1
|
Page AJ. Plasticity of gastrointestinal vagal afferents in terms of feeding-related physiology and pathophysiology. J Physiol 2024; 602:4763-4776. [PMID: 37737742 DOI: 10.1113/jp284075] [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: 06/09/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023] Open
Abstract
Gastrointestinal vagal afferents play an important role in communicating food related information from the gut to the brain. This information initiates vago-vagal reflexes essential for gut functions, including gut motility and secretions. These afferents also play a role in energy homeostasis, signalling the arrival, amount and nutrient composition of a meal to the central nervous system where it is processed ultimately leading to termination of a meal. Vagal afferent responses to food related stimuli demonstrate a high degree of plasticity, responding to short term changes in nutritional demand, such as the fluctuations that occur across a 24-hr or in response to a fast, as well as long term changes in energy demand, such as occurs during pregnancy. This plasticity is disrupted in disease states, such as obesity or chronic stress where there is hypo- and hypersensitivity of these afferents, respectively. Improved understanding of the plasticity of these afferents will enable identification of new treatment options for diseases associated with vagal afferent function.
Collapse
Affiliation(s)
- Amanda J Page
- Vagal Afferent Research Group, School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme, South Australian Health and Medical Research Institute, SAHMRI, Adelaide, South Australia, Australia
| |
Collapse
|
2
|
da Silva MDV, da Silva Bonassa L, Piva M, Basso CR, Zaninelli TH, Machado CCA, de Andrade FG, Miqueloto CA, Sant Ana DDMG, Aktar R, Peiris M, Aziz Q, Blackshaw LA, Verri WA, de Almeida Araújo EJ. Perineuronal net in the extrinsic innervation of the distal colon of mice and its remodeling in ulcerative colitis. J Neurochem 2024; 168:1937-1955. [PMID: 38426587 DOI: 10.1111/jnc.16080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 03/02/2024]
Abstract
The perineuronal net (PNN) is a well-described highly specialized extracellular matrix structure found in the central nervous system. Thus far, no reports of its presence or connection to pathological processes have been described in the peripheral nervous system. Our study demonstrates the presence of a PNN in the spinal afferent innervation of the distal colon of mice and characterizes structural and morphological alterations induced in an ulcerative colitis (UC) model. C57Bl/6 mice were given 3% dextran sulfate sodium (DSS) to induce acute or chronic UC. L6/S1 dorsal root ganglia (DRG) were collected. PNNs were labeled using fluorescein-conjugated Wisteria Floribunda (WFA) l lectin, and calcitonin gene-related peptide (CGRP) immunofluorescence was used to detect DRG neurons. Most DRG cell bodies and their extensions toward peripheral nerves were found surrounded by the PNN-like structure (WFA+), labeling neurons' cytoplasm and the pericellular surfaces. The amount of WFA+ neuronal cell bodies was increased in both acute and chronic UC, and the PNN-like structure around cell bodies was thicker in UC groups. In conclusion, a PNN-like structure around DRG neuronal cell bodies was described and found modulated by UC, as changes in quantity, morphology, and expression profile of the PNN were detected, suggesting a potential role in sensory neuron peripheral sensitization, possibly modulating the pain profile of ulcerative colitis.
Collapse
Affiliation(s)
- Matheus Deroco Veloso da Silva
- Laboratory of Neurogastroenterology, Department of Histology, State University of Londrina, Londrina, Paraná, Brazil
- Laboratory of Pain, Inflammation, Neuropathy and Cancer, Department of Pathology, State University of Londrina, Londrina, Paraná, Brazil
| | - Larissa da Silva Bonassa
- Laboratory of Neurogastroenterology, Department of Histology, State University of Londrina, Londrina, Paraná, Brazil
| | - Maiara Piva
- Laboratory of Pain, Inflammation, Neuropathy and Cancer, Department of Pathology, State University of Londrina, Londrina, Paraná, Brazil
| | - Camila Regina Basso
- Laboratory of Neurogastroenterology, Department of Histology, State University of Londrina, Londrina, Paraná, Brazil
| | - Tiago Henrique Zaninelli
- Laboratory of Pain, Inflammation, Neuropathy and Cancer, Department of Pathology, State University of Londrina, Londrina, Paraná, Brazil
| | - Camila Cristina Alves Machado
- Laboratory of Neurogastroenterology, Department of Histology, State University of Londrina, Londrina, Paraná, Brazil
| | - Fábio Goulart de Andrade
- Laboratory of Histopathological Analysis, Department of Histology, State University of Londrina, Londrina, Paraná, Brazil
| | - Carlos Alberto Miqueloto
- Laboratory of Neurogastroenterology, Department of Histology, State University of Londrina, Londrina, Paraná, Brazil
| | | | - Rubina Aktar
- Wingate Institute for Neurogastroenterology, Queen Mary University of London, London, UK
| | - Madusha Peiris
- Wingate Institute for Neurogastroenterology, Queen Mary University of London, London, UK
| | - Qasim Aziz
- Wingate Institute for Neurogastroenterology, Queen Mary University of London, London, UK
| | - L Ashley Blackshaw
- Wingate Institute for Neurogastroenterology, Queen Mary University of London, London, UK
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy and Cancer, Department of Pathology, State University of Londrina, Londrina, Paraná, Brazil
| | | |
Collapse
|
3
|
Wang L, Taché Y. The parasympathetic and sensory innervation of the proximal and distal colon in male mice. Front Neuroanat 2024; 18:1422403. [PMID: 39045348 PMCID: PMC11263295 DOI: 10.3389/fnana.2024.1422403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/24/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction The distributions of extrinsic neurons innervating the colon show differences in experimental animals from humans, including the vagal and spinal parasympathetic innervation to the distal colon. The neuroanatomical tracing to the mouse proximal colon has not been studied in details. This study aimed to trace the locations of extrinsic neurons projecting to the mouse proximal colon compared to the distal colon using dual retrograde tracing. Methods The parasympathetic and sensory neurons projecting to colon were assessed using Cholera Toxin subunit B conjugated to Alexa-Fluor 488 or 555 injected in the proximal and distal colon of the same mice. Results Retrograde tracing from the proximal and distal colon labeled neurons in the dorsal motor nucleus of the vagus (DMV) and the nodose ganglia, while the tracing from the distal colon did not label the parasympathetic neurons in the lumbosacral spinal cord at L6-S1. Neurons in the pelvic ganglia which were cholinergic projected to the distal colon. There were more neurons in the DMV and nodose ganglia projecting to the proximal than distal colon. The right nodose ganglion had a higher number of neurons than the left ganglion innervating the proximal colon. In the dorsal root ganglia (DRG), the highest number of neurons traced from the distal colon were at L6, and those from the proximal colon at T12. DRG neurons projected closely to the cholinergic neurons in the intermediolateral column of L6 spinal cord. Small percentages of neurons with dual projections to both the proximal and distal colon existed in the DMV, nodose ganglia and DRG. We also observed long projecting neurons traced from the caudal distal colon to the transverse and proximal colon, some of which were calbindin immunoreactive, while there were no retrogradely labeled neurons traced from the proximal to distal colon. Discussion These data demonstrated that the vagal motor and motor and sensory neurons innervate both the proximal and distal colon in mice, and the autonomic neurons in the intermediate zone of the lumbosacral spinal cord do not project directly to the mouse colon, which differs from that in humans.
Collapse
Affiliation(s)
- Lixin Wang
- CURE/Digestive Diseases Research Center, Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Yvette Taché
- CURE/Digestive Diseases Research Center, Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
4
|
Baidoo N, Sanger GJ. The human colon: Evidence for degenerative changes during aging and the physiological consequences. Neurogastroenterol Motil 2024:e14848. [PMID: 38887160 DOI: 10.1111/nmo.14848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/16/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND The incidence of constipation increases among the elderly (>65 years), while abdominal pain decreases. Causes include changes in lifestyle (e.g., diet and reduced exercise), disease and medications affecting gastrointestinal functions. Degenerative changes may also occur within the colo-rectum. However, most evidence is from rodents, animals with relatively high rates of metabolism and accelerated aging, with considerable variation in time course. In humans, cellular and non-cellular changes in the aging intestine are poorly investigated. PURPOSE To examine all available studies which reported the effects of aging on cellular and tissue functions of human isolated colon, noting the region studied, sex and age of tissue donors and study size. The focus on human colon reflects the ability to access full-thickness tissue over a wide age range, compared with other gastrointestinal regions. Details are important because of natural human variability. We found age-related changes within the muscle, in the enteric and nociceptor innervation, and in the submucosa. Some involve all regions of colon, but the ascending colon appears more vulnerable. Changes can be cell- and sublayer-dependent. Mechanisms are unclear but may include development of "senescent-like" and associated inflammaging, perhaps associated with increased mucosal permeability to harmful luminal contents. In summary, reduced nociceptor innervation can explain diminished abdominal pain among the elderly. Degenerative changes within the colon wall may have little impact on symptoms and colonic functions, because of high "functional reserve," but are likely to facilitate the development of constipation during age-related challenges (e.g., lifestyle, disease, and medications), now operating against a reduced functional reserve.
Collapse
Affiliation(s)
- Nicholas Baidoo
- School of Life Sciences, University of Westminster, London, UK
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gareth J Sanger
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
5
|
Chopra H, Jackels M, Suarez M, Vu PD, Broachwala M, AlFarra T, Sivanesan E. Dorsal root ganglion stimulation provides significant functional improvement from acute debilitating Crohn's disease: A novel use. INTERVENTIONAL PAIN MEDICINE 2024; 3:100389. [PMID: 39239493 PMCID: PMC11373045 DOI: 10.1016/j.inpm.2024.100389] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 09/07/2024]
Abstract
Crohn's disease is a chronic inflammatory bowel condition causing symptoms, notably pain, due to ongoing intestinal inflammation or complications like abscesses, strictures, and fistulas, which are common in IBD patients. Abdominal pain affects up to 60 % of IBD patients, irrespective of disease severity, prompting medical attention. Various medications like NSAIDs, antidepressants, antispasmodics, anticonvulsants, and opioids are used to manage pain, but they have limited effectiveness and potential side effects, even during remission. In this case, a 20-year-old Caucasian female college student [height 5'4″, weight 120lbs (54.4 kg)] with juvenile idiopathic arthritis and Crohn's disease experienced severe daily abdominal pain, negatively impacting her life. Despite a multimodal regimen, including gabapentin, nortriptyline, duloxetine, and acetaminophen, her pain persisted, significantly affecting her appetite, sleep, mood, activity level, and overall quality of life (QOL). To address this, dorsal root ganglion (DRG) stimulation was considered. The patient aimed for a 20 % pain reduction and improved QOL. Trial leads were placed along the T10 and T12 DRG, resulting in a 25 % pain reduction (8-6 out of 10) and substantial QOL improvement. She could eat, sleep without interruptions, walk longer distances, and be more active. The T12 lead was more effective than the T10, targeting upper abdomen stimulation. The patient and her mother were highly satisfied and opted for permanent implantation for the T11 and T12 DRG. While DRG stimulation was approved in 2016 for chronic pain, to our knowledge, this is the first reported case of its use in a patient with debilitating Crohn's disease.
Collapse
Affiliation(s)
| | | | | | - Peter D Vu
- The University of Texas Health Science Center at Houston, USA
| | | | | | | |
Collapse
|
6
|
Riehl L, Fürst J, Kress M, Rykalo N. The importance of the gut microbiome and its signals for a healthy nervous system and the multifaceted mechanisms of neuropsychiatric disorders. Front Neurosci 2024; 17:1302957. [PMID: 38249593 PMCID: PMC10797776 DOI: 10.3389/fnins.2023.1302957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024] Open
Abstract
Increasing evidence links the gut microbiome and the nervous system in health and disease. This narrative review discusses current views on the interaction between the gut microbiota, the intestinal epithelium, and the brain, and provides an overview of the communication routes and signals of the bidirectional interactions between gut microbiota and the brain, including circulatory, immunological, neuroanatomical, and neuroendocrine pathways. Similarities and differences in healthy gut microbiota in humans and mice exist that are relevant for the translational gap between non-human model systems and patients. There is an increasing spectrum of metabolites and neurotransmitters that are released and/or modulated by the gut microbiota in both homeostatic and pathological conditions. Dysbiotic disruptions occur as consequences of critical illnesses such as cancer, cardiovascular and chronic kidney disease but also neurological, mental, and pain disorders, as well as ischemic and traumatic brain injury. Changes in the gut microbiota (dysbiosis) and a concomitant imbalance in the release of mediators may be cause or consequence of diseases of the central nervous system and are increasingly emerging as critical links to the disruption of healthy physiological function, alterations in nutrition intake, exposure to hypoxic conditions and others, observed in brain disorders. Despite the generally accepted importance of the gut microbiome, the bidirectional communication routes between brain and gut are not fully understood. Elucidating these routes and signaling pathways in more detail offers novel mechanistic insight into the pathophysiology and multifaceted aspects of brain disorders.
