022] Open
Abstract
Neurons of the enteric nervous system (ENS) may undergo changes during maturation and aging, but knowledge of physiological stimuli-dependent changes in the ENS is still fragmentary. On the other hand, the frequency of many ENS-related intestinal illnesses depends on age and/or sex. The double immunofluorescence technique was used to study the influence of both of these factors on calcitonin gene-related peptide (CGRP)—positive enteric nervous structures—in the descending colon in young and adult female and castrated male pigs. The influence of age and gender on the number and neurochemical characterization (i.e., co-localization of CGRP with substance P, nitric oxide synthase, galanin, cocaine- and amphetamine-regulated transcript peptide and vesicular acetylcholine transporter) of CGRP-positive nerve structures in the colonic wall has been shown. These observations strongly suggest the participation of CGRP in adaptive processes in the ENS during GI tract maturation. Moreover, although the castration of males may mask some aspects of sex-dependent influences on the ENS, the sex-specific differences in CGRP-positive nervous structures were mainly visible in adult animals. This may suggest that the distribution and exact role of this substance in the ENS depend on the sex hormones.
Collapse
|
21
|
Tower J. Sex-Specific Gene Expression and Life Span Regulation. Trends Endocrinol Metab 2017; 28:735-747. [PMID: 28780002 PMCID: PMC5667568 DOI: 10.1016/j.tem.2017.07.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/09/2017] [Accepted: 07/10/2017] [Indexed: 11/18/2022]
Abstract
Aging-related diseases show a marked sex bias. For example, women live longer than men yet have more Alzheimer's disease and osteoporosis, whereas men have more cancer and Parkinson's disease. Understanding the role of sex will be important in designing interventions and in understanding basic aging mechanisms. Aging also shows sex differences in model organisms. Dietary restriction (DR), reduced insulin/IGF1-like signaling (IIS), and reduced TOR signaling each increase life span preferentially in females in both flies and mice. Maternal transmission of mitochondria to offspring may lead to greater control over mitochondrial functions in females, including greater life span and a larger response to diet. Consistent with this idea, males show greater loss of mitochondrial gene expression with age.
Collapse
Affiliation(s)
- John Tower
- Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|