1
|
Meerschaert KA, Chiu IM. The gut-brain axis and pain signalling mechanisms in the gastrointestinal tract. Nat Rev Gastroenterol Hepatol 2025; 22:206-221. [PMID: 39578592 DOI: 10.1038/s41575-024-01017-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 11/24/2024]
Abstract
Visceral pain is a major clinical problem and one of the most common reasons patients with gastrointestinal disorders seek medical help. Peripheral sensory neurons that innervate the gut can detect noxious stimuli and send signals to the central nervous system that are perceived as pain. There is a bidirectional communication network between the gastrointestinal tract and the nervous system that mediates pain through the gut-brain axis. Sensory neurons detect mechanical and chemical stimuli within the intestinal tissues, and receive signals from immune cells, epithelial cells and the gut microbiota, which results in peripheral sensitization and visceral pain. This Review focuses on molecular communication between these non-neuronal cell types and neurons in visceral pain. These bidirectional interactions can be dysregulated during gastrointestinal diseases to exacerbate visceral pain. We outline the anatomical pathways involved in pain processing in the gut and how cell-cell communication is integrated into this gut-brain axis. Understanding how bidirectional communication between the gut and nervous system is altered during disease could provide new therapeutic targets for treating visceral pain.
Collapse
Affiliation(s)
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Larauche M, Mulak A, Ha C, Million M, Arnett S, Germano P, Pearson JP, Currie MG, Taché Y. FAAH inhibitor URB597 shows anti-hyperalgesic action and increases brain and intestinal tissues fatty acid amides in a model of CRF 1 agonist mediated visceral hypersensitivity in male rats. Neurogastroenterol Motil 2024; 36:e14927. [PMID: 39344695 PMCID: PMC11781189 DOI: 10.1111/nmo.14927] [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: 04/11/2024] [Revised: 08/05/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND AND AIMS The endocannabinoid (eCB) system includes ligands (anandamide and 2-arachidonoyl glycerol, 2-AG), receptors and catabolizing enzymes (fatty acid amide hydrolase, FAAH and monoacylglycerol lipase) expressed in both the brain and gut. We investigated whether the FAAH inhibitor, URB597, influenced visceral pain to colorectal distension (CRD) in an acute stress-related model of visceral hypersensitivity induced by the selective corticotropin-releasing factor receptor subtype 1 (CRF1) agonist, cortagine. METHODS Male Sprague-Dawley rats were injected subcutaneously (SC) with URB597 (3 mg/kg) or vehicle and 2 h later, intraperitoneally with cortagine (10 μg/kg) or vehicle. The visceromotor responses (VMR) were assessed to a first CRD (baseline) before injections, and to a second CRD 15 min after the last treatment. Brain, jejunum, and proximal colon were collected from treated and naïve rats for levels quantification of three fatty acid amides (FAAs) [anandamide (arachidonyl-ethanolamide, AEA), oleoyl-ethanolamide (OEA) and palmitoyl-ethanolamide (PEA)], and 2-AG. In separate animals, defecation/diarrhea were monitored after URB597 and cortagine. KEY RESULTS URB597 inhibited cortagine-induced increased VMR at 40 mmHg (89.0 ± 14.8% vs. 132.5 ± 15.6% for vehicle SC, p < 0.05) and 60 mmHg (107.5 ± 16.1% vs. 176.9 ± 24.4% for vehicle SC, p < 0.001) while not influencing basal VMR. In URB597 plus cortagine group, FAAs levels increased in the brain and intestinal tissue while 2-AG did not change. URB597 did not modify cortagine-induced defecation/diarrhea versus vehicle. CONCLUSIONS AND INFERENCES URB597 shows efficacy to elevate brain and intestinal FAAs and to counteract the colonic hypersensitivity induced by peripheral activation of CRF1 signaling supporting a potential strategy of FAAH inhibitors to alleviate stress-related visceral hypersensitivity.
Collapse
Affiliation(s)
- Muriel Larauche
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90056, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Agata Mulak
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90056, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Present Affiliation: Department of Gastroenterology and Hepatology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Chrysanthy Ha
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90056, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Present Affiliation: Community Memorial Healthcare, Ventura, CA, USA
| | - Mulugeta Million
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90056, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Stacy Arnett
- Present Affiliation: Department of Pathology, St. Louis University, St. Louis, MO, USA
| | - Peter Germano
- Present Affiliation: Auron Therapeutics, Inc., Newton, MA, USA
| | - James P. Pearson
- Present Affiliation: Sea Pharmaceuticals LLC, Cambridge, MA, USA
| | - Mark G. Currie
- Present Affiliation: Sea Pharmaceuticals LLC, Cambridge, MA, USA
| | - Yvette Taché
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90056, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| |
Collapse
|
3
|
Tiwari N, Qiao LY. Sex Differences in Visceral Pain and Comorbidities: Clinical Outcomes, Preclinical Models, and Cellular and Molecular Mechanisms. Cells 2024; 13:834. [PMID: 38786056 PMCID: PMC11119472 DOI: 10.3390/cells13100834] [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: 03/19/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Sexual dimorphism of visceral pain has been documented in clinics and experimental animal models. Aside from hormones, emerging evidence suggests the sex-differential intrinsic neural regulation of pain generation and maintenance. According to the International Association for the Study of Pain (IASP) and the American College of Gastroenterology (ACG), up to 25% of the population have visceral pain at any one time, and in the United States 10-15 percent of adults suffer from irritable bowel syndrome (IBS). Here we examine the preclinical and clinical evidence of sex differences in visceral pain focusing on IBS, other forms of bowel dysfunction and IBS-associated comorbidities. We summarize preclinical animal models that provide a means to investigate the underlying molecular mechanisms in the sexual dimorphism of visceral pain. Neurons and nonneuronal cells (glia and immune cells) in the peripheral and central nervous systems, and the communication of gut microbiota and neural systems all contribute to sex-dependent nociception and nociplasticity in visceral painful signal processing. Emotion is another factor in pain perception and appears to have sexual dimorphism.
Collapse
Affiliation(s)
- Namrata Tiwari
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Liya Y. Qiao
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| |
Collapse
|
4
|
Larauche M, Kim YS, Mulak A, Duboc H, Taché Y. Intracerebroventricular administration of TRH Agonist, RX-77368 alleviates visceral pain induced by colorectal distension in rats. Peptides 2024; 175:171181. [PMID: 38423212 PMCID: PMC11841477 DOI: 10.1016/j.peptides.2024.171181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 01/18/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
Thyrotropin-releasing hormone (TRH) acts centrally to exert pleiotropic actions independently from its endocrine function, including antinociceptive effects against somatic pain in rodents. Whether exogenous or endogenous activation of TRH signaling in the brain modulates visceral pain is unknown. Adult male Sprague-Dawley rats received an intracerebroventricular (ICV) injection of the stable TRH analog, RX-77368 (10, 30 and 100 ng/rat) or saline (5 µl) or were semi-restrained and exposed to cold (4°C) for 45 min. The visceromotor response (VMR) to graded phasic colorectal distensions (CRD) was monitored using non-invasive intracolonic pressure manometry. Naloxone (1 mg/kg) was injected subcutaneously 10 min before ICV RX-77368 or saline. Fecal pellet output was monitored for 1 h after ICV injection. RX-77368 ICV (10, 30 and 100 ng/rat) reduced significantly the VMR by 56.7%, 67.1% and 81.1% at 40 mmHg and by 30.3%, 58.9% and 87.4% at 60 mmHg respectively vs ICV saline. Naloxone reduced RX-77368 (30 and 100 ng, ICV) analgesic response by 51% and 28% at 40 mmHg and by 30% and 33% at 60 mmHg respectively, but had no effect per se. The visceral analgesia was mimicked by the acute exposure to cold. At the doses of 30 and 100 ng, ICV RX-77368 induced defecation within 30 min. These data established the antinociceptive action of RX-77368 injected ICV in a model of visceral pain induced by colonic distension through recruitment of both opioid and non-opioid dependent mechanisms.
Collapse
Affiliation(s)
- Muriel Larauche
- Digestive Diseases Research Center and G. Oppenheimer Center for Neurobiology of Stress and Resilience, Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, and VA Greater Los Angeles Healthcare System, CA 90073, USA.