Collapse
Affiliation(s)
| | | | | | - Nadiia Rykalo
- Institute of Physiology, Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
7
|
Meling S, Tjora E, Eichele H, Nedergaard RB, Knop FK, Ejskjaer N, Carlsen S, Njølstad PR, Brock C, Søfteland E. Rectal sensitivity correlated with gastrointestinal-mediated glucose disposal, but not the incretin effect. Endocrinol Diabetes Metab 2024; 7:e463. [PMID: 38059537 PMCID: PMC10782140 DOI: 10.1002/edm2.463] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/08/2023] [Accepted: 11/19/2023] [Indexed: 12/08/2023] Open
Abstract
OBJECTIVE The mechanisms behind the diminished incretin effect in type 2 diabetes are uncertain, but impaired vagal transmission has been suggested. We aimed to investigate the association between the incretin effect and autonomic neuropathy, and the degree of dysglycaemia and duration of diabetes. DESIGN AND METHODS For a cross-sectional study, we included participants with either longstanding type 2 diabetes, recent onset, untreated diabetes and controls without diabetes matched for age, sex and body mass index. Autonomic nerve function was assessed with cardiovascular reflex tests, heart rate variability and sudomotor function. Visceral afferent nerves in the gut were tested performing rapid rectal balloon distention. An oral glucose tolerance test and an intravenous isoglycaemic glucose infusion were performed to calculate the incretin effect and gastrointestinal-mediated glucose disposal (GIGD). RESULTS Sixty-five participants were recruited. Participants with diabetes had rectal hyposensitivity for earliest sensation (3.7 ± 1.1 kPa in longstanding, 4.0 ± 1.3 in early), compared to controls (3.0 ± 0.9 kPa), p = .005. Rectal hyposensitivity for earliest sensation was not associated with the incretin effect (rho = -0.204, p = .106), but an association was found with GIGD (rho -0.341, p = .005). Incretin effect and GIGD were correlated with all glucose values, HbA1c and duration of diabetes. CONCLUSIONS Rectal hyposensitivity was uncovered in both longstanding and early type 2 diabetes, and was not associated with the incretin effect, but with GIGD, implying a potential link between visceral neuropathy and gastrointestinal handling of glucose. Both the incretin effect and GIGD were associated with the degree of dysglycaemia and the duration of diabetes. PREVIOUSLY PUBLISHED Some of the data have previously been published and presented as a poster on the American Diabetes Association 83rd Scientific Sessions: Meling et al; 1658-P: Rectal Hyposensitivity, a Potential Marker of Enteric Autonomic Nerve Dysfunction, Is Significantly Associated with Gastrointestinally Mediated Glucose Disposal in Persons with Type 2 Diabetes. Diabetes 20 June 2023; 72 (Supplement_1): 1658-P. https://doi.org/10.2337/db23-1658-P.
Collapse
Affiliation(s)
- Sondre Meling
- Department of MedicineStavanger University HospitalStavangerNorway
- Department of Clinical ScienceUniversity of BergenBergenNorway
| | - Erling Tjora
- Department of Clinical ScienceUniversity of BergenBergenNorway
- Children and Youth ClinicHaukeland University HospitalBergenNorway
| | - Heike Eichele
- Department of Biological and Medical Psychology, Faculty of PsychologyUniversity of BergenBergenNorway
- Regional resource Centre for Autism, ADHD and Tourette Syndrome Western Norway, Division of PsychiatryHaukeland University HospitalBergenNorway
| | - Rasmus B. Nedergaard
- Mech‐Sense, Department of Gastroenterology and HepatologyAalborg University HospitalAalborgDenmark
| | - Filip K. Knop
- Center for Clinical Metabolic ResearchCopenhagen University Hospital—Herlev and GentofteCopenhagenDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Steno Diabetes Center CopenhagenGentofteDenmark
- Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Niels Ejskjaer
- Department of Clinical Medicine, Faculty of MedicineAalborg University HospitalAalborgDenmark
- Steno Diabetes Center North DenmarkAalborg University HospitalAalborgDenmark
- Department of EndocrinologyAalborg University HospitalAalborgDenmark
| | - Siri Carlsen
- Department of MedicineStavanger University HospitalStavangerNorway
| | - Pål R. Njølstad
- Department of Clinical ScienceUniversity of BergenBergenNorway
- Children and Youth ClinicHaukeland University HospitalBergenNorway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical ScienceUniversity of BergenBergenNorway
| | - Christina Brock
- Mech‐Sense, Department of Gastroenterology and HepatologyAalborg University HospitalAalborgDenmark
- Department of Clinical Medicine, Faculty of MedicineAalborg University HospitalAalborgDenmark
- Steno Diabetes Center North DenmarkAalborg University HospitalAalborgDenmark
| | - Eirik Søfteland
- Department of Clinical ScienceUniversity of BergenBergenNorway
- Department of MedicineHaukeland University HospitalBergenNorway
| |
Collapse
|
8
|
Bhebhe CN, Higham JP, Gupta RA, Raine T, Bulmer DC. K V7 but not dual small and intermediate K Ca channel openers inhibit the activation of colonic afferents by noxious stimuli. Am J Physiol Gastrointest Liver Physiol 2023; 325:G436-G445. [PMID: 37667839 PMCID: PMC10894664 DOI: 10.1152/ajpgi.00141.2023] [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: 07/11/2023] [Revised: 08/21/2023] [Accepted: 08/28/2023] [Indexed: 09/06/2023]
Abstract
In numerous subtypes of central and peripheral neurons, small and intermediate conductance Ca2+-activated K+ (SK and IK, respectively) channels are important regulators of neuronal excitability. Transcripts encoding SK channel subunits, as well as the closely related IK subunit, are coexpressed in the soma of colonic afferent neurons with receptors for the algogenic mediators ATP and bradykinin, P2X3 and B2, highlighting the potential utility of these channels as drug targets for the treatment of abdominal pain in gastrointestinal diseases such as irritable bowel syndrome. Despite this, pretreatment with the dual SK/IK channel opener SKA-31 had no effect on the colonic afferent response to ATP, bradykinin, or noxious ramp distention of the colon. Inhibition of SK or IK channels with apamin or TRAM-34, respectively, yielded no change in spontaneous baseline afferent activity, indicating these channels are not tonically active. In contrast to its lack of effect in electrophysiological experiments, comparable concentrations of SKA-31 abolished ongoing peristaltic activity in the colon ex vivo. Treatment with the KV7 channel opener retigabine blunted the colonic afferent response to all applied stimuli. Our data therefore highlight the potential utility of KV7, but not SK/IK, channel openers as analgesic agents for the treatment of abdominal pain.NEW & NOTEWORTHY Despite marked coexpression of small (Kcnn1, Kcnn2) and intermediate (Kcnn4) conductance calcium-activated potassium channel transcripts with P2X3 (P2rx3) or bradykinin B2 (Bdkrb2) receptors in colonic sensory neurons, pharmacological activation of these channels had no effect on the colonic afferent response to ATP, bradykinin or luminal distension of the colon. This is in contrast to the robust inhibitory effect of the KV7 channel opener, retigabine.
Collapse
Affiliation(s)
- Charity N Bhebhe
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - James P Higham
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Rohit A Gupta
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Tim Raine
- Department of Gastroenterology, Addenbrookes Hospital, Cambridge University Teaching Hospitals, Cambridge, United Kingdom
| | - David C Bulmer
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
9
|
Ma J, Nguyen D, Madas J, Kwiat AM, Toledo Z, Bizanti A, Kogut N, Mistareehi A, Bendowski K, Zhang Y, Chen J, Li DP, Powley TL, Furness JB, Cheng Z. Spinal afferent innervation in flat-mounts of the rat stomach: anterograde tracing. Sci Rep 2023; 13:17675. [PMID: 37853008 PMCID: PMC10584867 DOI: 10.1038/s41598-023-43120-y] [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: 04/12/2023] [Accepted: 09/20/2023] [Indexed: 10/20/2023] Open
Abstract
The dorsal root ganglia (DRG) project spinal afferent axons to the stomach. However, the distribution and morphology of spinal afferent axons in the stomach have not been well characterized. In this study, we used a combination of state-of-the-art techniques, including anterograde tracer injection into the left DRG T7-T11, avidin-biotin and Cuprolinic Blue labeling, Zeiss M2 Imager, and Neurolucida to characterize spinal afferent axons in flat-mounts of the whole rat stomach muscular wall. We found that spinal afferent axons innervated all regions with a variety of distinct terminal structures innervating different gastric targets: (1) The ganglionic type: some axons formed varicose contacts with individual neurons within myenteric ganglia. (2) The muscle type: most axons ran in parallel with the longitudinal and circular muscles and expressed spherical varicosities. Complex terminal structures were observed within the circular muscle layer. (3) The ganglia-muscle mixed type: some individual varicose axons innervated both myenteric neurons and the circular muscle, exhibiting polymorphic terminal structures. (4) The vascular type: individual varicose axons ran along the blood vessels and occasionally traversed the vessel wall. This work provides a foundation for future topographical anatomical and functional mapping of spinal afferent axon innervation of the stomach under normal and pathophysiological conditions.
Collapse
Affiliation(s)
- Jichao Ma
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Duyen Nguyen
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Jazune Madas
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Andrew M Kwiat
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Zulema Toledo
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Ariege Bizanti
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Nicole Kogut
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Anas Mistareehi
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Kohlton Bendowski
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Yuanyuan Zhang
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Jin Chen
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - De-Pei Li
- Department of Medicine, Center for Precision Medicine, School of Medicine, University of Missouri, Columbia, MO, 65212, USA
| | - Terry L Powley
- Department of Psychological Sciences, Purdue University, West Lafayette, IN, 479062, USA
| | - John B Furness
- Department of Anatomy and Physiology, University of Melbourne, and Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Zixi Cheng
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA.
| |
Collapse
|
10
|
Hibberd TJ, Ramsay S, Spencer-Merris P, Dinning PG, Zagorodnyuk VP, Spencer NJ. Circadian rhythms in colonic function. Front Physiol 2023; 14:1239278. [PMID: 37711458 PMCID: PMC10498548 DOI: 10.3389/fphys.2023.1239278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/17/2023] [Indexed: 09/16/2023] Open
Abstract
A rhythmic expression of clock genes occurs within the cells of multiple organs and tissues throughout the body, termed "peripheral clocks." Peripheral clocks are subject to entrainment by a multitude of factors, many of which are directly or indirectly controlled by the light-entrainable clock located in the suprachiasmatic nucleus of the hypothalamus. Peripheral clocks occur in the gastrointestinal tract, notably the epithelia whose functions include regulation of absorption, permeability, and secretion of hormones; and in the myenteric plexus, which is the intrinsic neural network principally responsible for the coordination of muscular activity in the gut. This review focuses on the physiological circadian variation of major colonic functions and their entraining mechanisms, including colonic motility, absorption, hormone secretion, permeability, and pain signalling. Pathophysiological states such as irritable bowel syndrome and ulcerative colitis and their interactions with circadian rhythmicity are also described. Finally, the classic circadian hormone melatonin is discussed, which is expressed in the gut in greater quantities than the pineal gland, and whose exogenous use has been of therapeutic interest in treating colonic pathophysiological states, including those exacerbated by chronic circadian disruption.
Collapse
Affiliation(s)
- Timothy J. Hibberd
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Stewart Ramsay
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | | | - Phil G. Dinning
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Colorectal Surgical Unit, Division of Surgery, Flinders Medical Centre, Adelaide, SA, Australia
| | | | - Nick J. Spencer
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
11
|
Wolfson RL, Abdelaziz A, Rankin G, Kushner S, Qi L, Mazor O, Choi S, Sharma N, Ginty DD. DRG afferents that mediate physiologic and pathologic mechanosensation from the distal colon. Cell 2023; 186:3368-3385.e18. [PMID: 37541195 PMCID: PMC10440726 DOI: 10.1016/j.cell.2023.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 04/23/2023] [Accepted: 07/06/2023] [Indexed: 08/06/2023]
Abstract
The properties of dorsal root ganglia (DRG) neurons that innervate the distal colon are poorly defined, hindering our understanding of their roles in normal physiology and gastrointestinal (GI) disease. Here, we report genetically defined subsets of colon-innervating DRG neurons with diverse morphologic and physiologic properties. Four colon-innervating DRG neuron populations are mechanosensitive and exhibit distinct force thresholds to colon distension. The highest threshold population, selectively labeled using Bmpr1b genetic tools, is necessary and sufficient for behavioral responses to high colon distension, which is partly mediated by the mechanosensory ion channel Piezo2. This Aδ-HTMR population mediates behavioral over-reactivity to colon distension caused by inflammation in a model of inflammatory bowel disease. Thus, like cutaneous DRG mechanoreceptor populations, colon-innervating mechanoreceptors exhibit distinct anatomical and physiological properties and tile force threshold space, and genetically defined colon-innervating HTMRs mediate pathophysiological responses to colon distension, revealing a target population for therapeutic intervention.