| | - Yong Sung Kim
- Digestive Diseases Research Center and G. Oppenheimer Center for Neurobiology of Stress and Resilience, Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, and VA Greater Los Angeles Healthcare System, CA 90073, USA
| | - Agata Mulak
- Digestive Diseases Research Center and G. Oppenheimer Center for Neurobiology of Stress and Resilience, Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, and VA Greater Los Angeles Healthcare System, CA 90073, USA
| | - Henri Duboc
- Digestive Diseases Research Center and G. Oppenheimer Center for Neurobiology of Stress and Resilience, Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, and VA Greater Los Angeles Healthcare System, CA 90073, USA
| | - Yvette Taché
- Digestive Diseases Research Center and G. Oppenheimer Center for Neurobiology of Stress and Resilience, Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, and VA Greater Los Angeles Healthcare System, CA 90073, USA
| |
Collapse
|
5
|
Larauche M, Erchegyi J, Miller C, Sim MS, Rivier J, Behan D, Taché Y. Peripheral CRF-R1/CRF-R2 antagonist, astressin C, induces a long-lasting blockade of acute stress-related visceral pain in male and female rats. Peptides 2022; 157:170881. [PMID: 36185037 PMCID: PMC10389693 DOI: 10.1016/j.peptides.2022.170881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/06/2022] [Accepted: 09/16/2022] [Indexed: 11/15/2022]
Abstract
Peptide CRF antagonists injected peripherally alleviate stress-induced visceral hypersensitivity (SIVH) to colorectal distension (CRD) in rodents. Here we further evaluated the dose and time-dependent inhibitory activity of several long-acting peptide CRF receptor antagonists related to astressin on SIVH, focusing on astressin C (AstC), which previously showed high efficacy on stress-related alterations of HPA axis and gut secretomotor functions. Male and female Sprague-Dawley rats pretreated subcutaneously (SC) with AstC were injected intraperitoneally (IP) with CRF 15 min later. The visceromotor responses (VMR) to graded phasic CRD (10, 20, 40 and 60 mmHg) were monitored at basal, 15 min and up to 1-8 days after pretreatment. Two other astressin analogs, hexanoyl-astressin D (Hex-AstD) and [CαMeVal19,32]-AstC, were also tested. The response to IP CRF was sex-dependent with female rats requiring a higher dose to exhibit visceral hyperalgesia. Pretreatment with AstC (30-1000 µg/kg) resulted in a dose-related inhibition of IP CRF-induced SIVH and diarrhea in both sexes. The highest dose prevented SIVH and diarrhea up to 5-7 days after a single SC injection and was lost on day 7 (females) and day 8 (males) but reinstated after a second injection of AstC on day 8 or 9 respectively. [CαMeVal19,32]-AstC and Hex-AstD (1000 µg/kg in males) also prevented SIVH. These data show the potent long-lasting anti-hyperalgesic effect of AstC in an acute model of SIVH in both male and female rats. This highlights the potential of long-acting peripheral CRF antagonists to treat stress-sensitive irritable bowel syndrome.
Collapse
Affiliation(s)
- Muriel Larauche
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, West Los Angeles, CA, USA.
| | | | | | - Myung Shin Sim
- Department of Medicine, Statistic Core, UCLA, Los Angeles, CA, USA
| | - Jean Rivier
- Sentia Medical Sciences, Inc., San Diego, CA, USA
| | | | - Yvette Taché
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, West Los Angeles, CA, USA
| |
Collapse
|
6
|
López‐Estévez S, López‐Torrellardona JM, Parera M, Martínez V. Long-lasting visceral hypersensitivity in a model of DSS-induced colitis in rats. Neurogastroenterol Motil 2022; 34:e14441. [PMID: 36239298 PMCID: PMC9787759 DOI: 10.1111/nmo.14441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 05/31/2022] [Accepted: 07/18/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Persistent visceral hypersensitivity is a key component of functional and inflammatory gastrointestinal diseases. Current animal models fail to fully reproduce the characteristics of visceral pain in humans, particularly as it relates to persistent hypersensitivity. This work explores the validity of DSS-induced colitis in rats as a model to mimic chronic intestinal hypersensitivity. METHODS Exposure to DSS (5% for 7 days) was used to induce colitis in rats. Thereafter, changes in viscerosensitivity (visceromotor responses to colorectal distension-CRD), the presence of somatic referred pain (mechanosensitivity of the hind paws, von Frey test) and the expression (qRT-PCR) of sensory-related markers (colon, lumbosacral DRGs, and lumbosacral spinal cord) were assessed at different times during the 35 days period after colitis induction. RESULTS Following colitis, a sustained increase in visceromotor responses to CRD were observed, indicative of the presence of visceral hypersensitivity. Responses in animals without colitis remained stable over time. In colitic animals, somatic referred hypersensitivity was also detected. DSS-induced colitis was associated to a differential expression of sensory-related markers (with both pro- and anti-nociceptive action) in the colon, lumbosacral DRGs and lumbosacral spinal cord; indicating the presence of peripheral and central sensitization. CONCLUSIONS AND INFERENCES DSS-induced colitis in rats is associated to the generation of a long-lasting state of visceral (colonic) hypersensitivity, despite clinical colitis resolution. This model reproduces the changes in intestinal sensitivity characteristics of inflammatory and functional gastrointestinal disorders in humans and can be used in the characterization of new pharmacological treatments against visceral pain.
Collapse
Affiliation(s)
- Sergio López‐Estévez
- Department of Cell Biology, Physiology and ImmunologyUniversitat Autònoma de BarcelonaBarcelonaSpain
- Neuroscience InstituteUniversitat Autònoma de BarcelonaBarcelonaSpain
| | | | - Marc Parera
- Department of Cell Biology, Physiology and ImmunologyUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Vicente Martínez
- Department of Cell Biology, Physiology and ImmunologyUniversitat Autònoma de BarcelonaBarcelonaSpain
- Neuroscience InstituteUniversitat Autònoma de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
7
|
Nozu T, Miyagishi S, Ishioh M, Takakusaki K, Okumura T. Peripheral apelin mediates visceral hypersensitivity and impaired gut barrier in a rat irritable bowel syndrome model. Neuropeptides 2022; 94:102248. [PMID: 35526468 DOI: 10.1016/j.npep.2022.102248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 12/19/2022]
Abstract
Growing evidence indicates that visceral hypersensitivity and impaired gut barrier play an important role in the pathophysiology of irritable bowel syndrome (IBS). In animal models, these changes are known to be mediated via corticotropin-releasing factor (CRF)-Toll like receptor 4 (TLR4)-proinflammatory cytokine signaling. Apelin, an endogenous ligand of APJ, was reported to modulate CRF-induced enhanced colonic motility. In this context, we hypothesized that apelin also modulates visceral sensation and gut barrier, and tested this hypothesis. We measured visceral pain threshold in response to colonic balloon distention by abdominal muscle contractions assessed by electromyogram in rats. Colonic permeability was estimated by quantifying the absorbed Evans blue in colonic tissue. Intraperitoneal (ip) administration of [Ala13]-apelin-13, an APJ antagonist, blocked lipopolysaccharide (LPS)- or CRF-induced visceral hypersensitivity and colonic hyperpermeability (IBS model) in a dose-response manner. These inhibitory effects were blocked by compound C, an AMPK inhibitor, NG-nitro-L-arginine methyl ester, a nitric oxide (NO) synthesis inhibitor or naloxone in the LPS model. On the other hand, ip [Pyr1]-apelin-13, an APJ agonist, caused visceral hypersensitivity and colonic hyperpermeability, and these effects were reversed by astressin, a CRF receptor antagonist, TAK-242, a TLR4 antagonist or anakinra, an interleukin-1 receptor antagonist. APJ system modulated CRF-TLR4-proinflammatory cytokine signaling to cause visceral hypersensitivity and colonic hyperpermeability. APJ antagonist blocked these GI changes in IBS models, which were mediated via AMPK, NO and opioid signaling. Apelin may contribute to the IBS pathophysiology, and the inhibition of apelinergic signaling may be a promising therapeutic option for IBS.
Collapse
Affiliation(s)
- Tsukasa Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan; Center for Medical Education, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan.
| | - Saori Miyagishi
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| | - Masatomo Ishioh
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| | - Kaoru Takakusaki
- Division of Neuroscience, Department of Physiology, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| | - Toshikatsu Okumura
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan; Department of General Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| |
Collapse
|
8
|
Chen T, Chen S, Zheng X, Zhu Y, Huang Z, Jia L, OuYang L, Lei W. The pathological involvement of spinal cord EphB2 in visceral sensitization in male rats. Stress 2022; 25:166-178. [PMID: 35435121 DOI: 10.1080/10253890.2022.2054698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Patients with post-traumatic stress disorder (PTSD) are usually at an increased risk for chronic disorders, such as irritable bowel syndrome (IBS), characterized by hyperalgesia and allodynia, but its subsequent effect on visceral hyperalgesia and the mechanism remain unclear. The present study employed single prolonged stress (SPS), a model of PTSD-pain comorbidity, behavioral evaluation, intrathecal drug delivery, immunohistochemistry, Western blotting, and RT-PCR techniques. When detecting visceral sensitivity, the score of the abdominal withdrawal reflex (AWR) induced by graded colorectal distention (CRD) was used. The AWR score was reduced in the SPS day 1 group but increased in the SPS day 7 and SPS day 14 groups at 40 mmHg and 60 mmHg, and the score was increased significantly with EphrinB1-Fc administration. The EphB2+ cell density and EphB2 protein and mRNA levels were downregulated in the SPS day 1 group and then upregulated significantly in the SPS day 7 group; these changes were more noticeable with EphrinB1-Fc administration compared with the SPS-only group. The C-Fos-positive reaction induced by SPS was mainly localized in neurons of the spinal dorsal horn, in which the C-Fos-positive cell density and its protein and mRNA levels were upregulated on SPS days 7 and 14; these changes were statistically significant in the SPS + EphrinB1-Fc group compared with the SPS alone group. The present study confirmed the time window for the AWR value, EphB2 and C-Fos changes, and the effect of EphrinB1-Fc on these changes, which suggests that spinal cord EphB2 activation exacerbates visceral pain after SPS.