Collapse
Affiliation(s)
- Rachel L Wolfson
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Division of Gastroenterology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Amira Abdelaziz
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Genelle Rankin
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Sarah Kushner
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Lijun Qi
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Ofer Mazor
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Seungwon Choi
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Nikhil Sharma
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Department of Systems Biology, Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY 10032, USA
| | - David D Ginty
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
12
|
Bayrer JR, Castro J, Venkataraman A, Touhara KK, Rossen ND, Morrie RD, Maddern J, Hendry A, Braverman KN, Garcia-Caraballo S, Schober G, Brizuela M, Castro Navarro FM, Bueno-Silva C, Ingraham HA, Brierley SM, Julius D. Gut enterochromaffin cells drive visceral pain and anxiety. Nature 2023; 616:137-142. [PMID: 36949192 PMCID: PMC10827380 DOI: 10.1038/s41586-023-05829-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 02/10/2023] [Indexed: 03/24/2023]
Abstract
Gastrointestinal (GI) discomfort is a hallmark of most gut disorders and represents an important component of chronic visceral pain1. For the growing population afflicted by irritable bowel syndrome, GI hypersensitivity and pain persist long after tissue injury has resolved2. Irritable bowel syndrome also exhibits a strong sex bias, afflicting women three times more than men1. Here, we focus on enterochromaffin (EC) cells, which are rare excitable, serotonergic neuroendocrine cells in the gut epithelium3-5. EC cells detect and transduce noxious stimuli to nearby mucosal nerve endings3,6 but involvement of this signalling pathway in visceral pain and attendant sex differences has not been assessed. By enhancing or suppressing EC cell function in vivo, we show that these cells are sufficient to elicit hypersensitivity to gut distension and necessary for the sensitizing actions of isovalerate, a bacterial short-chain fatty acid associated with GI inflammation7,8. Remarkably, prolonged EC cell activation produced persistent visceral hypersensitivity, even in the absence of an instigating inflammatory episode. Furthermore, perturbing EC cell activity promoted anxiety-like behaviours which normalized after blockade of serotonergic signalling. Sex differences were noted across a range of paradigms, indicating that the EC cell-mucosal afferent circuit is tonically engaged in females. Our findings validate a critical role for EC cell-mucosal afferent signalling in acute and persistent GI pain, in addition to highlighting genetic models for studying visceral hypersensitivity and the sex bias of gut pain.
Collapse
Affiliation(s)
- James R Bayrer
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA.
| | - Joel Castro
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Archana Venkataraman
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
| | - Kouki K Touhara
- Department of Physiology, University of California, San Francisco, CA, USA
| | - Nathan D Rossen
- Department of Physiology, University of California, San Francisco, CA, USA
- Tetrad Graduate Program, University of California, San Francisco, CA, USA
| | - Ryan D Morrie
- Department of Physiology, University of California, San Francisco, CA, USA
- Maze Therapeutics, San Francisco, CA, USA
| | - Jessica Maddern
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Aenea Hendry
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Kristina N Braverman
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Jansen, Johnson & Johnson, San Diego, CA, USA
| | - Sonia Garcia-Caraballo
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Gudrun Schober
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Mariana Brizuela
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | | | - Carla Bueno-Silva
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Holly A Ingraham
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.
| | - Stuart M Brierley
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia.
- Hopwood Centre for Neurobiology, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia.
| | - David Julius
- Department of Physiology, University of California, San Francisco, CA, USA.
| |
Collapse
|
13
|
Osman S, Tashtush A, Reed DE, Lomax AE. Analysis of the spinal and vagal afferent innervation of the mouse colon using neuronal retrograde tracers. Cell Tissue Res 2023:10.1007/s00441-023-03769-3. [PMID: 37004577 DOI: 10.1007/s00441-023-03769-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/24/2023] [Indexed: 04/04/2023]
Abstract
The gut-brain axis has received increasing attention recently due to evidence that colonic microbes can affect brain function and behavior. However, little is known about the innervation of the colon by a major component of the gut-brain axis, vagal afferent neurons. Furthermore, it is currently unknown whether individual NG neurons or DRG neurons innervate both the proximal and distal colon. We aimed to quantify the number of vagal and spinal afferent neurons that innervate the colon; and determine whether these individual neurons simultaneously innervate the mouse proximal and distal colon. C57Bl/6 mice received injections of a combination of retrograde tracers that were either injected into the muscularis externa of the proximal or the distal colon: fast blue, DiI and DiO. Five to seven percent of lumbosacral and thoracolumbar spinal afferent neurons, and 25% of vagal afferent neurons were labelled by injections of DiI and DiO into the colon. We also found that approximately 8% of NG neurons innervate the distal colon. Ten percent of labeled thoracolumbar and 15% of labeled lumbosacral DRG neurons innervate both the distal and proximal colon. Eighteen percent of labeled NG neurons innervated both the distal and proximal colon. In conclusion, vagal afferent innervation of the distal colon is less extensive than the proximal colon, whereas a similar gradient was not observed for the spinal afferent innervation. Furthermore, overlap appears to exist between the receptive fields of vagal and spinal afferent neurons that innervate the proximal and distal colon.
Collapse
Affiliation(s)
- Samira Osman
- Gastrointestinal Diseases Research Unit, Queen's University, 76 Stuart Street, Kingston, ON, K7L 2V7, Canada
| | - Ayssar Tashtush
- Gastrointestinal Diseases Research Unit, Queen's University, 76 Stuart Street, Kingston, ON, K7L 2V7, Canada
- Department of Physiology, Jordan University of Science and Technology, Irbid, Jordan
| | - David E Reed
- Gastrointestinal Diseases Research Unit, Queen's University, 76 Stuart Street, Kingston, ON, K7L 2V7, Canada
- Department of Medicine, Queen's University, ON, Kingston, Canada
| | - Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University, 76 Stuart Street, Kingston, ON, K7L 2V7, Canada.
- Department of Medicine, Queen's University, ON, Kingston, Canada.
- Department of Biomedical and Molecular Sciences, Queen's University, ON, Kingston, Canada.
| |
Collapse
|
14
|
Ramsay S, Zagorodnyuk V. Role of circadian rhythms and melatonin in bladder function in heath and diseases. Auton Neurosci 2023; 246:103083. [PMID: 36871511 DOI: 10.1016/j.autneu.2023.103083] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/23/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023]
Abstract
The circadian system modulates all visceral organ physiological processes including urine storage and voiding. The "master clock" of the circadian system lies within suprachiasmatic nucleus of the hypothalamus while "peripheral clocks" are found in most peripheral tissue and organs, including the urinary bladder. Disruptions of circadian rhythms can cause organ malfunction and disorder or exacerbate pre-existing ones. It has been suggested that nocturia, which develops mostly in the elderly, could be a circadian-related disorder of the bladder. In the bladder, many types of gap junctions and ion channels in the detrusor, urothelium and sensory nerves are likely under strict local peripheral circadian control. The pineal hormone, melatonin, is a circadian rhythm synchroniser capable of controlling a variety of physiological processes in the body. Melatonin predominantly acts via the melatonin 1 and melatonin 2 G-protein coupled receptors expressed in the central nervous system, and many peripheral organs and tissues. Melatonin could be beneficial in the treatment of nocturia and other common bladder disorders. The ameliorating action of melatonin on bladder function is likely due to multiple mechanisms which include central effects on voiding and peripheral effects on the detrusor and bladder afferents. More studies are warranted to determine the precise mechanisms of circadian rhythm coordination of the bladder function and melatonin influences on the bladder in health and diseases.
Collapse
Affiliation(s)
- Stewart Ramsay
- Discipline of Human Physiology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, South Australia, Australia
| | - Vladimir Zagorodnyuk
- Discipline of Human Physiology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, South Australia, Australia.
| |
Collapse
|
15
|
Qi L, Lin SH, Ma Q. Spinal VGLUT3 lineage neurons drive visceral mechanical allodynia but not sensitized visceromotor reflexes. Neuron 2023; 111:669-681.e5. [PMID: 36584681 DOI: 10.1016/j.neuron.2022.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 09/08/2022] [Accepted: 11/30/2022] [Indexed: 12/30/2022]
Abstract
Visceral pain is among the most prevalent and bothersome forms of chronic pain, but their transmission in the spinal cord is still poorly understood. Here, we conducted focal colorectal distention (fCRD) to drive both visceromotor responses (VMRs) and aversion. We first found that spinal CCK neurons were necessary for noxious fCRD to drive both VMRs and aversion under naive conditions. We next showed that spinal VGLUT3 neurons mediate visceral allodynia, whose ablation caused loss of aversion evoked by low-intensity fCRD in mice with gastrointestinal (GI) inflammation or spinal circuit disinhibition. Importantly, these neurons were dispensable for driving sensitized VMRs under both inflammatory and central disinhibition conditions. Anatomically, a subset of VGLUT3 neurons projected to parabrachial nuclei, whose photoactivation sufficiently generated aversion in mice with GI inflammation, without influencing VMRs. Our studies suggest the presence of different spinal substrates that transmit nociceptive versus affective dimensions of visceral sensory information.
Collapse
Affiliation(s)
- Lu Qi
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Shing-Hong Lin
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Qiufu Ma
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
16
|
Xu J, McGinnis A, Ji RR. Piezo2 mediates visceral mechanosensation: A new therapeutic target for gut pain? Neuron 2023; 111:450-452. [PMID: 36796326 DOI: 10.1016/j.neuron.2023.01.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Mechanical distension/stretch in the colon provokes visceral hypersensitivity and pain. In this issue of Neuron, Xie et al. report that mechanosensitive Piezo2 channels, expressed by TRPV1-lineage nociceptors, are involved in visceral mechanical nociception and hypersensitivity.
Collapse
Affiliation(s)
- Jing Xu
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Aidan McGinnis
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
17
|
Gaither TW, Vincent NW, Piqueiras E, Barzallo D, Siapno A, Williams KC, Russell M, Litwin MS. Atlas of the receptive anal sex experience among people with prostates. J Sex Med 2023; 20:126-138. [PMID: 36763914 DOI: 10.1093/jsxmed/qdac024] [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: 06/03/2022] [Revised: 09/15/2022] [Accepted: 11/21/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND Receptive anal intercourse (RAI) is commonly practiced among individuals of all sexual orientations. However, negative stigmatization by society and health care professionals leads to the underreporting or this practice. AIM We sought to assess and describe the subjective role of the prostate as a pleasure center in participants with diverse RAI experiences. The secondary aim was to describe nonprostatic areas within the anorectal region that produce erotic sensation and/or pain. METHODS The exploratory sequential multimethod study design included focus groups and semistructured interviews with 30 individuals with prostates who had engaged in RAI. We used graphic elicitation of natal male anatomy to enhance visualization and assess participant perspectives. OUTCOMES The main outcome of interest was the identification of anatomic locations of erogenous sensation and pain during RAI. RESULTS Among the participants (median age 38, range 24-77 years), most participants (90%) identified as cisgender male. Three major themes emerged within the motivations for RAI, including (1) deriving intrinsic pleasure, (2) providing both pleasure for a partner and a way to improve intimacy/connection, and (3) an inability to be the insertive partner due to physical or mental challenges. The data suggest that the anorectal region produces a variety of erogenous sensations which participants find pleasurable. Overall, 2 major areas of erogenous sensation occur along the anterior rectal wall and within the anus. Within the context of RAI, 2 distinct categories of pain emerged, including pain with insertion and pain at other times. CLINICAL IMPLICATIONS Understanding where erogenous sensation originates for each individual may predict sexual functioning after various surgical interventions. Timing and location of pain may aid in further characterizing anodyspareunia. STRENGTHS AND LIMITATIONS Our study utilized a sequential design (from focus groups to interviews) with diverse RAI experiences, especially regarding age, geographic location, and prostate pathology. We included individuals of diverse gender identities, but too few to evaluate these groups independently from cisgender men. CONCLUSION People with prostates experience pleasure in multiple areas during RAI. Contrary to some lay literature, the prostate region is not the subjective pleasure center for all individuals. Timing and location of pain during RAI may inform areas for intervention. Providing a language for pleasure and pain during RAI may improve communication between not only sexual partners but also clinicians and patients.
Collapse
Affiliation(s)
- Thomas W Gaither
- Department of Urology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | | | - Eduardo Piqueiras
- Department of Urology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Devin Barzallo
- Case Western Reserve School of Medicine, Cleveland, OH, United States
| | - Allen Siapno
- Department of Urology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Kristen C Williams
- Department of Urology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Marcia Russell
- Department of Surgery, Section of Colorectal Surgery, David Geffen School of Medicine, Los Angeles, United States, CA.,Surgical and Perioperative Careline, VA Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Mark S Litwin
- Department of Urology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States.,Department of Health Policy and Management, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, United States.,School of Nursing, University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
18
|
Muacevic A, Adler JR. Acute Colonic Pseudo-Obstruction Secondary to Renal Calculus: A Case Report and Review of Pathophysiology. Cureus 2023; 15:e34756. [PMID: 36777972 PMCID: PMC9905947 DOI: 10.7759/cureus.34756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Acute colonic pseudo-obstruction (ACPO) is obstruction of the large bowel without a mechanical cause. The exact mechanism remains incompletely understood but is thought to result from disruption to the autonomic regulation of the colon, typically in the context of hospitalized patients with medical illness, precipitating medications, or recent surgical intervention. This paper presents an unusual case of ACPO in an ambulatory patient with a recently passed renal calculus, explores the anatomy and physiology underlying the autonomic dysfunction theory of ACPO pathogenesis in the context of the case, and provides a 3D reconstruction of the patient's CT to illustrate the abrupt caliber change at the splenic flexure characteristic of ACPO.