Collapse
Affiliation(s)
- Tao Chen
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Si Chen
- Department of Human Anatomy and Histology & Embryology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Xuefeng Zheng
- Neuroscience Laboratory for Cognitive and Developmental Disorders, Department of Anatomy, Medical College of Jinan University, Guangzhou, China
| | - Yaofeng Zhu
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ziyun Huang
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Linju Jia
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lisi OuYang
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wanlong Lei
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
9
|
Nozu T, Miyagishi S, Ishioh M, Takakusaki K, Okumura T. Phlorizin attenuates visceral hypersensitivity and colonic hyperpermeability in a rat model of irritable bowel syndrome. Biomed Pharmacother 2021; 139:111649. [PMID: 33957565 DOI: 10.1016/j.biopha.2021.111649] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/07/2021] [Accepted: 04/19/2021] [Indexed: 12/18/2022] Open
Abstract
Visceral hypersensitivity and impaired gut barrier are crucial contributors to the pathophysiology of irritable bowel syndrome (IBS), and those are mediated via corticotropin-releasing factor (CRF)-Toll like receptor 4-pro-inflammatory cytokine signaling. Phlorizin is an inhibitor of sodium-linked glucose transporters (SGLTs), and known to have anti-cytokine properties. Thus, we hypothesized that phlorizin may improve these gastrointestinal changes in IBS, and tested this hypothesis in rat IBS models, i.e., lipopolysaccharide (LPS) or CRF-induced visceral hypersensitivity and colonic hyperpermeability. The visceral pain threshold in response to colonic balloon distention was estimated by abdominal muscle contractions by electromyogram, and colonic permeability was measured by quantifying the absorbed Evans blue in colonic tissue. Subcutaneous (s.c.) injection of phlorizin inhibited visceral hypersensitivity and colonic hyperpermeability induced by LPS in a dose-dependent manner. Phlorizin also blocked CRF-induced these gastrointestinal changes. Phlorizin is known to inhibit both SGLT1 and SGLT2, but intragastric administration of phlorizin may only inhibit SGLT1 because gut mainly expresses SGLT1. We found that intragastric phlorizin did not display any effects, but ipragliflozin, an orally active and selective SGLT2 inhibitor improved the gastrointestinal changes in the LPS model. Compound C, an adenosine monophosphate-activated protein kinase (AMPK) inhibitor, NG-nitro-L-arginine methyl ester, a nitric oxide (NO) synthesis inhibitor and naloxone, an opioid receptor antagonist reversed the effects of phlorizin. In conclusions, phlorizin improved visceral hypersensitivity and colonic hyperpermeability in IBS models. These effects may result from inhibition of SGLT2, and were mediated via AMPK, NO and opioid pathways. Phlorizin may be effective for the treatment of IBS.
Collapse
Affiliation(s)
- Tsukasa Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan; Center for Medical Education, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan.
| | - Saori Miyagishi
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan
| | - Masatomo Ishioh
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan
| | - Kaoru Takakusaki
- Division of Neuroscience, Department of Physiology, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| | - Toshikatsu Okumura
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan; Department of General Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| |
Collapse
|
10
|
Mauvais-Jarvis F, Berthold HK, Campesi I, Carrero JJ, Dakal S, Franconi F, Gouni-Berthold I, Heiman ML, Kautzky-Willer A, Klein SL, Murphy A, Regitz-Zagrosek V, Reue K, Rubin JB. Sex- and Gender-Based Pharmacological Response to Drugs. Pharmacol Rev 2021; 73:730-762. [PMID: 33653873 PMCID: PMC7938661 DOI: 10.1124/pharmrev.120.000206] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In humans, the combination of all sex-specific genetic, epigenetic, and hormonal influences of biologic sex produces different in vivo environments for male and female cells. We dissect how these influences of sex modify the pharmacokinetics and pharmacodynamics of multiple drugs and provide examples for common drugs acting on specific organ systems. We also discuss how gender of physicians and patients may influence the therapeutic response to drugs. We aim to highlight sex as a genetic modifier of the pharmacological response to drugs, which should be considered as a necessary step toward precision medicine that will benefit men and women. SIGNIFICANCE STATEMENT: This study discusses the influences of biologic sex on the pharmacokinetics and pharmacodynamics of drugs and provides examples for common drugs acting on specific organ systems. This study also discusses how gender of physicians and patients influence the therapeutic response to drugs.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Heiner K Berthold
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Ilaria Campesi
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Juan-Jesus Carrero
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Santosh Dakal
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Flavia Franconi
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Ioanna Gouni-Berthold
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Mark L Heiman
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Alexandra Kautzky-Willer
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Sabra L Klein
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Anne Murphy
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Vera Regitz-Zagrosek
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Karen Reue
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| | - Joshua B Rubin
- Section of Endocrinology, John W. Deming Department of Medicine, Diabetes Discovery and Sex-Based Medicine Laboratory, Tulane University School of Medicine and Southeast Louisiana Veterans Health Care System Medical Center, New Orleans, Louisiana (F.M.-J.); Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), Bielefeld, Germany (H.K.B.); Department of Biomedical Sciences, University of Sassari, Sassari, Italy (I.C.); Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (J.-J.C.); W. Harry Feinstone Department of Molecular Microbiology and Immunology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (S.D., S.L.K.); Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy (F.F.); Polyclinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University of Cologne, Cologne, Germany (I.G.-B.); Scioto Biosciences, Indianapolis, Indiana (M.L.H.); Department of Internal Medicine III, Clinical Division of Endocrinology, Metabolism and Gender Medicine, Medical University of Vienna, Vienna and Gender Institute Gars am Kamp, Vienna, Austria (A.K.-W.); Neuroscience Institute, Georgia State University, Atlanta, Georgia (A.M.); Berlin Institute of Gender Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany and University of Zürich, Switzerland (V.R.-Z.); Department of Human Genetics, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California (K.R.); and Departments of Medicine, Pediatrics, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (J.B.R.)
| |
Collapse
|
11
|
The Infant-Derived Bifidobacterium bifidum Strain CNCM I-4319 Strengthens Gut Functionality. Microorganisms 2020; 8:microorganisms8091313. [PMID: 32872165 PMCID: PMC7565306 DOI: 10.3390/microorganisms8091313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/21/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
Bifidobacteria are among the first colonisers of the gastrointestinal tract of breast-fed newborns due to, among other things, their ability to metabolise oligosaccharides naturally occurring in human milk. The presence of bifidobacteria in the infant gut has been shown to promote intestinal health and homeostasis as well as to preserve a functional gut barrier, thus positively influencing host health and well-being. Among human-associated gut commensals, Bifidobacterium bifidum has been described as the only species capable of the extracellular degradation of both mucin-type glycans and HMOs, thereby giving this species a special role as a commensal gut forager of both host and diet-derived glycans. In the present study, we assess the possible beneficial properties and probiotic potential of B. bifidum strain CNCM I-4319. In silico genome analysis and growth experiments confirmed the expected ability of this strain to consume HMOs and mucin. By employing various animal models, we were also able to assess the ability of B. bifidum CNCM I-4319 to preserve gut integrity and functionality from stress-induced and inflammatory damage, thereby enforcing its potential as an effective probiotic strain.
Collapse
|
12
|
Ji Y, Hu B, Klontz C, Li J, Dessem D, Dorsey SG, Traub RJ. Peripheral mechanisms contribute to comorbid visceral hypersensitivity induced by preexisting orofacial pain and stress in female rats. Neurogastroenterol Motil 2020; 32:e13833. [PMID: 32155308 PMCID: PMC7319894 DOI: 10.1111/nmo.13833] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/24/2020] [Accepted: 02/18/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Stress exacerbates many chronic pain syndromes including irritable bowel syndrome (IBS). Among these patient populations, many suffer from comorbid or chronic overlapping pain conditions and are predominantly female. Nevertheless, basic studies investigating chronic psychological stress-induced changes in pain sensitivity have been mostly carried out in male rodents. Our laboratory developed a model of comorbid pain hypersensitivity (CPH) (stress in the presence of preexisting orofacial pain inducing chronic visceral pain hypersensitivity that significantly outlasts transient stress-induced pain hypersensitivity (SIH)) facilitating the study of pain associated with IBS. Since CPH and SIH are phenotypically similar until SIH resolves and CPH persists, it is unclear if underlying mechanisms are similar. METHODS In the present study, the visceromotor response (VMR) to colorectal distention was recorded in the SIH and CPH models in intact females and ovariectomized rats plus estradiol replacement (OVx + E2). Over several months, rats were determined to be susceptible or resilient to stress and the role of peripheral corticotrophin-releasing factor (CRF) underlying in the pain hypersensitivity was examined. KEY RESULTS Stress alone induced transient (3-4 weeks) visceral hypersensitivity, though some rats were resilient. Comorbid conditions increased susceptibility to stress prolonging hypersensitivity beyond 13 weeks. Both models had robust peripheral components; hypersensitivity was attenuated by the CRF receptor antagonist astressin and the mast cell stabilizer disodium cromoglycate (DSCG). However, DSCG was less effective in the CPH model compared to the SIH model. CONCLUSIONS AND INFERENCES The data indicate many similarities but some differences in mechanisms contributing to comorbid pain conditions compared to transient stress-induced pain.
Collapse
Affiliation(s)
- Yaping Ji
- Department of Neural and Pain SciencesSchool of DentistryUniversity of Maryland BaltimoreBaltimoreMDUSA
| | - Bo Hu
- Department of Neural and Pain SciencesSchool of DentistryUniversity of Maryland BaltimoreBaltimoreMDUSA,Present address:
Key laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchXi’an Jiao Tong University College of StomatologyXi’anShaanxiChina
| | - Charles Klontz
- Department of Neural and Pain SciencesSchool of DentistryUniversity of Maryland BaltimoreBaltimoreMDUSA
| | - Jiyun Li
- Department of Neural and Pain SciencesSchool of DentistryUniversity of Maryland BaltimoreBaltimoreMDUSA
| | - Dean Dessem
- Department of Neural and Pain SciencesSchool of DentistryUniversity of Maryland BaltimoreBaltimoreMDUSA,UM Center to Advance Chronic Pain ResearchUniversity of Maryland BaltimoreBaltimoreMDUSA
| | - Susan G. Dorsey
- UM Center to Advance Chronic Pain ResearchUniversity of Maryland BaltimoreBaltimoreMDUSA,Department of Pain and Translational Symptom ScienceSchool of NursingUniversity of Maryland BaltimoreBaltimoreMDUSA
| | - Richard J. Traub
- Department of Neural and Pain SciencesSchool of DentistryUniversity of Maryland BaltimoreBaltimoreMDUSA,UM Center to Advance Chronic Pain ResearchUniversity of Maryland BaltimoreBaltimoreMDUSA
| |
Collapse
|
13
|
Nozu T, Miyagishi S, Nozu R, Takakusaki K, Okumura T. Losartan improves visceral sensation and gut barrier in a rat model of irritable bowel syndrome. Neurogastroenterol Motil 2020; 32:e13819. [PMID: 32056324 DOI: 10.1111/nmo.13819] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/03/2020] [Accepted: 01/22/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Lipopolysaccharide (LPS) or repeated water avoidance stress (WAS) induces visceral allodynia and colonic hyperpermeability via corticotropin-releasing factor (CRF) and proinflammatory cytokines, which is considered to be a rat irritable bowel syndrome (IBS) model. As losartan is known to inhibit proinflammatory cytokine release, we hypothesized that it improves these visceral changes. METHODS The threshold of visceromotor response (VMR), that is, abdominal muscle contractions induced by colonic balloon distention was electrophysiologically measured in rats. Colonic permeability was determined in vivo by quantifying the absorbed Evans blue in colonic tissue for 15 minutes spectrophotometrically. KEY RESULTS Lipopolysaccharide (1 mg kg-1 ) subcutaneously (s.c.) reduced the threshold of VMR and increased colonic permeability. Losartan (5-25 mg kg-1 s.c.) for 3 days inhibited these changes in a dose-dependent manner. Moreover, repeated WAS (1 hour daily for 3 days) or intraperitoneal injection of CRF (50 µg kg-1 ) induced the similar visceral changes as LPS, which were also eliminated by losartan. These effects by losartan in LPS model were reversed by GW9662 (a peroxisome proliferator-activated receptor-γ [PPAR-γ] antagonist), NG -nitro-L-arginine methyl ester (a nitric oxide [NO] synthesis inhibitor), or naloxone (an opioid receptor antagonist). Moreover, it also inhibited by sulpiride (a dopamine D2 receptor antagonist) or domperidone (a peripheral dopamine D2 antagonist). CONCLUSION & INFERENCES Losartan prevented visceral allodynia and colonic hyperpermeability in rat IBS models. These actions may be PPAR-γ-dependent and also mediated by the NO, opioid, and dopamine D2 pathways. Losartan may be useful for IBS treatment.
Collapse
Affiliation(s)
- Tsukasa Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, Asahikawa, Japan.,Center for Medical Education, Asahikawa Medical University, Asahikawa, Japan
| | - Saori Miyagishi
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Rintaro Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, Asahikawa, Japan
| | - Kaoru Takakusaki
- Research Center for Brain Function and Medical Engineering, Asahikawa Medical University, Asahikawa, Japan
| | - Toshikatsu Okumura
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan.,Department of General Medicine, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
14
|
Butyrate inhibits visceral allodynia and colonic hyperpermeability in rat models of irritable bowel syndrome. Sci Rep 2019; 9:19603. [PMID: 31862976 PMCID: PMC6925246 DOI: 10.1038/s41598-019-56132-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/06/2019] [Indexed: 12/11/2022] Open
Abstract
Lipopolysaccharide (LPS) or repeated water avoidance stress (WAS) induces visceral allodynia and gut hyperpermeability via corticotropin-releasing factor (CRF) and proinflammatory cytokines, which is a rat irritable bowel syndrome (IBS) model. As butyrate is known to suppress the release of proinflammatory cytokine, we hypothesized that butyrate alleviates these colonic changes in IBS models. The visceral pain was assessed by electrophysiologically measuring the threshold of abdominal muscle contractions in response to colonic distention. Colonic permeability was determined by measuring the absorbance of Evans blue in colonic tissue. Colonic instillation of sodium butyrate (SB; 0.37-2.9 mg/kg) for 3 days inhibited LPS (1 mg/kg)-induced visceral allodynia and colonic hyperpermeability dose-dependently. Additionally, the visceral changes induced by repeated WAS (1 h for 3 days) or CRF (50 µg/kg) were also blocked by SB. These effects of SB in the LPS model were eliminated by compound C, an AMPK inhibitor, or GW9662, a PPAR-γ antagonist, NG-nitro-L-arginine methyl ester, a NO synthesis inhibitor, naloxone or sulpiride. SB attenuated visceral allodynia and colonic hyperpermeability in animal IBS models. These actions may be AMPK and PPAR-γ dependent and also mediated by the NO, opioid and central dopamine D2 pathways. Butyrate may be effective for the treatment of IBS.
Collapse
|
15
|
Larauche M, Moussaoui N, Biraud M, Bae W, Duboc H, Million M, Taché Y. Brain corticotropin-releasing factor signaling: Involvement in acute stress-induced visceral analgesia in male rats. Neurogastroenterol Motil 2019; 31:e13489. [PMID: 30298965 PMCID: PMC6347489 DOI: 10.1111/nmo.13489] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 09/14/2018] [Accepted: 09/17/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Water avoidance stress (WAS) induces a naloxone-independent visceral analgesia in male rats under non-invasive conditions of monitoring. The objective of the study was to examine the role of brain CRF signaling in acute stress-induced visceral analgesia (SIVA). METHODS Adult male Sprague-Dawley rats were chronically implanted with an intracerebroventricular (ICV) cannula. The visceromotor response (VMR) to graded phasic colorectal distension (CRD: 10, 20, 40, 60 mm Hg, 20 seconds, 4 minutes intervals) was monitored using manometry. The VMR to a first CRD (baseline) was recorded 5 minutes after an ICV saline injection, followed 1 hour later by ICV injection of either CRF (30, 100, or 300 ng and 1, 3, or 5 μg/rat) or saline and a second CRD, 5 minutes later. Receptor antagonists against CRF1 /CRF2 (astressin-B, 30 μg/rat), CRF2 (astressin2 -B, 10 μg/rat), oxytocin (tocinoic acid, 20 μg/rat), or vehicle were injected ICV 5 minutes before CRF (300 ng/rat, ICV) or 15 minutes before WAS (1 hour). KEY RESULTS ICV CRF (100 and 300 ng) reduced the VMR to CRD at 60 mm Hg by -36.6% ± 6.8% and -48.7% ± 11.7%, respectively, vs baseline (P < 0.001), while other doses had no effect and IP CRF (10 µg/kg) induced visceral hyperalgesia. Astressin-B and tocinoic acid injected ICV induced hyperalgesia and prevented the analgesic effect of ICV CRF (300 ng/rat) and WAS, while astressin2 -B only blocked WAS-induced SIVA. CONCLUSIONS & INFERENCES These data support a role for brain CRF signaling via CRF2 in SIVA in a model of WAS and CRD likely mediated by the activation of brain oxytocin pathway.
Collapse
Affiliation(s)
- M. Larauche
- Department of Medicine, UCLA, G Oppenheimer Center for
Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center,
Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of
Medicine, Los Angeles, CA, United States,VA Greater Los Angeles Healthcare System, Los Angeles, CA,
United States
| | - N. Moussaoui
- Department of Medicine, UCLA, G Oppenheimer Center for
Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center,
Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of
Medicine, Los Angeles, CA, United States,VA Greater Los Angeles Healthcare System, Los Angeles, CA,
United States,Present address: Inserm U1048/I2MC Obesity Research
Laboratory, 1 avenue Jean Poulhès BP 84225 31432 Toulouse Cedex 4,
France
| | - M. Biraud
- Department of Medicine, UCLA, G Oppenheimer Center for
Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center,
Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of
Medicine, Los Angeles, CA, United States,VA Greater Los Angeles Healthcare System, Los Angeles, CA,
United States,Present address: 1060 William Moore drive CVM Main
Building, RM C305, Raleigh, NC 27607, USA
| | - W.K. Bae
- Department of Medicine, UCLA, G Oppenheimer Center for
Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center,
Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of
Medicine, Los Angeles, CA, United States,VA Greater Los Angeles Healthcare System, Los Angeles, CA,
United States,Present address: Department of Internal Medicine, Ilsan
Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - H. Duboc
- Department of Medicine, UCLA, G Oppenheimer Center for
Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center,
Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of
Medicine, Los Angeles, CA, United States,VA Greater Los Angeles Healthcare System, Los Angeles, CA,
United States,Present address: CRI INSERM UMR 1149, University Paris
Diderot, Sorbonne Paris Cité and DHU Unity, APHP, F-75890 Paris, France
| | - M. Million
- Department of Medicine, UCLA, G Oppenheimer Center for
Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center,
Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of
Medicine, Los Angeles, CA, United States,VA Greater Los Angeles Healthcare System, Los Angeles, CA,
United States
| | - Y. Taché
- Department of Medicine, UCLA, G Oppenheimer Center for
Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center,
Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of
Medicine, Los Angeles, CA, United States,VA Greater Los Angeles Healthcare System, Los Angeles, CA,
United States
| |
Collapse
|
16
|
Averitt DL, Eidson LN, Doyle HH, Murphy AZ. Neuronal and glial factors contributing to sex differences in opioid modulation of pain. Neuropsychopharmacology 2019; 44:155-165. [PMID: 29973654 PMCID: PMC6235988 DOI: 10.1038/s41386-018-0127-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/21/2018] [Accepted: 05/27/2018] [Indexed: 12/12/2022]
Abstract
Morphine remains one of the most widely prescribed opioids for alleviation of persistent and/or severe pain; however, multiple preclinical and clinical studies report that morphine is less efficacious in females compared to males. Morphine primarily binds to the mu opioid receptor, a prototypical G-protein coupled receptor densely localized in the midbrain periaqueductal gray. Anatomical and physiological studies conducted in the 1960s identified the periaqueductal gray, and its descending projections to the rostral ventromedial medulla and spinal cord, as an essential descending inhibitory circuit mediating opioid-based analgesia. Remarkably, the majority of studies published over the following 30 years were conducted in males with the implicit assumption that the anatomical and physiological characteristics of this descending inhibitory circuit were comparable in females; not surprisingly, this is not the case. Several factors have since been identified as contributing to the dimorphic effects of opioids, including sex differences in the neuroanatomical and neurophysiological characteristics of the descending inhibitory circuit and its modulation by gonadal steroids. Recent data also implicate sex differences in opioid metabolism and neuroimmune signaling as additional contributing factors. Here we cohesively present these lines of evidence demonstrating a neural basis for sex differences in opioid modulation of pain, with a focus on the PAG as a sexually dimorphic core of descending opioid-induced inhibition and argue for the development of sex-specific pain therapeutics.