Collapse
|
19
|
Spencer NJ, Hibberd T, Xie Z, Hu H. How should we define a nociceptor in the gut-brain axis? Front Neurosci 2022; 16:1096405. [PMID: 36601592 PMCID: PMC9806170 DOI: 10.3389/fnins.2022.1096405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
In the past few years, there has been extraordinary interest in how the gut communicates with the brain. This is because substantial and gathering data has emerged to suggest that sensory nerve pathways between the gut and brain may contribute much more widely in heath and disease, than was originally presumed. In the skin, the different types of sensory nerve endings have been thoroughly characterized, including the morphology of different nerve endings and the sensory modalities they encode. This knowledge is lacking for most types of visceral afferents, particularly spinal afferents that innervate abdominal organs, like the gut. In fact, only recently have the nerve endings of spinal afferents in any visceral organ been identified. What is clear is that spinal afferents play the major role in pain perception from the gut to the brain. Perhaps surprisingly, the majority of spinal afferent nerve endings in the gut express the ion channel TRPV1, which is often considered to be a marker of "nociceptive" neurons. And, a majority of gut-projecting spinal afferent neurons expressing TRPV1 are activated at low thresholds, in the "normal" physiological range, well below the normal threshold for detection of painful sensations. This introduces a major conundrum regarding visceral nociception. How should we define a "nociceptor" in the gut? We discuss the notion that nociception from the gut wall maybe a process encrypted into multiple different morphological types of spinal afferent nerve ending, rather than a single class of sensory ending, like free-endings, suggested to underlie nociception in skin.
Collapse
Affiliation(s)
- Nick J. Spencer
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia,*Correspondence: Nick J. Spencer,
| | - Tim Hibberd
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| | - Zili Xie
- Department of Anesthesiology, The Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - Hongzhen Hu
- Department of Anesthesiology, The Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
20
|
Tuck CJ, Abu Omar A, De Palma G, Osman S, Jiménez-Vargas NN, Yu Y, Bennet SM, Lopez-Lopez C, Jaramillo-Polanco JO, Baker CC, Bennett AS, Guzman-Rodriguez M, Tsang Q, Alward T, Rolland S, Morissette C, Verdu EF, Bercik P, Vanner SJ, Lomax AE, Reed DE. Changes in signalling from faecal neuroactive metabolites following dietary modulation of IBS pain. Gut 2022; 72:gutjnl-2022-327260. [PMID: 36591617 DOI: 10.1136/gutjnl-2022-327260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 11/23/2022] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Dietary therapies for irritable bowel syndrome (IBS) have received increasing interest but predicting which patients will benefit remains a challenge due to a lack of mechanistic insight. We recently found evidence of a role for the microbiota in dietary modulation of pain signalling in a humanised mouse model of IBS. This randomised cross-over study aimed to test the hypothesis that pain relief following reduced consumption of fermentable carbohydrates is the result of changes in luminal neuroactive metabolites. DESIGN IBS (Rome IV) participants underwent four trial periods: two non-intervention periods, followed by a diet low (LFD) and high in fermentable carbohydrates for 3 weeks each. At the end of each period, participants completed questionnaires and provided stool. The effects of faecal supernatants (FS) collected before (IBS FS) and after a LFD (LFD FS) on nociceptive afferent neurons were assessed in mice using patch-clamp and ex vivo colonic afferent nerve recording techniques. RESULTS Total IBS symptom severity score and abdominal pain were reduced by the LFD (N=25; p<0.01). Excitability of neurons was increased in response to IBS FS, but this effect was reduced (p<0.01) with LFD FS from pain-responders. IBS FS from pain-responders increased mechanosensitivity of nociceptive afferent nerve axons (p<0.001), an effect lost following LFD FS administration (p=NS) or when IBS FS was administered in the presence of antagonists of histamine receptors or protease inhibitors. CONCLUSIONS In a subset of IBS patients with improvement in abdominal pain following a LFD, there is a decrease in pronociceptive signalling from FS, suggesting that changes in luminal mediators may contribute to symptom response.
Collapse
Affiliation(s)
- Caroline J Tuck
- Department of Sport, Exercise and Nutrition Sciences, La Trobe University, Melbourne, Victoria, Australia
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Amal Abu Omar
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
- Department of Physiology, Jordan University of Science and Technology, Irbid, Jordan
| | - Giada De Palma
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Samira Osman
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | | | - Yang Yu
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Sean Mp Bennet
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Cintya Lopez-Lopez
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | | | - Corey C Baker
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Aidan Sw Bennett
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | | | - Quentin Tsang
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Taylor Alward
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Sebastien Rolland
- Department of Medicine, Hopital Maisonneuve-Rosemont, Montreal, Québec, Canada
| | - Celine Morissette
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Elena F Verdu
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Premysl Bercik
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Stephen J Vanner
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - David E Reed
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
21
|
Nishida K, Matsumura S, Kobayashi T. Involvement of Brn3a-positive spinal dorsal horn neurons in the transmission of visceral pain in inflammatory bowel disease model mice. FRONTIERS IN PAIN RESEARCH (LAUSANNE, SWITZERLAND) 2022; 3:979038. [PMID: 36570085 PMCID: PMC9768036 DOI: 10.3389/fpain.2022.979038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/16/2022] [Indexed: 12/12/2022]
Abstract
The spinal dorsal horn plays a crucial role in the transmission and processing of somatosensory information. Although spinal neural circuits that process several distinct types of somatic sensations have been studied extensively, those responsible for visceral pain transmission remain poorly understood. In the present study, we analyzed dextran sodium sulfate (DSS)-induced inflammatory bowel disease (IBD) mouse models to characterize the spinal dorsal horn neurons involved in visceral pain transmission. Immunostaining for c-fos, a marker of neuronal activity, demonstrated that numerous c-fos-positive cells were found bilaterally in the lumbosacral spinal dorsal horn, and their distribution was particularly abundant in the shallow dorsal horn. Characterization of these neurons by several molecular markers revealed that the percentage of the Pit1-Oct1-Unc86 domain (POU domain)-containing transcription factor Brn3a-positive neurons among the c-fos-positive neurons in the shallow dorsal horn was 30%-40% in DSS-treated mice, which was significantly higher than that in the somatic pain model mice. We further demonstrated by neuronal tracing that, within the shallow dorsal horn, Brn3a-positive neurons were more highly represented in spino-solitary projection neurons than in spino-parabrachial projection neurons. These results raise the possibility that Brn3a-positive spinal dorsal horn neurons make a large contribution to visceral pain transmission, part of which is mediated through the spino-solitary pathway.
Collapse
|
22
|
Dodds KN, Kyloh MA, Travis L, Cox M, Hibberd TJ, Spencer NJ. Anatomical distribution of CGRP-containing lumbosacral spinal afferent neurons in the mouse uterine horn. Front Neurosci 2022; 16:1012329. [PMID: 36248657 PMCID: PMC9554138 DOI: 10.3389/fnins.2022.1012329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Sensory stimuli from the uterus are detected by spinal afferent neurons whose cell bodies arise from thoracolumbar and lumbosacral dorsal root ganglia (DRG). Using an in vivo survival surgical technique developed in our laboratory to remove select DRG from live mice, we recently quantified the topographical distribution of thoracolumbar spinal afferents innervating the mouse uterine horn, revealed by loss of immunoreactivity to calcitonin gene-related peptide (CGRP). Here, we used the same technique to investigate the distribution of lumbosacral uterine spinal afferents, in which L5-S1 DRG were unilaterally removed from adult female C57BL/6J mice (N = 6). Following 10–12 days recovery, CGRP immunoreactivity was quantified along the length of uterine horns using fluorescence immunohistochemistry. Relative to myometrial thickness, overall CGRP density in uterine tissues ipsilateral to L5-S1 DRG removal was reduced compared to the DRG-intact, contralateral side (P = 0.0265). Regionally, however, myometrial CGRP density was unchanged in the cranial, mid, and caudal portions. Similarly, CGRP-expressing nerve fiber counts, network lengths, junctions, and the proportion of area occupied by CGRP immunoreactivity were unaffected by DRG removal (P ≥ 0.2438). Retrograde neuronal tracing from the caudal uterine horn revealed fewer spinal afferents here arise from lumbosacral than thoracolumbar DRG (P = 0.0442) (N = 4). These data indicate that, unlike thoracolumbar DRG, lumbosacral spinal afferent nerves supply relatively modest sensory innervation across the mouse uterine horn, with no regional specificity. We conclude most sensory information between the mouse uterine horn and central nervous system is likely relayed via thoracolumbar spinal afferents.
Collapse
|
23
|
Kyloh MA, Hibberd TJ, Castro J, Harrington AM, Travis L, Dodds KN, Wiklendt L, Brierley SM, Zagorodnyuk VP, Spencer NJ. Disengaging spinal afferent nerve communication with the brain in live mice. Commun Biol 2022; 5:915. [PMID: 36104503 PMCID: PMC9475039 DOI: 10.1038/s42003-022-03876-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 08/23/2022] [Indexed: 11/24/2022] Open
Abstract
Our understanding of how abdominal organs (like the gut) communicate with the brain, via sensory nerves, has been limited by a lack of techniques to selectively activate or inhibit populations of spinal primary afferent neurons within dorsal root ganglia (DRG), of live animals. We report a survival surgery technique in mice, where select DRG are surgically removed (unilaterally or bilaterally), without interfering with other sensory or motor nerves. Using this approach, pain responses evoked by rectal distension were abolished by bilateral lumbosacral L5-S1 DRG removal, but not thoracolumbar T13-L1 DRG removal. However, animals lacking T13-L1 or L5-S1 DRG both showed reduced pain sensitivity to distal colonic distension. Removal of DRG led to selective loss of peripheral CGRP-expressing spinal afferent axons innervating visceral organs, arising from discrete spinal segments. This method thus allows spinal segment-specific determination of sensory pathway functions in conscious, free-to-move animals, without genetic modification. A surgical method in mice can selectively remove dorsal root ganglia (DRG) at specific spinal levels without interfering with other nerves, providing insight on thoracolumbar vs. lumbosacral DRG contributions to pain signalling and behaviour.
Collapse
|
24
|
Balogh M, Zhang J, Gaffney CM, Kalakuntla N, Nguyen NT, Trinh RT, Aguilar C, Pham HV, Milutinovic B, Nichols JM, Mahalingam R, Shepherd AJ. Sensory neuron dysfunction in orthotopic mouse models of colon cancer. J Neuroinflammation 2022; 19:204. [PMID: 35962398 PMCID: PMC9375288 DOI: 10.1186/s12974-022-02566-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/07/2022] [Indexed: 11/10/2022] Open
Abstract
Reports of neurological sequelae related to colon cancer are largely restricted to rare instances of paraneoplastic syndromes, due to autoimmune reactions. Systemic inflammation associated with tumor development influences sensory neuron function in other disease models, though the extent to which this occurs in colorectal cancer is unknown. We induced orthotopic colorectal cancer via orthotopic injection of two colorectal cancer cell lines (MC38 and CT26) in two different mouse strains (C57BL/6 and Balb/c, respectively). Behavioral tests of pain sensitivity and activity did not detect significant alterations in sensory sensitivity or diminished well-being throughout tumor development. However, immunohistochemistry revealed widespread reductions in intraepidermal nerve fiber density in the skin of tumor-bearing mice. Though loss of nerve fiber density was not associated with increased expression of cell injury markers in dorsal root ganglia, lumbar dorsal root ganglia neurons of tumor-bearing animals showed deficits in mitochondrial function. These neurons also had reduced cytosolic calcium levels in live-cell imaging and reduced spontaneous activity in multi-electrode array analysis. Bulk RNA sequencing of DRGs from tumor-bearing mice detected activation of gene expression pathways associated with elevated cytokine and chemokine signaling, including CXCL10. This is consistent with the detection of CXCL10 (and numerous other cytokines, chemokines and growth factors) in MC38 and CT26 cell-conditioned media, and the serum of tumor-bearing mice. Our study demonstrates in a pre-clinical setting that colon cancer is associated with latent sensory neuron dysfunction and implicates cytokine/chemokine signaling in this process. These findings may have implications for determining risk factors and treatment responsiveness related to neuropathy in colorectal cancer.
Collapse
Affiliation(s)
- Mihály Balogh
- The MD Anderson Pain Research Consortium and the Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, 9700 AD, Groningen, The Netherlands
| | - Jixiang Zhang
- The MD Anderson Pain Research Consortium and the Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caitlyn M Gaffney
- The MD Anderson Pain Research Consortium and the Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neha Kalakuntla
- The MD Anderson Pain Research Consortium and the Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas T Nguyen
- The MD Anderson Pain Research Consortium and the Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate School of Biomedical Sciences, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ronnie T Trinh
- The MD Anderson Pain Research Consortium and the Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Clarissa Aguilar
- The MD Anderson Pain Research Consortium and the Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - Hoang Vu Pham
- The MD Anderson Pain Research Consortium and the Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bojana Milutinovic
- The MD Anderson Pain Research Consortium and the Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Neurosurgery, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James M Nichols
- The MD Anderson Pain Research Consortium and the Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rajasekaran Mahalingam
- The MD Anderson Pain Research Consortium and the Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew J Shepherd
- The MD Anderson Pain Research Consortium and the Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
25
|
Verkuijl SJ, Trzpis M, Broens PMA. The anorectal defaecation reflex: a prospective intervention study. Colorectal Dis 2022; 24:845-853. [PMID: 35194918 PMCID: PMC9541108 DOI: 10.1111/codi.16101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/30/2022] [Accepted: 02/14/2022] [Indexed: 12/13/2022]
Abstract
AIM Our hypothesis is that there may be a neural pathway with sensory afferent neurons in the anal canal that leads to rectal contraction to assist defaecation. We aimed to compare rectal motility between healthy participants with or without anal anaesthesia. METHOD This prospective intervention study consisted of two test sessions: a baseline session followed by an identical second session. During each session we performed the anal electrosensitivity test, the rectoanal inhibitory reflex test and rapid phasic barostat distensions. Prior to the second session, participants were randomly assigned to receive either a local anal anaesthetic or a placebo. RESULTS We included 23 healthy participants aged 21.1 ± 0.5 years, 13 of whom received an anal anaesthetic and 10 a placebo. All participants showed a transient rectal contraction during the first test session, which decreased significantly after anal anaesthesia (18.6 ml vs. 4.9 ml, p = 0.019). The maximum rectal contraction was comparable to the baseline results in the placebo group. Furthermore, the electrosensitivity at the highest centimetre of the anal canal correlated with the maximum rectal contraction (r = -0.452, p = 0.045). CONCLUSION All healthy study participants display an involuntary, reproducible rectal reflex contraction that appears to be innervated by afferent nerves in the proximal anal canal. The rectal reflex contraction appears to play a role in defaecation and we therefore refer to this phenomenon as the anorectal defaecation reflex. Knowledge of the anorectal defaecation reflex may have consequences for the diagnostics and treatment of constipation.
Collapse
Affiliation(s)
- Sanne J. Verkuijl
- Department of SurgeryAnorectal Physiology LaboratoryUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands,Department of SurgeryDivision of Paediatric SurgeryUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Monika Trzpis
- Department of SurgeryAnorectal Physiology LaboratoryUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Paul M. A. Broens
- Department of SurgeryAnorectal Physiology LaboratoryUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands,Department of SurgeryDivision of Paediatric SurgeryUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
26
|
Balik V, Šulla I. Autonomic Dysreflexia following Spinal Cord Injury. Asian J Neurosurg 2022; 17:165-172. [PMID: 36120615 PMCID: PMC9473833 DOI: 10.1055/s-0042-1751080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
AbstractAutonomic dysreflexia (AD) is a potentially life-threatening condition of the autonomic nervous system following spinal cord injury at or above T6. One of the most common symptoms is a sudden increase in blood pressure induced by afferent sensory stimulation owing to unmodulated reflex sympathetic hyperactivity. Such episodes of high blood pressure might be associated with a high risk of cerebral or retinal hemorrhage, seizures, heart failure, or pulmonary edema. In-depth knowledge is, therefore, crucial for the proper management of the AD, especially for spine surgeons, who encounter these patients quite often in their clinical practice. Systematical review of the literature dealing with strategies to prevent and manage this challenging condition was done by two independent reviewers. Studies that failed to assess primary (prevention, treatment strategies and management) and secondary outcomes (clinical symptomatology, presentation) were excluded. A bibliographical search revealed 85 eligible studies that provide a variety of preventive and treatment measures for the subjects affected by AD. As these measures are predominantly based on noncontrolled trials, long-term prospectively controlled multicenter studies are warranted to validate these preventive and therapeutic proposals.
Collapse
Affiliation(s)
- Vladimír Balik
- Department of Neurosurgery, Svet Zdravia Hospital, Michalovce, Slovakia
| | - Igor Šulla
- Department of Anatomy, University of Veterinary Medicine and Pharmacy, Histology and Physiology, Košice, Slovakia
| |
Collapse
|
27
|
Dodds KN, Travis L, Kyloh MA, Jones LA, Keating DJ, Spencer NJ. The gut-brain axis: spatial relationship between spinal afferent nerves and 5-HT-containing enterochromaffin cells in mucosa of mouse colon. Am J Physiol Gastrointest Liver Physiol 2022; 322:G523-G533. [PMID: 35293258 DOI: 10.1152/ajpgi.00019.2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cross talk between the gastrointestinal tract and brain is of significant relevance for human health and disease. However, our understanding of how the gut and brain communicate has been limited by a lack of techniques to identify the precise spatial relationship between extrinsic nerve endings and their proximity to specific cell types that line the inner surface of the gastrointestinal tract. We used an in vivo anterograde tracing technique, previously developed in our laboratory, to selectively label single spinal afferent axons and their nerve endings in mouse colonic mucosa. The closest three-dimensional distances between spinal afferent nerve endings and axonal varicosities to enterochromaffin (EC) cells, which contain serotonin (5-hydroxytryptamine; 5-HT), were then measured. The mean distances (± standard deviation) between any varicosity along a spinal afferent axon or its nerve ending, and the nearest EC cell, were 5.7 ± 6.0 μm (median: 3.6 μm) and 26.9 ± 18.6 μm (median: 24.1 μm), respectively. Randomization of the spatial location of EC cells revealed similar results to this actual data. These distances are ∼200-1,000 times greater than those between pre- and postsynaptic membranes (15-25 nm) that underlie synaptic transmission in the vertebrate nervous system. Our findings suggest that colonic 5-HT-containing EC cells release substances to activate centrally projecting spinal afferent nerves likely via diffusion, as such signaling is unlikely to occur with the spatial fidelity of a synapse.NEW & NOTEWORTHY We show an absence of close physical contact between spinal afferent nerves and 5-HT-containing EC cells in mouse colonic mucosa. Similar relative distances were observed between randomized EC cells and spinal afferents compared with actual data. This spatial relationship suggests that substances released from colonic 5-HT-containing EC cells are unlikely to act via synaptic transmission to neighboring spinal afferents that relay sensory information from the gut lumen to the brain.
Collapse
Affiliation(s)
- Kelsi N Dodds
- Visceral Neurophysiology Laboratory, Flinders Health and Medical Research Institute & College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, South Australia, Australia
| | - Lee Travis
- Visceral Neurophysiology Laboratory, Flinders Health and Medical Research Institute & College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, South Australia, Australia
| | - Melinda A Kyloh
- Visceral Neurophysiology Laboratory, Flinders Health and Medical Research Institute & College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, South Australia, Australia
| | - Lauren A Jones
- Molecular & Cellular Physiology Laboratory, Flinders Health and Medical Research Institute & College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, South Australia, Australia
| | - Damien J Keating
- Molecular & Cellular Physiology Laboratory, Flinders Health and Medical Research Institute & College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, South Australia, Australia
| | - Nick J Spencer
- Visceral Neurophysiology Laboratory, Flinders Health and Medical Research Institute & College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, South Australia, Australia
| |
Collapse
|
28
|
Najjar S, Ahn S, Umrau K, Erdem Arslan M, Jennings TA, Whyte C, Lee H. Increasing Trend of Calretinin-Positive Mucosal Innervation from Aganglionic Zone toward Transition Zone in Hirschsprung's Disease. Eur J Pediatr Surg 2022; 32:191-197. [PMID: 33513627 DOI: 10.1055/s-0041-1722859] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Diagnosis of very short-segment Hirschsprung's disease (vsHD) by rectal suction biopsy is challenging as its aganglionic zone (AZ) overlaps with physiologic hypoganglionic zone and calretinin-positive mucosal nerves may extend from the transition zone (TZ) into AZ. We studied whether an increasing trend/gradient of calretinin-positive mucosal nerves along the distance from AZ toward TZ aids in diagnosis of HD. MATERIALS AND METHODS In this study, 46 rectal suction biopsies from non-HD and HD, and 15 pull-through specimens from short-segment HD were evaluated by mucosal calretinin immunostain (CI) and image processing and analysis (IPA) to measure pixel count (PC, the percentage of calretinin stained pixels in the mucosa). Consecutive longitudinal sections of proximal AZ toward distal TZ in HD pull-through specimens were utilized as a vsHD surrogate model. First, we studied variability of mucosal CI in non-HD biopsies along the distance from dentate line. Second, we determined a cutoff point of mucosal CI by IPA that separated non-HD versus HD and applied this cutoff to longitudinal sections from proximal AZ to distal TZ segments in HD pull-through specimens. Third, we studied whether an increasing trend of mucosal CI was universally observed in HD pull-through. RESULTS Our findings included a significant variability in PC along the biopsy distance in non-HD cases. Positive mucosal CI was found in proximal AZ in 6 (43%) of 14 HD pull-through, among which 1 case lacked submucosal nerve hypertrophy in the proximal AZ. All 14 HD pull-through cases showed an increasing trend/gradient of PC from AZ toward TZ. CONCLUSION Based on our findings, the presence or absence of mucosal CI positivity and submucosal nerve hypertrophy may not reliably diagnose vsHD in rectal suction biopsy. While we acknowledge that the density of mucosal innervation in variable contexts and anatomical locations is unknown and yet to be explored, our study suggests that an increasing trend of positive mucosal CI from AZ toward TZ by IPA might prove to be a useful tool for the diagnosis of vsHD in the future.
Collapse
Affiliation(s)
- Saleh Najjar
- Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, New York, United States
| | - Sangtae Ahn
- GE Global Research, Niskayuna, New York, United States
| | - Kavita Umrau
- Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, New York, United States
| | - Mustafa Erdem Arslan
- Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, New York, United States
| | - Timothy A Jennings
- Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, New York, United States
| | - Christine Whyte
- Department of Pediatric Surgery, Albany Medical College, Albany, New York, United States
| | - Hwajeong Lee
- Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, United States
| |
Collapse
|
29
|
Brierley SM, Grundy L, Castro J, Harrington AM, Hannig G, Camilleri M. Guanylate cyclase-C agonists as peripherally acting treatments of chronic visceral pain. Trends Pharmacol Sci 2022; 43:110-122. [PMID: 34865885 PMCID: PMC8760167 DOI: 10.1016/j.tips.2021.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/28/2021] [Accepted: 11/02/2021] [Indexed: 02/03/2023]
Abstract
Irritable bowel syndrome (IBS) is a chronic gastrointestinal disorder characterized by abdominal pain and altered bowel habit that affects ~11% of the global population. Over the past decade, preclinical and clinical studies have revealed a variety of novel mechanisms relating to the visceral analgesic effects of guanylate cyclase-C (GC-C) agonists. Here we discuss the mechanisms by which GC-C agonists target the GC-C/cyclic guanosine-3',5'-monophosphate (cGMP) pathway, resulting in visceral analgesia as well as clinically relevant relief of abdominal pain and other sensations in IBS patients. Due to the preponderance of evidence we focus on linaclotide, a 14-amino acid GC-C agonist with very low oral bioavailability that acts within the gut. Collectively, the weight of experimental and clinical evidence supports the concept that GC-C agonists act as peripherally acting visceral analgesics.
Collapse
Affiliation(s)
- Stuart M. Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, 5042, AUSTRALIA.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, AUSTRALIA.,Discipline of Medicine, University of Adelaide, North Terrace, Adelaide, South Australia 5000, AUSTRALIA.,Corresponding Author: Prof. Stuart M. Brierley, Ph.D. Visceral Pain Research Group, Level 7, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, SA 5000, AUSTRALIA.
| | - Luke Grundy
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, 5042, AUSTRALIA.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, AUSTRALIA
| | - Joel Castro
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, 5042, AUSTRALIA.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, AUSTRALIA
| | - Andrea M. Harrington
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, 5042, AUSTRALIA.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, AUSTRALIA
| | | | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiologic Research Program, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
30
|
Neural signalling of gut mechanosensation in ingestive and digestive processes. Nat Rev Neurosci 2022; 23:135-156. [PMID: 34983992 DOI: 10.1038/s41583-021-00544-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2021] [Indexed: 12/29/2022]
Abstract
Eating and drinking generate sequential mechanosensory signals along the digestive tract. These signals are communicated to the brain for the timely initiation and regulation of diverse ingestive and digestive processes - ranging from appetite control and tactile perception to gut motility, digestive fluid secretion and defecation - that are vital for the proper intake, breakdown and absorption of nutrients and water. Gut mechanosensation has been investigated for over a century as a common pillar of energy, fluid and gastrointestinal homeostasis, and recent discoveries of specific mechanoreceptors, contributing ion channels and the well-defined circuits underlying gut mechanosensation signalling and function have further expanded our understanding of ingestive and digestive processes at the molecular and cellular levels. In this Review, we discuss our current understanding of the generation of mechanosensory signals from the digestive periphery, the neural afferent pathways that relay these signals to the brain and the neural circuit mechanisms that control ingestive and digestive processes, focusing on the four major digestive tract parts: the oral and pharyngeal cavities, oesophagus, stomach and intestines. We also discuss the clinical implications of gut mechanosensation in ingestive and digestive disorders.