Collapse
Affiliation(s)
- Dayna L Averitt
- Department of Biology, Texas Woman's University, Denton, TX, 76204, USA
| | - Lori N Eidson
- Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Hillary H Doyle
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30303, USA
| | - Anne Z Murphy
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
17
|
Nozu T, Miyagishi S, Nozu R, Takakusaki K, Okumura T. Pioglitazone improves visceral sensation and colonic permeability in a rat model of irritable bowel syndrome. J Pharmacol Sci 2018; 139:46-49. [PMID: 30522964 DOI: 10.1016/j.jphs.2018.11.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/05/2018] [Accepted: 11/15/2018] [Indexed: 01/16/2023] Open
Abstract
Visceral hypersensitivity and impaired gut barrier with minor inflammation are considered to play an important role in the pathophysiology of irritable bowel syndrome (IBS). Since pioglitazone is known to have anti-inflammatory property, we hypothesized that pioglitazone is beneficial for treating IBS. In this study, the effect was tested in rat IBS models such as lipopolysaccharide or repeated water avoidance stress-induced visceral allodynia and increased colonic permeability. Pioglitazone blocked these visceral changes, and GW9662, a peroxisome proliferator-activated receptor gamma (PPAR-γ) antagonist fully reversed the effect by pioglitazone. These results suggest that PPAR-γ activation by pioglitazone may be useful for IBS treatment.
Collapse
Affiliation(s)
- Tsukasa Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan.
| | - Saori Miyagishi
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| | - Rintaro Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| | - Kaoru Takakusaki
- Research Center for Brain Function and Medical Engineering, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| | - Toshikatsu Okumura
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan; Department of General Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan
| |
Collapse
|
18
|
Aubert P, Oleynikova E, Rizvi H, Ndjim M, Le Berre-Scoul C, Grohard PA, Chevalier J, Segain JP, Le Drean G, Neunlist M, Boudin H. Maternal protein restriction induces gastrointestinal dysfunction and enteric nervous system remodeling in rat offspring. FASEB J 2018; 33:770-781. [DOI: 10.1096/fj.201800079r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Philippe Aubert
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Elena Oleynikova
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Hina Rizvi
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Marième Ndjim
- Institute National de la Recherche Agronomique (INRA) Unité Mixte de Recherche 1280Physiologie des Adaptations Nutritionnelles (PhAN)Institut des Maladies de l'Appareil Digestif Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Catherine Le Berre-Scoul
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Pierre Antoine Grohard
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Julien Chevalier
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Jean-Pierre Segain
- Institute National de la Recherche Agronomique (INRA) Unité Mixte de Recherche 1280Physiologie des Adaptations Nutritionnelles (PhAN)Institut des Maladies de l'Appareil Digestif Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Gwenola Le Drean
- Institute National de la Recherche Agronomique (INRA) Unité Mixte de Recherche 1280Physiologie des Adaptations Nutritionnelles (PhAN)Institut des Maladies de l'Appareil Digestif Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Michel Neunlist
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Helene Boudin
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| |
Collapse
|
19
|
Opposing Roles of Estradiol and Testosterone on Stress-Induced Visceral Hypersensitivity in Rats. THE JOURNAL OF PAIN 2018; 19:764-776. [PMID: 29496640 DOI: 10.1016/j.jpain.2018.02.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 01/30/2018] [Accepted: 02/14/2018] [Indexed: 12/19/2022]
Abstract
Chronic stress produces maladaptive pain responses, manifested as alterations in pain processing and exacerbation of chronic pain conditions including irritable bowel syndrome. Female predominance, especially during reproductive years, strongly suggests a role of gonadal hormones. However, gonadal hormone modulation of stress-induced pain hypersensitivity is not well understood. In the present study, we tested the hypothesis that estradiol is pronociceptive and testosterone is antinociceptive in a model of stress-induced visceral hypersensitivity (SIVH) in rats by recording the visceromotor response to colorectal distention after a 3-day forced swim (FS) stress paradigm. FS induced visceral hypersensitivity that persisted at least 2 weeks in female, but only 2 days in male rats. Ovariectomy blocked and orchiectomy facilitated SIVH. Furthermore, estradiol injection in intact male rats increased SIVH and testosterone in intact female rats attenuated SIVH. Western blot analyses indicated estradiol increased excitatory glutamate ionotropic receptor NMDA type subunit 1 expression and decreased inhibitory metabotropic glutamate receptor 2 expression after FS in male thoracolumbar spinal cord. In addition, the presence of estradiol during stress increased spinal brain-derived neurotrophic factor (BDNF) expression independent of sex. In contrast, testosterone blocked the stress-induced increase in BDNF expression in female rats. These data suggest that estradiol facilitates and testosterone attenuates SIVH by modulating spinal excitatory and inhibitory glutamatergic receptor expression. PERSPECTIVE SIVH is more robust in female rats. Estradiol facilitates whereas testosterone dampens the development of SIVH. This could partially explain the greater prevalence of certain chronic visceral pain conditions in women. An increase in spinal BDNF is concomitant with increased stress-induced pain. Pharmaceutical interventions targeting this molecule could provide promising alleviation of SIVH in women.
Collapse
|
20
|
Ferdousi M, Finn DP. Stress-induced modulation of pain: Role of the endogenous opioid system. PROGRESS IN BRAIN RESEARCH 2018; 239:121-177. [DOI: 10.1016/bs.pbr.2018.07.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
21
|
Abstract
More than eighty years after Hans Selye (1907-1982) first developed a concept describing how different types of environmental stressors affect physiological functions and promote disease development (called the "general adaptation syndrome") in 1936, we herein review advances in theoretical, mechanistic, and clinical knowledge in stress research, especially in the area of gastroenterology, and summarize progress and future perspectives arising from an interdisciplinary psychoneurobiological framework in which genetics, epigenetics, and other advanced ( omics) technologies in the last decade continue to refine knowledge about how stress affects the brain-gut axis in health and gastrointestinal disease. We demonstrate that neurobiological stress research continues to be a driving force for scientific progress in gastroenterology and related clinical areas, inspiring translational research from animal models to clinical applications, while highlighting some areas that remain incompletely understood, such as the roles of sex/gender and gut microbiota in health and disease. Future directions of research should include not only the genetics of the stress response and resilience but also epigenetic contributions.
Collapse
Affiliation(s)
- Sigrid Elsenbruch
- Institute of Medical Psychology & Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Paul Enck
- Department of Internal Medicine VI: Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
22
|
Probiotics reduce repeated water avoidance stress-induced colonic microinflammation in Wistar rats in a sex-specific manner. PLoS One 2017; 12:e0188992. [PMID: 29244820 PMCID: PMC5731730 DOI: 10.1371/journal.pone.0188992] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 11/16/2017] [Indexed: 12/21/2022] Open
Abstract
The colonic response to stress is greater in female rats than in male rats. The aim of this study was to evaluate the effect of probiotics in the repeated water avoidance stress (rWAS)-induced colonic microinflammation model of Wistar rats in a sex-specific manner. The three groups (no-stress, WAS, and WAS with probiotics) were exposed to r-WAS for 1 h daily for 10 days, and Lactobacillus farciminis was administered by oral gavage for 10 days to animals in the probiotics group. The visceromotor response (VMR) to colorectal distension (CRD) was assessed using a barostat and noninvasive manometry before and after WAS exposure. Immunohistochemistry for mast cells and real-time polymerase chain reaction (RT-PCR) for detection of mucosal cytokines were performed using distal colon tissue after the animals were sacrificed. Significant reduction of VMR to CRD (visceral analgesia) was observed at 60 mmHg in the female WAS group (P = 0.045), but not in males. In addition, the female WAS with probiotics group showed a significantly lower colonic mucosal mast cell count in comparison to the female WAS group (P = 0.013), but this phenomenon was not observed in the male group. The colonic mucosal mRNA levels of interferon-γ (IFNR), tumor necrosis factor-α (TNFA), interleukin (IL) 6, and IL17 were higher in the female WAS group than in the male WAS group. The mRNA levels of IFNR, TNFA, and IL6 were significantly decreased in WAS females who received probiotics (all P < 0.050). In conclusion, rWAS is induced in a sex-specific manner. A 10-day-long treatment with L. farciminis is an effective therapy for rWAS-induced colonic microinflammation in female rates, but not in male rats.