Collapse
|
31
|
Meerschaert KA, Davis BM, Smith-Edwards KM. New Insights on Extrinsic Innervation of the Enteric Nervous System and Non-neuronal Cell Types That Influence Colon Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1383:133-139. [PMID: 36587153 DOI: 10.1007/978-3-031-05843-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The enteric nervous system not only innervates the colon to execute various functions in a semi-autonomous manner but also receives neural input from three extrinsic sources, (1) vagal, (2) thoracolumbar (splanchnic), and (3) lumbosacral (pelvic) pathways, that permit bidirectional communication between the colon and central nervous system. Extrinsic pathways signal sensory input via afferent fibers, as well as motor autonomic output via parasympathetic or sympathetic efferent fibers, but the shared and unique roles for each pathway in executing sensory-motor control of colon function have not been well understood. Here, we describe the recently developed approaches that have provided new insights into the diverse mechanisms utilized by extrinsic pathways to influence colon functions related to visceral sensation, motility, and inflammation. Based on the cumulative results from anatomical, molecular, and functional studies, we propose pathway-specific functions for vagal, thoracolumbar, and lumbosacral innervation of the colon.
Collapse
Affiliation(s)
| | - Brian M Davis
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | | |
Collapse
|
32
|
Defaye M, Abdullah NS, Iftinca M, Hassan A, Agosti F, Zhang Z, Cumenal M, Zamponi GW, Altier C. Gut-innervating TRPV1+ Neurons Drive Chronic Visceral Pain via Microglial P2Y12 Receptor. Cell Mol Gastroenterol Hepatol 2021; 13:977-999. [PMID: 34954381 PMCID: PMC8867057 DOI: 10.1016/j.jcmgh.2021.12.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 12/15/2021] [Accepted: 12/15/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS Chronic abdominal pain is a common symptom of inflammatory bowel diseases (IBDs). Peripheral and central mechanisms contribute to the transition from acute to chronic pain during active disease and clinical remission. Lower mechanical threshold and hyperexcitability of visceral afferents induce gliosis in central pain circuits, leading to persistent visceral hypersensitivity (VHS). In the spinal cord, microglia, the immune sentinels of the central nervous system, undergo activation in multiple models of VHS. Here, we investigated the mechanisms of microglia activation to identify centrally acting analgesics for chronic IBD pain. METHODS Using Designer Receptors Exclusively Activated by Designer Drugs (DREADD) expressed in transient receptor potential vanilloid member 1-expressing visceral neurons that sense colonic inflammation, we tested whether neuronal activity was indispensable to control microglia activation and VHS. We then investigated the neuron-microglia signaling system involved in visceral pain chronification. RESULTS We found that chemogenetic inhibition of transient receptor potential vanilloid member 1+ visceral afferents prevents microglial activation in the spinal cord and subsequent VHS in colitis mice. In contrast, chemogenetic activation, in the absence of colitis, enhanced microglial activation associated with VHS. We identified a purinergic signaling mechanism mediated by neuronal adenosine triphosphate (ATP) and microglial P2Y12 receptor, triggering VHS in colitis. Inhibition of P2RY12 prevented microglial reactivity and chronic VHS post-colitis. CONCLUSIONS Overall, these data provide novel insights into the central mechanisms of chronic visceral pain and suggest that targeting microglial P2RY12 signaling could be harnessed to relieve pain in patients with IBD who are in remission.
Collapse
Affiliation(s)
- Manon Defaye
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Inflammation Research Network-Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Nasser S. Abdullah
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Inflammation Research Network-Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Mircea Iftinca
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Inflammation Research Network-Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Ahmed Hassan
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Inflammation Research Network-Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Francina Agosti
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Inflammation Research Network-Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Zizhen Zhang
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Melissa Cumenal
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Inflammation Research Network-Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Gerald W. Zamponi
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Inflammation Research Network-Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada,Correspondence Address correspondence to: Christophe Altier, PhD, Associate Professor, Canada Research Chair in Inflammatory Pain, Department of Physiology & Pharmacology, Inflammation Research Network, Snyder Institute for Chronic Diseases, University of Calgary, HS 1665, 3330 Hospital Dr NW, Calgary, AB, T2N4N1 Canada. tel: (403) 220-7549.
| |
Collapse
|
33
|
Huizinga JD, Hussain A, Chen JH. Interstitial cells of Cajal and human colon motility in health and disease. Am J Physiol Gastrointest Liver Physiol 2021; 321:G552-G575. [PMID: 34612070 DOI: 10.1152/ajpgi.00264.2021] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Our understanding of human colonic motility, and autonomic reflexes that generate motor patterns, has increased markedly through high-resolution manometry. Details of the motor patterns are emerging related to frequency and propagation characteristics that allow linkage to interstitial cells of Cajal (ICC) networks. In studies on colonic motor dysfunction requiring surgery, ICC are almost always abnormal or significantly reduced. However, there are still gaps in our knowledge about the role of ICC in the control of colonic motility and there is little understanding of a mechanistic link between ICC abnormalities and colonic motor dysfunction. This review will outline the various ICC networks in the human colon and their proven and likely associations with the enteric and extrinsic autonomic nervous systems. Based on our extensive knowledge of the role of ICC in the control of gastrointestinal motility of animal models and the human stomach and small intestine, we propose how ICC networks are underlying the motor patterns of the human colon. The role of ICC will be reviewed in the autonomic neural reflexes that evoke essential motor patterns for transit and defecation. Mechanisms underlying ICC injury, maintenance, and repair will be discussed. Hypotheses are formulated as to how ICC dysfunction can lead to motor abnormalities in slow transit constipation, chronic idiopathic pseudo-obstruction, Hirschsprung's disease, fecal incontinence, diverticular disease, and inflammatory conditions. Recent studies on ICC repair after injury hold promise for future therapies.
Collapse
Affiliation(s)
- Jan D Huizinga
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Amer Hussain
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Ji-Hong Chen
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
34
|
Noor-Mohammadi E, Ligon CO, Mackenzie K, Stratton J, Shnider S, Greenwood-Van Meerveld B. A Monoclonal Anti-Calcitonin Gene-Related Peptide Antibody Decreases Stress-Induced Colonic Hypersensitivity. J Pharmacol Exp Ther 2021; 379:270-279. [PMID: 34620725 DOI: 10.1124/jpet.121.000731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/24/2021] [Indexed: 12/20/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a brain-gut disorder characterized by abdominal pain and altered bowel habits. Although the etiology of IBS remains unclear, stress in adulthood or in early life has been shown to be a significant factor in the development of IBS symptomatology. Evidence suggests that aberrant calcitonin gene-related peptide (CGRP) signaling may be involved in afferent sensitization and visceral organ hypersensitivity. Here, we used a monoclonal anti-CGRP divalent antigen-binding fragment [F(ab')2] antibody to test the hypothesis that inhibition of peripheral CGRP signaling reverses colonic hypersensitivity induced by either chronic adult stress or early life stress. A cohort of adult male rats was exposed to repeated water avoidance stress. Additionally, a second cohort consisting of female rats was exposed to a female-specific neonatal odor-attachment learning paradigm of unpredictable early life stress. Colonic sensitivity was then assessed in adult animals via behavioral responses to colorectal distension (CRD). To analyze spinal nociceptive signaling in response to CRD, dorsal horn extracellular signal-regulated kinase (ERK) 1/2 phosphorylation was measured via immunohistochemistry. Repeated psychologic stress in adulthood or unpredictable stress in early life induced colonic hypersensitivity and enhanced evoked ERK1/2 phosphorylation in the spinal cord after CRD in rats. These phenotypes were reversed by administration of a monoclonal anti-CGRP F(ab')2 fragment antibody. Stress-induced changes in visceral sensitivity and spinal nociceptive signaling were reversed by inhibition of peripheral CGRP signaling, which suggests a prominent role for CGRP in central sensitization and the development of stress-induced visceral hypersensitivity. SIGNIFICANCE STATEMENT: Targeting peripheral calcitonin gene-related peptide (CGRP) with a monoclonal anti-CGRP divalent antigen-binding fragment antibody reduced central sensitization and attenuated colonic hypersensitivity induced by either chronic adult stress or early life stress. CGRP-targeting antibodies are approved for migraine prevention, and the results of this study suggest that targeting CGRP may provide a novel treatment strategy for irritable bowel syndrome-related, stress-induced visceral pain.
Collapse
Affiliation(s)
- Ehsan Noor-Mohammadi
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma (E.N.-M., C.O.L., B.G.-V.M); and Teva Pharmaceutical Industries, Ltd., Redwood City, California (K.M., J.S., S.S.)
| | - Casey Owen Ligon
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma (E.N.-M., C.O.L., B.G.-V.M); and Teva Pharmaceutical Industries, Ltd., Redwood City, California (K.M., J.S., S.S.)
| | - Kimberly Mackenzie
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma (E.N.-M., C.O.L., B.G.-V.M); and Teva Pharmaceutical Industries, Ltd., Redwood City, California (K.M., J.S., S.S.)
| | - Jennifer Stratton
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma (E.N.-M., C.O.L., B.G.-V.M); and Teva Pharmaceutical Industries, Ltd., Redwood City, California (K.M., J.S., S.S.)
| | - Sara Shnider
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma (E.N.-M., C.O.L., B.G.-V.M); and Teva Pharmaceutical Industries, Ltd., Redwood City, California (K.M., J.S., S.S.)
| | - Beverley Greenwood-Van Meerveld
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma (E.N.-M., C.O.L., B.G.-V.M); and Teva Pharmaceutical Industries, Ltd., Redwood City, California (K.M., J.S., S.S.)
| |
Collapse
|
35
|
Bakshi N, Hart AL, Lee MC, Williams ACDC, Lackner JM, Norton C, Croft P. Chronic pain in patients with inflammatory bowel disease. Pain 2021; 162:2466-2471. [PMID: 34534174 PMCID: PMC8442739 DOI: 10.1097/j.pain.0000000000002304] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/19/2021] [Accepted: 03/31/2021] [Indexed: 12/19/2022]
Affiliation(s)
- Nikul Bakshi
- Research Department, Crohn's and Colitis UK, Hatfield, United Kingdom
| | | | - Michael C. Lee
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Amanda C de C. Williams
- Research Department of Clinical, Educational and Health Psychology, University College London, and Pain Management Centre, University College Hospitals NHS Foundation Trust, London, United Kingdom
| | - Jeffrey M. Lackner
- Division of Behavioral Medicine, Department of Medicine, University at Buffalo, SUNY, Buffalo, NY, United States
| | - Christine Norton
- Division of Care for Long-Term Conditions, Florence Nightingale Faculty of Nursing, Midwifery and Palliative Care, King's College, London, United Kingdom
| | - Peter Croft
- Primary Care Centre Versus Arthritis and Centre for Prognosis Research, Keele University, Keele, United Kingdom
| |
Collapse
|
36
|
Takahashi K, Khwaja IG, Schreyer JR, Bulmer D, Peiris M, Terai S, Aziz Q. Post-inflammatory Abdominal Pain in Patients with Inflammatory Bowel Disease During Remission: A Comprehensive Review. CROHN'S & COLITIS 360 2021; 3:otab073. [PMID: 36777266 PMCID: PMC9802269 DOI: 10.1093/crocol/otab073] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Indexed: 11/13/2022] Open
Abstract
Patients with inflammatory bowel disease often experience ongoing pain even after achieving mucosal healing (i.e., post-inflammatory pain). Factors related to the brain-gut axis, such as peripheral and central sensitization, altered sympatho-vagal balance, hypothalamic-pituitary-adrenal axis activation, and psychosocial factors, play a significant role in the development of post-inflammatory pain. A comprehensive study investigating the interaction between multiple predisposing factors, including clinical psycho-physiological phenotypes, molecular mechanisms, and multi-omics data, is still needed to fully understand the complex mechanism of post-inflammatory pain. Furthermore, current treatment options are limited and new treatments consistent with the underlying pathophysiology are needed to improve clinical outcomes.
Collapse
Affiliation(s)
- Kazuya Takahashi
- Centre for Neuroscience, Surgery and Trauma, Wingate Institute of Neurogastroenterology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Iman Geelani Khwaja
- Centre for Neuroscience, Surgery and Trauma, Wingate Institute of Neurogastroenterology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jocelyn Rachel Schreyer
- Centre for Neuroscience, Surgery and Trauma, Wingate Institute of Neurogastroenterology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - David Bulmer
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Madusha Peiris
- Centre for Neuroscience, Surgery and Trauma, Wingate Institute of Neurogastroenterology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Qasim Aziz
- Centre for Neuroscience, Surgery and Trauma, Wingate Institute of Neurogastroenterology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
37
|
Gershon MD, Margolis KG. The gut, its microbiome, and the brain: connections and communications. J Clin Invest 2021; 131:143768. [PMID: 34523615 PMCID: PMC8439601 DOI: 10.1172/jci143768] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Modern research on gastrointestinal behavior has revealed it to be a highly complex bidirectional process in which the gut sends signals to the brain, via spinal and vagal visceral afferent pathways, and receives sympathetic and parasympathetic inputs. Concomitantly, the enteric nervous system within the bowel, which contains intrinsic primary afferent neurons, interneurons, and motor neurons, also senses the enteric environment and controls the detailed patterns of intestinal motility and secretion. The vast microbiome that is resident within the enteric lumen is yet another contributor, not only to gut behavior, but to the bidirectional signaling process, so that the existence of a microbiota-gut-brain "connectome" has become apparent. The interaction between the microbiota, the bowel, and the brain now appears to be neither a top-down nor a bottom-up process. Instead, it is an ongoing, tripartite conversation, the outline of which is beginning to emerge and is the subject of this Review. We emphasize aspects of the exponentially increasing knowledge of the microbiota-gut-brain "connectome" and focus attention on the roles that serotonin, Toll-like receptors, and macrophages play in signaling as exemplars of potentially generalizable mechanisms.