Collapse
|
23
|
Nozu T, Miyagishi S, Nozu R, Takakusaki K, Okumura T. Repeated water avoidance stress induces visceral hypersensitivity: Role of interleukin-1, interleukin-6, and peripheral corticotropin-releasing factor. J Gastroenterol Hepatol 2017; 32:1958-1965. [PMID: 28299830 DOI: 10.1111/jgh.13787] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 03/03/2017] [Accepted: 03/13/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIM Repeated water avoidance stress (WAS) induces visceral hypersensitivity. Additionally, it is also known to activate corticotropin-releasing factor (CRF), mast cells, and pro-inflammatory cytokines systems, but their precise roles on visceral sensation have not been determined definitely. The aim of the study was to explore this issue. METHODS Abdominal muscle contractions induced by colonic balloon distention, that is, visceromotor response (VMR) was detected electrophysiologically in conscious rats. WAS or sham stress as control for 1 h daily was loaded, and the threshold of VMR was determined before and at 24 h after the stress. RESULTS Repeated WAS for three consecutive days reduced the threshold of VMR, but sham stress did not induce any change. Astressin, a CRF receptor antagonist (50 μg/kg) intraperitoneally (ip) at 10 min before each WAS session, prevented the visceral allodynia, but the antagonist (200 μg/kg) ip at 30 min and 15 h before measurement of the threshold after completing 3-day stress session did not modify the response. Ketotifen, a mast cell stabilizer (3 mg/kg), anakinra, an interleukin (IL)-1 receptor antagonist (20 mg/kg) or IL-6 antibody (16.6 μg/kg) ip for two times before the measurement abolished the response. CONCLUSIONS Repeated WAS for three consecutive days induced visceral allodynia, which was mediated through mast cells, IL-1, and IL-6 pathways. Inhibition of peripheral CRF signaling prevented but did not reverse this response, suggesting that peripheral CRF may be an essential trigger but may not contribute to the maintenance of repeated WAS-induced visceral allodynia.
Collapse
Affiliation(s)
- Tsukasa Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Saori Miyagishi
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Rintaro Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Kaoru Takakusaki
- Research Center for Brain Function and Medical Engineering, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Toshikatsu Okumura
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| |
Collapse
|
24
|
Romero A, García-Carmona JA, Laorden ML, Puig MM. Role of CRF1 receptor in post-incisional plasma extravasation and nociceptive responses in mice. Toxicol Appl Pharmacol 2017; 332:121-128. [DOI: 10.1016/j.taap.2017.04.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/12/2017] [Accepted: 04/19/2017] [Indexed: 12/30/2022]
|
25
|
Rolland-Fourcade C, Denadai-Souza A, Cirillo C, Lopez C, Jaramillo JO, Desormeaux C, Cenac N, Motta JP, Larauche M, Taché Y, Berghe PV, Neunlist M, Coron E, Kirzin S, Portier G, Bonnet D, Alric L, Vanner S, Deraison C, Vergnolle N. Epithelial expression and function of trypsin-3 in irritable bowel syndrome. Gut 2017; 66:1767-1778. [PMID: 28096305 PMCID: PMC5595105 DOI: 10.1136/gutjnl-2016-312094] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 12/26/2016] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Proteases are key mediators of pain and altered enteric neuronal signalling, although the types and sources of these important intestinal mediators are unknown. We hypothesised that intestinal epithelium is a major source of trypsin-like activity in patients with IBS and this activity signals to primary afferent and enteric nerves and induces visceral hypersensitivity. DESIGN Trypsin-like activity was determined in tissues from patients with IBS and in supernatants of Caco-2 cells stimulated or not. These supernatants were also applied to cultures of primary afferents. mRNA isoforms of trypsin (PRSS1, 2 and 3) were detected by reverse transcription-PCR, and trypsin-3 protein expression was studied by western blot analysis and immunohistochemistry. Electrophysiological recordings and Ca2+ imaging in response to trypsin-3 were performed in mouse primary afferent and in human submucosal neurons, respectively. Visceromotor response to colorectal distension was recorded in mice administered intracolonically with trypsin-3. RESULTS We showed that stimulated intestinal epithelial cells released trypsin-like activity specifically from the basolateral side. This activity was able to activate sensory neurons. In colons of patients with IBS, increased trypsin-like activity was associated with the epithelium. We identified that trypsin-3 was the only form of trypsin upregulated in stimulated intestinal epithelial cells and in tissues from patients with IBS. Trypsin-3 was able to signal to human submucosal enteric neurons and mouse sensory neurons, and to induce visceral hypersensitivity in vivo, all by a protease-activated receptor-2-dependent mechanism. CONCLUSIONS In IBS, the intestinal epithelium produces and releases the active protease trypsin-3, which is able to signal to enteric neurons and to induce visceral hypersensitivity.
Collapse
Affiliation(s)
| | | | - Carla Cirillo
- Laboratory for Enteric Neuroscience (LENS), TARGID, University of Leuven, Leuven, Belgium
| | - Cintya Lopez
- Gastrointestinal Diseases Research Unit, , General Hospital, Queen's University School of Medicine, Kingston, Ontario, Canada
| | - Josue Obed Jaramillo
- Gastrointestinal Diseases Research Unit, , General Hospital, Queen's University School of Medicine, Kingston, Ontario, Canada
| | - Cleo Desormeaux
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Nicolas Cenac
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Jean-Paul Motta
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Muriel Larauche
- Oppenheimer Family Center for Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, UCLA David Geffen School of Medicine, Los Angeles, California, USA
| | - Yvette Taché
- Oppenheimer Family Center for Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, UCLA David Geffen School of Medicine, Los Angeles, California, USA
| | - Pieter Vanden Berghe
- Laboratory for Enteric Neuroscience (LENS), TARGID, University of Leuven, Leuven, Belgium
| | - Michel Neunlist
- Inserm, UMR913, Nantes, France,Nantes University, Nantes, France,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - Emmanuel Coron
- Inserm, UMR913, Nantes, France,Nantes University, Nantes, France,Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Hopital Hôtel-Dieu, Nantes, France
| | - Sylvain Kirzin
- Department of Internal Medicine and Digestive Diseases, Pole Digestif, CHU Toulouse, Toulouse, France
| | - Guillaume Portier
- Department of Internal Medicine and Digestive Diseases, Pole Digestif, CHU Toulouse, Toulouse, France
| | - Delphine Bonnet
- Department of Internal Medicine and Digestive Diseases, Pole Digestif, CHU Toulouse, Toulouse, France
| | - Laurent Alric
- Department of Internal Medicine and Digestive Diseases, Pole Digestif, CHU Toulouse, Toulouse, France
| | - Stephen Vanner
- Gastrointestinal Diseases Research Unit, , General Hospital, Queen's University School of Medicine, Kingston, Ontario, Canada
| | - Celine Deraison
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France,Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
26
|
He YQ, Lang XQ, Lin L, Ji L, Yuan XY, Chen Q, Ran YM, Chen HS, Li L, Wang JM, Wang ZG, Gregersen H, Zou DW, Liang HP, Yang M. P2X3 receptor-mediated visceral hyperalgesia and neuronal sensitization following exposure to PTSD-like stress in the dorsal root ganglia of rats. Neurogastroenterol Motil 2017; 29. [PMID: 27781340 DOI: 10.1111/nmo.12976] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 09/22/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND Patients with posttraumatic stress disorder (PTSD) often share co-morbidity with chronic pain conditions. Recent studies suggest a role of P2X3 receptors and ATP signaling in pain conditions. However, the underlying mechanisms of visceral hyperalgesia following exposure to PTSD-like stress conditions remain unclarified. METHODS The behavior and hormones relevant for PTSD were studied. Visceromotor responses (VMR) and the abdominal withdrawal reflexes (AWR) to colorectal distention (CRD) were recorded to determine P2X3-receptor-mediated alteration of hyperalgesia following single-prolonged stress (SPS) exposure. Immunofluorescence, Western blotting, and patch-clamp were used. KEY RESULTS The escape latency, adrenocorticotropic hormone and cortisol were increased on days 7-14. Visceromotor responses and AWR was reduced at day 1 in SPS rats but increased to higher levels than in controls after exposure to day 7. Intrathecal administration of the P2X3-receptor antagonist TNP-ATP abolished the CRD response. Based on immunofluorescence and Western blotting analysis, SPS-treated rats exhibited reduced P2X3 expression in dorsal root ganglia (DRG) after day 1 compared with controls. P2X3 expression in DRG was enhanced on day 7 after SPS and the increase of the P2X3 expression was maintained on day 14 and 21 compared with controls. The P2X3-receptor agonist α,β-me ATP (10 μM) induced a fast desensitizing inward current in DRG neurons of both control and SPS-treated rats. The average peak current densities in SPS-treated group were increased 3.6-fold. TNP-ATP (100 nM) markedly blocked all fast α,β-me ATP-induced inward currents in the DRG neurons both in control and SPS-treated rats. CONCLUSIONS & INFERENCES The data indicate an important role of P2X3 signaling in visceral hyperalgesia following PTSD-like stress.
Collapse
Affiliation(s)
- Y-Q He
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - X-Q Lang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - L Lin
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - L Ji
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - X-Y Yuan
- Department of Gastroenterology, The Ninth People's Hospital of Chongqing, Chongqing, China
| | - Q Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Y-M Ran
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - H-S Chen
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - L Li
- Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - J-M Wang
- Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Z-G Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China.,Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - H Gregersen
- GIOME and the Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, China
| | - D-W Zou
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - H-P Liang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China.,Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - M Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China.,Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
27
|
Pitcher MH, Gonzalez-Cano R, Vincent K, Lehmann M, Cobos EJ, Coderre TJ, Baeyens JM, Cervero F. Mild Social Stress in Mice Produces Opioid-Mediated Analgesia in Visceral but Not Somatic Pain States. THE JOURNAL OF PAIN 2017; 18:716-725. [PMID: 28219667 DOI: 10.1016/j.jpain.2017.02.422] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 01/14/2017] [Accepted: 02/02/2017] [Indexed: 12/30/2022]
Abstract
Visceral pain has a greater emotional component than somatic pain. To determine if the stress-induced analgesic response is differentially expressed in visceral versus somatic pain states, we studied the effects of a mild social stressor in either acute visceral or somatic pain states in mice. We show that the presence of an unfamiliar conspecific mouse (stranger) in an adjacent cubicle of a standard transparent observation box produced elevated plasma corticosterone levels compared with mice tested alone, suggesting that the mere presence of a stranger is stressful. We then observed noxious visceral or somatic stimulation-induced nociceptive behavior in mice tested alone or in mildly stressful conditions (ie, beside an unfamiliar stranger). Compared with mice tested alone, the presence of a stranger produced a dramatic opioid-dependent reduction in pain behavior associated with visceral but not somatic pain. This social stress-induced reduction of visceral pain behavior relied on visual but not auditory/olfactory cues. These findings suggest that visceral pain states may provoke heightened responsiveness to mild stressors, an effect that could interfere with testing outcomes during simultaneous behavioral testing of multiple rodents. PERSPECTIVE In mice, mild social stress due to the presence of an unfamiliar conspecific mouse reduces pain behavior associated with noxious visceral but not somatic stimulation, suggesting that stress responsiveness may be enhanced in visceral pain versus somatic pain states.