Collapse
Affiliation(s)
| | - Kara Gross Margolis
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
38
|
Zhang L, Yang C, Li Y, Niu S, Liang X, Zhang Z, Luo Q, Luo H. Dynamic Changes in the Levels of Amyloid-β 42 Species in the Brain and Periphery of APP/PS1 Mice and Their Significance for Alzheimer's Disease. Front Mol Neurosci 2021; 14:723317. [PMID: 34512259 PMCID: PMC8430227 DOI: 10.3389/fnmol.2021.723317] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/09/2021] [Indexed: 11/13/2022] Open
Abstract
Although amyloid-β42 (Aβ42) has been used as one of the core biomarkers for Alzheimer’s disease (AD) diagnosis, the dynamic changes of its different forms in the brain, blood, and even intestines and its correlation with the progression of AD disease remain obscure. Herein, we screened Aβ42-specific preferred antibody pairs 1F12/1F12 and 1F12/2C6 to accurately detect Aβ42 types using sandwich ELISA, including total Aβ42, Aβ42 oligomers (Aβ42Os), and Aβ42 monomers (Aβ42Ms). The levels of Aβ42 species in the brain, blood, and intestines of different aged APP/PS1 mice were quantified to study their correlation with AD progression. Total Aβ42 levels in the blood were not correlated with AD progression, but Aβ42Ms level in the blood of 9-month-old APP/PS1 mice was significantly reduced, and Aβ42Os level in the brain was significantly elevated compared to 3-month-old APP/PS1, demonstrating that the levels of Aβ42Ms and Aβ42Os in the blood and brain were correlated with AD progression. Interestingly, in 9-month-old APP/PS1 mice, the level of Aβ42 in the intestine was higher than that in 3-month-old APP/PS1 mice, indicating that the increased level of Aβ42 in the gastrointestinal organs may also be related to the progression of AD. Meanwhile, changes in the gut microbiota composition of APP/PS1 mice with age were also observed. Therefore, the increase in Aβ derived from intestinal tissues and changes in microbiome composition can be used as a potential early diagnosis tool for AD, and further used as an indicator of drug intervention to reduce brain amyloid.
Collapse
Affiliation(s)
- Liding Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Changwen Yang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Yanqing Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Shiqi Niu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohan Liang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihong Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China.,School of Biomedical Engineering, Hainan University, Haikou, China
| | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China.,School of Biomedical Engineering, Hainan University, Haikou, China
| | - Haiming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
39
|
Trager RJ, Mok SR, Schlick KJ, Perez JA, Dusek JA. Association between radicular low back pain and constipation: a retrospective cohort study using a real-world national database. Pain Rep 2021; 6:e954. [PMID: 34476303 PMCID: PMC8397289 DOI: 10.1097/pr9.0000000000000954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/02/2021] [Accepted: 07/20/2021] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Previous observational studies have reported an association between lumbosacral radiculopathy (LSR), a form of low back pain (LBP) with nerve root involvement, and constipation. However, it is unclear whether this association is due to confounding variables such as comorbidities and medications. OBJECTIVES This study explores the possible association between LSR and constipation, with the hypothesis that adults with LSR have increased odds of developing constipation compared with those with nonradicular LBP. METHODS Adults aged 18 to 49 years with incident LSR and nonradicular LBP were identified from a national 70 million patient electronic health records network (TriNetX). Propensity score matching (PSM) was used to control for covariates and determine the odds ratio (OR) of constipation over a 1-year follow-up. Lumbar stenosis, cauda equina syndrome, and inflammatory bowel diseases were excluded. RESULTS After PSM, 503,062 patients were in each cohort. Before PSM, the likelihood of constipation was identical between cohorts (LSR 10.8% vs 10.9%; OR [confidence interval] = 0.99 [0.98-1.0], P = 0.251). This association was unchanged after PSM (LSR 10.8% vs 11.1%; OR [confidence interval] = 0.98 [0.97-0.99]; P = 0.003). CONCLUSIONS The study hypothesis can be refuted given that the OR approximated the null in a large propensity-matched sample. Patients with LSR have equivalent odds of constipation compared with those with nonradicular LBP, suggesting that LSR is not a direct cause of constipation. The similar risk of constipation between cohorts could be explained by factors common to LBP in general, such as pain severity, physical inactivity, and constipating medications.
Collapse
Affiliation(s)
- Robert James Trager
- Connor Integrative Health Network, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Shaffer R.S. Mok
- UH Digestive Health Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Kayla J. Schlick
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Jaime A. Perez
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Jeffery A. Dusek
- Connor Integrative Health Network, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| |
Collapse
|
40
|
Najjar SA, Albers KM. Pain in Inflammatory Bowel Disease: Optogenetic Strategies for Study of Neural-Epithelial Signaling. CROHN'S & COLITIS 360 2021; 3:otab040. [PMID: 34805983 PMCID: PMC8600958 DOI: 10.1093/crocol/otab040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Abdominal pain is common in patients with active inflammation of the colon but can persist even in its absence, suggesting other mechanisms of pain signaling. Recent findings suggest colon epithelial cells are direct regulators of pain-sensing neurons. Optogenetic activation of epithelial cells evoked nerve firing and pain-like behaviors. Inhibition of epithelial cells caused the opposite effect, reducing responses to colon distension and inflammatory hypersensitivity. Thus, epithelial cells alone can regulate the activation of pain circuits. Future goals are to define the anatomical and cellular mechanisms that underlie epithelial-neural pain signaling and how it is altered in response to colon inflammation.
Collapse
Affiliation(s)
- Sarah A Najjar
- Department of Neurobiology and Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Present address: Department of Pediatrics, Columbia University Medical Center, New York, New York, USA
| | - Kathryn M Albers
- Department of Neurobiology and Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Address correspondence to: Kathryn M. Albers, PhD, Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15216, USA ()
| |
Collapse
|
41
|
Borgmann D, Ciglieri E, Biglari N, Brandt C, Cremer AL, Backes H, Tittgemeyer M, Wunderlich FT, Brüning JC, Fenselau H. Gut-brain communication by distinct sensory neurons differently controls feeding and glucose metabolism. Cell Metab 2021; 33:1466-1482.e7. [PMID: 34043943 PMCID: PMC8280952 DOI: 10.1016/j.cmet.2021.05.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 02/23/2021] [Accepted: 05/03/2021] [Indexed: 01/26/2023]
Abstract
Sensory neurons relay gut-derived signals to the brain, yet the molecular and functional organization of distinct populations remains unclear. Here, we employed intersectional genetic manipulations to probe the feeding and glucoregulatory function of distinct sensory neurons. We reconstruct the gut innervation patterns of numerous molecularly defined vagal and spinal afferents and identify their downstream brain targets. Bidirectional chemogenetic manipulations, coupled with behavioral and circuit mapping analysis, demonstrated that gut-innervating, glucagon-like peptide 1 receptor (GLP1R)-expressing vagal afferents relay anorexigenic signals to parabrachial nucleus neurons that control meal termination. Moreover, GLP1R vagal afferent activation improves glucose tolerance, and their inhibition elevates blood glucose levels independent of food intake. In contrast, gut-innervating, GPR65-expressing vagal afferent stimulation increases hepatic glucose production and activates parabrachial neurons that control normoglycemia, but they are dispensable for feeding regulation. Thus, distinct gut-innervating sensory neurons differentially control feeding and glucoregulatory neurocircuits and may provide specific targets for metabolic control.
Collapse
Affiliation(s)
- Diba Borgmann
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Translational Neurocircuitry Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Anatomy II, Neuroanatomy, University Hospital Cologne, Joseph-Stelzmann Str. 9, 50937 Cologne, Germany
| | - Elisa Ciglieri
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50937 Cologne, Germany
| | - Nasim Biglari
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Claus Brandt
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Anna Lena Cremer
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Heiko Backes
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Marc Tittgemeyer
- Translational Neurocircuitry Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany
| | - F Thomas Wunderlich
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50937 Cologne, Germany; Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany; Center of Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Straße 21, 50931 Cologne, Germany
| | - Jens C Brüning
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50937 Cologne, Germany; Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany; Center of Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Straße 21, 50931 Cologne, Germany
| | - Henning Fenselau
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50937 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany.
| |
Collapse
|
42
|
Garrett A, Rakhilin N, Wang N, McKey J, Cofer G, Anderson RB, Capel B, Johnson GA, Shen X. Mapping the peripheral nervous system in the whole mouse via compressed sensing tractography. J Neural Eng 2021; 18. [PMID: 33979784 DOI: 10.1088/1741-2552/ac0089] [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: 12/03/2020] [Accepted: 05/12/2021] [Indexed: 11/12/2022]
Abstract
Objective.The peripheral nervous system (PNS) connects the central nervous system with the rest of the body to regulate many physiological functions and is therapeutically targeted to treat diseases such as epilepsy, depression, intestinal dysmotility, chronic pain, and more. However, we still lack understanding of PNS innervation in most organs because the large span, diffuse nature, and small terminal nerve bundle fibers have precluded whole-organism, high resolution mapping of the PNS. We sought to produce a comprehensive peripheral nerve atlas for use in future interrogation of neural circuitry and selection of targets for neuromodulation.Approach.We used diffusion tensor magnetic resonance imaging (DT-MRI) with high-speed compressed sensing to generate a tractogram of the whole mouse PNS. The tractography generated from the DT-MRI data is validated using lightsheet microscopy on optically cleared, antibody stained tissue.Main results.Herein we demonstrate the first comprehensive PNS tractography in a whole mouse. Using this technique, we scanned the whole mouse in 28 h and mapped PNS innervation and fiber network in multiple organs including heart, lung, liver, kidneys, stomach, intestines, and bladder at 70µm resolution. This whole-body PNS tractography map has provided unparalleled information; for example, it delineates the innervation along the gastrointestinal tract by multiple sacral levels and by the vagal nerves. The map enabled a quantitative tractogram that revealed relative innervation of the major organs by each vertebral foramen as well as the vagus nerve.Significance.This novel high-resolution nerve atlas provides a potential roadmap for future neuromodulation therapies and other investigations into the neural circuits which drive homeostasis and disease throughout the body.
Collapse
Affiliation(s)
- Aliesha Garrett
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States of America
| | - Nikolai Rakhilin
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States of America
| | - Nian Wang
- Duke Center for In Vivo Microscopy, Duke University Medical Center, Durham, NC, United States of America
| | - Jennifer McKey
- Department of Cell Biology, School of Medicine, Duke University, Durham, NC, United States of America
| | - Gary Cofer
- Duke Center for In Vivo Microscopy, Duke University Medical Center, Durham, NC, United States of America
| | - Robert Bj Anderson
- Duke Center for In Vivo Microscopy, Duke University Medical Center, Durham, NC, United States of America
| | - Blanche Capel
- Department of Cell Biology, School of Medicine, Duke University, Durham, NC, United States of America
| | - G Allan Johnson
- Duke Center for In Vivo Microscopy, Duke University Medical Center, Durham, NC, United States of America
| | - Xiling Shen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States of America
| |
Collapse
|
43
|
Brizuela M, Castro J, Harrington AM, Brierley SM. Pruritogenic mechanisms and gut sensation: putting the "irritant" into irritable bowel syndrome. Am J Physiol Gastrointest Liver Physiol 2021; 320:G1131-G1141. [PMID: 33949199 DOI: 10.1152/ajpgi.00331.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Chronic abdominal pain is a common clinical condition experienced by patients with irritable bowel syndrome (IBS). A general lack of suitable treatment options for the management of visceral pain is the major contributing factor to the debilitating nature of the disease. Understanding the underlying causes of chronic visceral pain is pivotal to identifying new effective therapies for IBS. This review provides the current evidence, demonstrating that mediators and receptors that induce itch in the skin also act as "gut irritants" in the gastrointestinal tract. Activation of these receptors triggers specific changes in the neuronal excitability of sensory pathways responsible for the transmission of nociceptive information from the periphery to the central nervous system leading to visceral hypersensitivity and visceral pain. Accumulating evidence points to significant roles of irritant mediators and their receptors in visceral hypersensitivity and thus constitutes potential targets for the development of more effective therapeutic options for IBS.