Collapse
Affiliation(s)
- Mark H Pitcher
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland.
| | | | - Kathleen Vincent
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
| | - Michael Lehmann
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Enrique J Cobos
- Department of Pharmacology, University of Granada, Granada, Spain
| | - Terence J Coderre
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
| | - José M Baeyens
- Department of Pharmacology, University of Granada, Granada, Spain
| | - Fernando Cervero
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
28
|
Tsang SW, Auyeung KKW, Bian ZX, Ko JKS. Pathogenesis, Experimental Models and Contemporary Pharmacotherapy of Irritable Bowel Syndrome: Story About the Brain-Gut Axis. Curr Neuropharmacol 2017; 14:842-856. [PMID: 27009115 PMCID: PMC5333584 DOI: 10.2174/1570159x14666160324144154] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 02/07/2016] [Accepted: 03/22/2016] [Indexed: 11/22/2022] Open
Abstract
Background Although the precise pathophysiology of irritable bowel syndrome (IBS) remains unknown, it is generally considered to be a disorder of the brain-gut axis, representing the disruption of communication between the brain and the digestive system. The present review describes advances in understanding the pathophysiology and experimental approaches in studying IBS, as well as providing an update of the therapies targeting brain-gut axis in the treatment of the disease. Methods Causal factors of IBS are reviewed. Following this, the preclinical experimental models of IBS will be introduced. Besides, both current and future therapeutic approaches of IBS will be discussed. Results When signal of the brain-gut axis becomes misinterpreted, it may lead to dysregulation of both central and enteric nervous systems, altered intestinal motility, increased visceral sensitivity and consequently contributing to the development of IBS. Interference of the brain-gut axis can be modulated by various psychological and environmental factors. Although there is no existing animal experiment that can represent this complex multifactorial disease, these in vivo models are clinically relevant readouts of gastrointestinal functions being essential to the identification of effective treatments of IBS symptoms as well as their molecular targets. Understanding the brain-gut axis is essential in developing the effective therapy for IBS. Therapies include improvement of GI motor functions, relief of visceral hypersensitivity and pain, attenuation of autonomic dysfunctions and suppression of mucosal immune activation. Conclusion Target-oriented therapies that provide symptomatic, psychological and physiological benefits could surely help to improve the quality of life of IBS patients.
Collapse
Affiliation(s)
| | | | | | - J K S Ko
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Hong Kong SAR, China
| |
Collapse
|
29
|
Lee JY, Kim N, Kim YS, Nam RH, Ham MH, Lee HS, Jo W, Shim Y, Choi YJ, Yoon H, Shin CM, Lee DH. Repeated Water Avoidance Stress Alters Mucosal Mast Cell Counts, Interleukin-1β Levels with Sex Differences in the Distal Colon of Wistar Rats. J Neurogastroenterol Motil 2016; 22:694-704. [PMID: 27466288 PMCID: PMC5056580 DOI: 10.5056/jnm16007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 05/23/2016] [Accepted: 06/14/2016] [Indexed: 12/13/2022] Open
Abstract
Background/Aims This study was aimed at evaluating differences in the effects of repeated water avoidance stress (rWAS) on colonic movement, mucosal mast cell counts, cytokine levels, and visceromotor response (VMR) to colorectal distension (CRD) in rats of both sexes. Methods Wistar rats were divided into stress and no-stress groups. Rats in the stress group were exposed to rWAS (1 hr/day) for 10 days. Mucosal mast cells were immunohistochemically stained with anti-mast cell tryptase antibody and counted. The colonic mucosal cytokine levels were measured with enzyme-linked immunosorbent assay. The VMR to CRD (visceral analgesia) was assessed by using a barostat and noninvasive manometry. Results The mean number of fecal pellets in the rWAS group increased significantly as compared with that in the no-stress group in both sexes. After adjustment for body weight, the female rats had a significantly higher pellet output than the male rats. The mucosal mast cell count of the female rWAS group was higher than that of the male rWAS group (13.0 ± 0.9 vs 8.8 ± 0.6; P < 0.001). The colonic mucosal interleukin-1β level was also higher only in the female rats of the rWAS group than in those of the no-stress group. On days 10 and 11, a decrease in VMR to CRD was observed at 40 and 60 mmHg in both sexes of the rWAS group, without a sex-based difference. Conclusions The colonic response to stress appeared to be more sensitive in the female rats than in the male rats. However, stress-induced visceral analgesia had no sex-related difference and the underlying mechanism needs to be further evaluated.
Collapse
Affiliation(s)
- Ju Yup Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea.,Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Yong Sung Kim
- Department of Gastroenterology and Digestive Disease Research Institute, Wonkwang University School of Medicine, Iksan, Jeollabuk-do, Korea
| | - Ryoung Hee Nam
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Min Hee Ham
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Wonjun Jo
- Department of BioNano Technology and Gachon BioNano Research Institute, Gachon University, Seongnam, Gyeonggi-do, Korea
| | - Youngkwang Shim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Yoon Jin Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| |
Collapse
|
30
|
Cao DY, Bai G, Ji Y, Karpowicz JM, Traub RJ. EXPRESS: Histone hyperacetylation modulates spinal type II metabotropic glutamate receptor alleviating stress-induced visceral hypersensitivity in female rats. Mol Pain 2016; 12:1744806916660722. [PMID: 27385724 PMCID: PMC4956148 DOI: 10.1177/1744806916660722] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 06/13/2016] [Accepted: 06/27/2016] [Indexed: 12/14/2022] Open
Abstract
Stress is often a trigger to exacerbate chronic pain including visceral hypersensitivity associated with irritable bowel syndrome, a female predominant functional bowel disorder. Epigenetic mechanisms that mediate stress responses are a potential target to interfere with visceral pain. The purpose of this study was to examine the effect of a histone deacetylase inhibitor, suberoylanilide hydroxamic acid, on visceral hypersensitivity induced by a subchronic stressor in female rats and to investigate the involvement of spinal glutamate receptors. Three daily sessions of forced swim induced visceral hypersensitivity. Intrathecal suberoylanilide hydroxamic acid prevented or reversed the stress-induced visceral hypersensitivity, increased spinal histone 3 acetylation and increased mGluR2 and mGluR3 expression. Chromatin immunoprecipitation (ChIP) analysis revealed enrichment of H3K9Ac and H3K18Ac at several promoter Grm2 and Grm3 regions. The mGluR2/3 antagonist LY341495 reversed the inhibitory effect of suberoylanilide hydroxamic acid on the stress-induced visceral hypersensitivity. In surprising contrast, stress and/or suberoylanilide hydroxamic acid had no effect on spinal NMDA receptor expression or function. These data reveal histone modification modulates mGluR2/3 expression in the spinal cord to attenuate stressinduced visceral hypersensitivity. HDAC inhibitors may provide a potential approach to relieve visceral hypersensitivity associated with irritable bowel syndrome.
Collapse
Affiliation(s)
| | - Guang Bai
- University of Maryland School of Dentistry
| | - Yaping Ji
- University of Maryland School of Dentistry
| | - Jane M Karpowicz
- University of Maryland School of DentistryUniversity of Maryland School of DentistryUniversity of Maryland School of Dentistry
| | - Richard J Traub
- University of Maryland School of DentistryUniversity of Maryland School of DentistryUniversity of Maryland School of Dentistry
| |
Collapse
|
31
|
Mondelaers SU, Theofanous SA, Florens MV, Perna E, Aguilera-Lizarraga J, Boeckxstaens GE, Wouters MM. Effect of genetic background and postinfectious stress on visceral sensitivity in Citrobacter rodentium-infected mice. Neurogastroenterol Motil 2016; 28:647-58. [PMID: 26728091 DOI: 10.1111/nmo.12759] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 11/24/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND Infectious gastroenteritis is a major risk factor to develop postinfectious irritable bowel syndrome (PI-IBS). It remains unknown why only a subgroup of infected individuals develops PI-IBS. We hypothesize that immunogenetic predisposition is an important risk factor. Hence, we studied the effect of Citrobacter rodentium infection on visceral sensitivity in Th1-predominant C57BL/6 and Th2-predominant Balb/c mice. METHODS Eight-week-old mice were gavaged with C. rodentium, followed by 1 h of water avoidance stress (WAS) at 5 weeks PI. At 10, 14 days, and 5 weeks PI, samples were assessed for histology and inflammatory gene expression by RT-qPCR. Visceral sensitivity was evaluated by visceromotor response recordings (VMR) to colorectal distension. KEY RESULTS Citrobacter rodentium evoked a comparable colonic inflammatory response at 14 days PI characterized by increased crypt length and upregulation of Th1/Th17 cytokine mRNA levels (puncorrected < 0.05) in both C57BL/6 and Balb/c mice. At 5 weeks PI, inflammatory gene mRNA levels returned to baseline in both strains. The VMR was maximal at 14 days PI in C57BL/6 (150 ± 47%; p = 0.02) and Balb/c mice (243 ± 52%; p = 0.03). At 3 weeks PI, the VMR remained increased in Balb/c (176 ± 23%; p = 0.02), but returned to baseline in C57BL/6 mice. At 5 weeks PI, WAS could not re-introduce visceral hypersensitivity (VHS). CONCLUSIONS & INFERENCES Citrobacter rodentium infection induces transient VHS in C57BL/6 and Balb/c mice, which persisted 1 week longer in Balb/c mice. Although other strain-related differences may contribute, a Th2 background may represent a risk factor for prolonged PI-VHS. As PI-VHS is transient, other factors are crucial for persistent VHS development as observed in PI-IBS.