Collapse
Affiliation(s)
- Mariana Brizuela
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Joel Castro
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Andrea M Harrington
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
44
|
Suslov AV, Chairkina E, Shepetovskaya MD, Suslova IS, Khotina VA, Kirichenko TV, Postnov AY. The Neuroimmune Role of Intestinal Microbiota in the Pathogenesis of Cardiovascular Disease. J Clin Med 2021; 10:1995. [PMID: 34066528 PMCID: PMC8124579 DOI: 10.3390/jcm10091995] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/19/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
Currently, a bidirectional relationship between the gut microbiota and the nervous system, which is considered as microbiota-gut-brain axis, is being actively studied. This axis is believed to be a key mechanism in the formation of somatovisceral functions in the human body. The gut microbiota determines the level of activation of the hypothalamic-pituitary system. In particular, the intestinal microbiota is an important source of neuroimmune mediators in the pathogenesis of cardiovascular disease. This review reflects the current state of publications in PubMed and Scopus databases until December 2020 on the mechanisms of formation and participation of neuroimmune mediators associated with gut microbiota in the development of cardiovascular disease.
Collapse
Affiliation(s)
- Andrey V. Suslov
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, 8-2 Trubetskaya Str., 119992 Moscow, Russia; (A.V.S.); (E.C.); (M.D.S.)
| | - Elizaveta Chairkina
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, 8-2 Trubetskaya Str., 119992 Moscow, Russia; (A.V.S.); (E.C.); (M.D.S.)
| | - Maria D. Shepetovskaya
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, 8-2 Trubetskaya Str., 119992 Moscow, Russia; (A.V.S.); (E.C.); (M.D.S.)
| | - Irina S. Suslova
- Central State Medical Academy of the Administrative Department of the President of the Russian Federation, 19-1A Marshal Timoshenko Str., 121359 Moscow, Russia;
| | - Victoria A. Khotina
- Research Institute of Human Morphology, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.A.K.); (A.Y.P.)
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Str., 125315 Moscow, Russia
| | - Tatiana V. Kirichenko
- Research Institute of Human Morphology, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.A.K.); (A.Y.P.)
- National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Str., 121552 Moscow, Russia
| | - Anton Y. Postnov
- Research Institute of Human Morphology, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.A.K.); (A.Y.P.)
- National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Str., 121552 Moscow, Russia
| |
Collapse
|
45
|
Tsushima H, Zhang Y, Muratsubaki T, Kanazawa M, Fukudo S. Oxytocin antagonist induced visceral pain and corticotropin-releasing hormone neuronal activation in the central nucleus of the amygdala during colorectal distention in mice. Neurosci Res 2021; 168:41-53. [PMID: 33932549 DOI: 10.1016/j.neures.2021.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/19/2022]
Abstract
Activation of neurons containing oxytocin and corticotropin-releasing hormone (CRH) in the paraventricular nucleus (PVN) of the hypothalamus, the anterior cingulate cortex (ACC), and the central nucleus of the amygdala (CeA) during colorectal distention (CRD) is likely to play a crucial role in animal models of irritable bowel syndrome (IBS). Earlier studies in rodents showed that the microbiome is involved in social behavior via oxytocin expression in the brain. However, the detailed mechanism of visceral sensation and oxytocin is largely unknown. We tested the following hypotheses: (1) that oxytocin neurons in the PVN are activated by CRD, and (2) that the activation of oxytocin neurons by CRD is related to anxiety-like behavior, visceral perception, and an activation of CRH CeA neurons or ACC neurons. Oxytocin antagonist caused visceral hypersensitivity and anxiety-like behavior. In the PVN, oxytocin neurons were activated by CRD. Noxious CRD activated the CeA, basolateral nucleus of the amygdala (BLA), and ACC. High-dose oxytocin antagonist suppressed ACC activity and activated CRH CeA neurons. These results support our hypotheses. Oxytocin likely regulates CRH CeA neurons in an inhibitory manner and the ACC in an excitatory manner. Further research into the interaction of oxytocin and CRH in visceral pain and anxiety is warranted.
Collapse
Affiliation(s)
- Hiromichi Tsushima
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yanli Zhang
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| | - Tomohiko Muratsubaki
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Motoyori Kanazawa
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Psychosomatic Medicine, Tohoku University Hospital, Sendai, Japan
| | - Shin Fukudo
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Psychosomatic Medicine, Tohoku University Hospital, Sendai, Japan.
| |
Collapse
|
46
|
Najjar SA, Ejoh LL, Loeza-Alcocer E, Edwards BS, Smith-Edwards KM, Epouhe AY, Gold MS, Davis BM, Albers KM. Optogenetic inhibition of the colon epithelium reduces hypersensitivity in a mouse model of inflammatory bowel disease. Pain 2021; 162:1126-1134. [PMID: 33048854 PMCID: PMC7969374 DOI: 10.1097/j.pain.0000000000002110] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/07/2020] [Indexed: 11/25/2022]
Abstract
ABSTRACT Visceral pain is a prevalent symptom of inflammatory bowel disease that can be difficult to treat. Pain and hypersensitivity are mediated by extrinsic primary afferent neurons (ExPANs) that innervate the colon. Recent studies indicate that the colon epithelium contributes to initiating ExPAN firing and nociceptive responses. Based on these findings, we hypothesized that the epithelium contributes to inflammation-induced hypersensitivity. A key prediction of this hypothesis is that inhibition of the epithelium would attenuate nociceptive signaling and inflammatory hypersensitivity. To test this hypothesis, the inhibitory yellow light-activated protein archaerhodopsin was targeted to the intestinal epithelium (villin-Arch) or the ExPANs (TRPV1-Arch) that innervate the colon. Visceral sensitivity was assessed by measuring the visceromotor response (VMR) to colorectal distension (CRD), with and without yellow light illumination of the colon lumen. Inhibition of the colon epithelium in healthy villin-Arch mice significantly diminished the CRD-induced VMR. Direct inhibition of ExPANs during CRD using TRPV1-Arch mice showed that ExPAN and epithelial inhibition were similarly effective in reducing the VMR to CRD. We then investigated the effect of epithelial and ExPAN inhibition in the dextran sulfate sodium model of inflammatory bowel disease. Inhibition of the colon epithelium significantly decreased dextran sulfate sodium-induced hypersensitivity and was comparable with the inhibition of ExPANs. Together, these results reveal the potential of targeting the colon epithelium for the treatment of pain.
Collapse
Affiliation(s)
- Sarah A. Najjar
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lindsay L. Ejoh
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Emanuel Loeza-Alcocer
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Brian S. Edwards
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kristen M. Smith-Edwards
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ariel Y. Epouhe
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael S. Gold
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Brian M. Davis
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kathryn M. Albers
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
47
|
Ramirez VT, Sladek J, Godinez DR, Rude KM, Chicco P, Murray K, Brust-Mascher I, Gareau MG, Reardon C. Sensory Nociceptive Neurons Contribute to Host Protection During Enteric Infection With Citrobacter rodentium. J Infect Dis 2021; 221:1978-1988. [PMID: 31960920 DOI: 10.1093/infdis/jiaa014] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/15/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Neurons are an integral component of the immune system that functions to coordinate responses to bacterial pathogens. Sensory nociceptive neurons that can detect bacterial pathogens are found throughout the body with dense innervation of the intestinal tract. METHODS In this study, we assessed the role of these nerves in the coordination of host defenses to Citrobacter rodentium. Selective ablation of nociceptive neurons significantly increased bacterial burden 10 days postinfection and delayed pathogen clearance. RESULTS Because the sensory neuropeptide CGRP (calcitonin gene-related peptide) regulates host responses during infection of the skin, lung, and small intestine, we assessed the role of CGRP receptor signaling during C rodentium infection. Although CGRP receptor blockade reduced certain proinflammatory gene expression, bacterial burden and Il-22 expression was unaffected. CONCLUSIONS Our data highlight that sensory nociceptive neurons exert a significant host protective role during C rodentium infection, independent of CGRP receptor signaling.
Collapse
Affiliation(s)
- Valerie T Ramirez
- Department of Anatomy, Physiology, & Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| | - Jessica Sladek
- Department of Anatomy, Physiology, & Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| | - Dayn Romero Godinez
- Department of Anatomy, Physiology, & Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| | - Kavi M Rude
- Department of Anatomy, Physiology, & Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| | - Pamela Chicco
- Department of Anatomy, Physiology, & Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| | - Kaitlin Murray
- Department of Anatomy, Physiology, & Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| | - Ingrid Brust-Mascher
- Department of Anatomy, Physiology, & Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| | - Melanie G Gareau
- Department of Anatomy, Physiology, & Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| | - Colin Reardon
- Department of Anatomy, Physiology, & Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California, USA
| |
Collapse
|
48
|
Cao J, O'Day DR, Pliner HA, Kingsley PD, Deng M, Daza RM, Zager MA, Aldinger KA, Blecher-Gonen R, Zhang F, Spielmann M, Palis J, Doherty D, Steemers FJ, Glass IA, Trapnell C, Shendure J. A human cell atlas of fetal gene expression. Science 2020; 370:370/6518/eaba7721. [PMID: 33184181 DOI: 10.1126/science.aba7721] [Citation(s) in RCA: 364] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022]
Abstract
The gene expression program underlying the specification of human cell types is of fundamental interest. We generated human cell atlases of gene expression and chromatin accessibility in fetal tissues. For gene expression, we applied three-level combinatorial indexing to >110 samples representing 15 organs, ultimately profiling ~4 million single cells. We leveraged the literature and other atlases to identify and annotate hundreds of cell types and subtypes, both within and across tissues. Our analyses focused on organ-specific specializations of broadly distributed cell types (such as blood, endothelial, and epithelial), sites of fetal erythropoiesis (which notably included the adrenal gland), and integration with mouse developmental atlases (such as conserved specification of blood cells). These data represent a rich resource for the exploration of in vivo human gene expression in diverse tissues and cell types.
Collapse
Affiliation(s)
- Junyue Cao
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Diana R O'Day
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Hannah A Pliner
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Paul D Kingsley
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Mei Deng
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Riza M Daza
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Michael A Zager
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.,Center for Data Visualization, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Kimberly A Aldinger
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Ronnie Blecher-Gonen
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Malte Spielmann
- Human Molecular Genomics Group, Max Planck Institute for Molecular Genetics, Berlin, Germany.,Institute of Human Genetics, University of Lübeck, Lübeck, Germany
| | - James Palis
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Dan Doherty
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA.,Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Ian A Glass
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA.,Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Cole Trapnell
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA. .,Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.,Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA. .,Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.,Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.,Howard Hughes Medical Institute, Seattle, WA, USA
| |
Collapse
|
49
|
Abstract
The gut-brain axis is a coordinated communication system that not only maintains homeostasis, but significantly influences higher cognitive functions and emotions, as well as neurological and behavioral disorders. Among the large populations of sensory and motor neurons that innervate the gut, insights into the function of primary afferent nociceptors, whose cell bodies reside in the dorsal root ganglia and nodose ganglia, have revealed their multiple crosstalk with several cell types within the gut wall, including epithelial, vascular, and immune cells. These bidirectional communications have immunoregulatory functions, control host response to pathogens, and modulate sensations associated with gastrointestinal disorders, through activation of immune cells and glia in the peripheral and central nervous system, respectively. Here, we will review the cellular and neurochemical basis of these interactions at the periphery, in dorsal root ganglia, and in the spinal cord. We will discuss the research gaps that should be addressed to get a better understanding of the multifunctional role of sensory neurons in maintaining gut homeostasis and regulating visceral sensitivity.
Collapse
Affiliation(s)
- Nasser Abdullah
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Manon Defaye
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
50
|
Lactobacillus rhamnosus GG soluble mediators ameliorate early life stress-induced visceral hypersensitivity and changes in spinal cord gene expression. Neuronal Signal 2020; 4:NS20200007. [PMID: 33343931 PMCID: PMC7726314 DOI: 10.1042/ns20200007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 10/26/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
Visceral hypersensitivity is a hallmark of many functional and stress-related gastrointestinal disorders, and there is growing evidence that the gut microbiota may play a role in its pathophysiology. It has previously been shown that early life stress-induced visceral sensitivity is reduced by various probiotic strains of bacteria (including Lactobacillus rhamnosus GG (LGG)) alone or in combination with prebiotic fibres in rat models. However, the exact mechanisms underpinning such effects remain unresolved. Here, we investigated if soluble mediators derived from LGG can mimic the bacteria's effects on visceral hypersensitivity and the microbiota-gut-brain axis. Rats were exposed to maternal separation (MS) from postnatal days 2-12. From weaning onwards both non-separated (NS) and MS offspring were provided drinking water with or without supplementation of standardized preparations of the LGG soluble mediators (LSM). Our results show that MS led to increased visceral sensitivity and exaggerated corticosterone plasma levels following restraint stress in adulthood, and both of these effects were ameliorated through LSM supplementation. Differential regulation of various genes in the spinal cord of MS versus NS rats was observed, 41 of which were reversed by LSM supplementation. At the microbiota composition level MS led to changes in beta diversity and abundance of specific bacteria including parabacteroides, which were ameliorated by LSM. These findings support probiotic soluble mediators as potential interventions in the reduction of symptoms of visceral hypersensitivity.
Collapse
|