Collapse
Affiliation(s)
- S U Mondelaers
- Translational Research Center for Gastrointestinal Disorders, Dept. of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - S A Theofanous
- Translational Research Center for Gastrointestinal Disorders, Dept. of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - M V Florens
- Translational Research Center for Gastrointestinal Disorders, Dept. of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - E Perna
- Translational Research Center for Gastrointestinal Disorders, Dept. of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - J Aguilera-Lizarraga
- Translational Research Center for Gastrointestinal Disorders, Dept. of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - G E Boeckxstaens
- Translational Research Center for Gastrointestinal Disorders, Dept. of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - M M Wouters
- Translational Research Center for Gastrointestinal Disorders, Dept. of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| |
Collapse
|
32
|
Nozu T, Miyagishi S, Nozu R, Takakusaki K, Okumura T. Water avoidance stress induces visceral hyposensitivity through peripheral corticotropin releasing factor receptor type 2 and central dopamine D2 receptor in rats. Neurogastroenterol Motil 2016; 28:522-31. [PMID: 26662216 DOI: 10.1111/nmo.12747] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 11/11/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Water avoidance stress (WAS) is reported to induce functional changes in visceral sensory function in rodents, but the results which have been demonstrated so far are not consistent, i.e., hypersensitivity or hyposensitivity. We determined the effect of WAS on visceral sensation and evaluated the mechanisms of the action. METHODS Visceral sensation was assessed by abdominal muscle contractions induced by colonic balloon distention, i.e., visceromotor response (VMR), measured electrophysiologically in conscious rats. The electromyogram electrodes were acutely implanted under anesthesia on the day of the experiment. The threshold of VMR was measured before and after WAS for 1 h. To explore the mechanisms of WAS-induced response, drugs were administered 10 min prior to the initiation of WAS. KEY RESULTS WAS significantly increased the threshold of VMR, and this effect was no longer detected at 24 h after. Intraperitoneal injection of astressin2 -B (200 μg/kg), a corticotropin releasing factor (CRF) receptor type 2 antagonist abolished the response by WAS. Subcutaneous (sc) injection of sulpiride (200 mg/kg), a dopamine D2 receptor antagonist blocked the response, while sc domperidone (10 mg/kg), a peripheral dopamine D2 receptor antagonist did not alter it. Naloxone (1 mg/kg, sc), an opioid antagonist did not modify it either. CONCLUSIONS & INFERENCES WAS induced visceral hyposensitivity through peripheral CRF receptor type 2 and central dopamine D2 receptor, but not through opioid pathways. As altered pain inhibitory system was reported to be observed in the patients with irritable bowel syndrome, CRF and dopamine signaling might contribute to the pathophysiology.
Collapse
Affiliation(s)
- T Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, Asahikawa, Japan
| | - S Miyagishi
- Department of General Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - R Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, Asahikawa, Japan
| | - K Takakusaki
- Research Center for Brain Function and Medical Engineering, Asahikawa Medical University, Asahikawa, Japan
| | - T Okumura
- Department of General Medicine, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
33
|
Gil DW, Wang J, Gu C, Donello JE, Cabrera S, Al-Chaer ED. Role of sympathetic nervous system in rat model of chronic visceral pain. Neurogastroenterol Motil 2016; 28:423-31. [PMID: 26670784 DOI: 10.1111/nmo.12742] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 10/31/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Changes in central pain modulation have been implicated in generalized pain syndromes such as irritable bowel syndrome (IBS). We have previously demonstrated that reduced descending inhibition unveils a role of sympathoneuronal outflow in decreasing peripheral sensory thresholds, resulting in stress-induced hyperalgesia. We investigated whether sympathetic nervous system (SNS) exacerbation of pain sensation when central pain inhibition is reduced is relevant to chronic pain disorders using a rat colon irritation (CI) model of chronic visceral hypersensitivity with hallmarks of IBS. METHODS Rats were treated to a series of colorectal balloon distensions (CRD) as neonates resulting in visceral and somatic hypersensitivity and altered stool function that persists into adulthood. The visceral sensitivity was assessed by recording electromyographic (EMG) responses to CRD. Somatic sensitivity was assessed by paw withdrawal thresholds to radiant heat. The effects on the hypersensitivity of (i) inhibiting sympathoneuronal outflow with pharmacological and surgical interventions and (ii) enhancing the outflow with water avoidance stress (WAS) were tested. KEY RESULTS The alpha2-adrenergic agonist, clonidine, and the alpha1-adrenergic antagonist, prazosin, reduced the visceral hypersensitivity and WAS enhanced the pain. Chemical sympathectomy with guanethidine and surgical sympathectomy resulted in a loss of the chronic visceral hypersensitivity. CONCLUSIONS & INFERENCES The results support a role of the SNS in driving the chronic visceral and somatic hypersensitivity seen in CI rats. The findings further suggest that treatments that decrease sympathetic outflow or block activation of adrenergic receptors on sensory nerves could be beneficial in the treatment of generalized pain syndromes.
Collapse
Affiliation(s)
| | - J Wang
- University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, USA
| | - C Gu
- University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, USA
| | | | | | - E D Al-Chaer
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut (AUB), Beirut, Lebanon.,Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, USA
| |
Collapse
|
34
|
Houghton LA, Heitkemper M, Crowell M, Emmanuel A, Halpert A, McRoberts JA, Toner B. Age, Gender and Women's Health and the Patient. Gastroenterology 2016; 150:S0016-5085(16)00183-9. [PMID: 27144622 DOI: 10.1053/j.gastro.2016.02.017] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 02/09/2016] [Indexed: 12/15/2022]
Abstract
Patients with functional gastrointestinal disorders (FGIDs) often experience distress, reduced quality of life, a perceived lack of validation, and an unsatisfactory experience with health care providers. A health care provider can provide the patient with a framework in which to understand and legitimize their symptoms, remove self-doubt or blame, and identify factors that contribute to symptoms that the patient can influence or control. This framework is implemented with the consideration of important factors that impact FGIDs, such as gender, age, society, and the patient's perspective. Although the majority of FGIDs, including globus, rumination syndrome, IBS, bloating, constipation, functional abdominal pain, sphincter of Oddi dyskinesia, pelvic floor dysfunction, and extra-intestinal manifestations, are more prevalent in women than men, functional chest pain, dyspepsia, vomiting, and anorectal pain do not appear to vary by gender. Studies suggest sex differences in somatic but not visceral pain perception, motility, and central processing of visceral pain; although further research is required in autonomic nervous system dysfunction, genetics and immunologic/microbiome. Gender differences in response to psychological treatments, antidepressants, fiber, probiotics, and anticholinergics have not been adequately studied. However, a greater clinical response to 5-HT3 antagonists but not 5-HT4 agonists has been reported in women compared with men.
Collapse
Affiliation(s)
- Lesley A Houghton
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA; Centre for Gastrointestinal Sciences, University of Manchester, Manchester, UK.
| | | | - Michael Crowell
- Division of Gastroenterology and Hepatology Mayo Clinic, Scottsdale, Arizona, USA
| | | | | | | | | |
Collapse
|
35
|
Xu YY, Ge JF, Qin G, Peng YN, Zhang CF, Liu XR, Liang LC, Wang ZZ, Chen FH, Li J. Acute, but not chronic, stress increased the plasma concentration and hypothalamic mRNA expression of NUCB2/nesfatin-1 in rats. Neuropeptides 2015; 54:47-53. [PMID: 26297350 DOI: 10.1016/j.npep.2015.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 08/02/2015] [Accepted: 08/02/2015] [Indexed: 01/03/2023]
Abstract
Nesfatin-1, a newly discovered satiety peptide, has recently been reported to be involved in the stress response. Stress-induced expression of nesfatin-1 has been reported and few studies focus on its expression in the hypothalamus, which is the center of the stress response. To test our hypothesis that peripheral and hypothalamic nesfatin-1 overexpression should play an important role in the stress response and the associated hyperactivity of hypothalamic-pituitary-adrenal (HPA) axis, acute stress (AS) was induced using water avoidance stress (WAS), and chronic unpredictable mild stress (CUMS) was also induced using 3 consecutive weeks of 7 different stressors. The behavior of CUMS rats was evaluated by an open field test (OFT), sucrose preference test (SPT), and forced swimming test (FST). The activity of the HPA axis was detected by measurement of the plasma corticosterone concentration and hypothalamic mRNA expression of corticotropin-releasing-hormone (CRH). The plasma concentration and hypothalamic mRNA expression of nesfatin-1 were measured with an enzyme-linked immunosorbent assay (ELISA) and real-time fluorescent quantitative PCR, respectively. The results showed that both AS and CUMS increased the plasma corticosterone concentration and hypothalamic CRH mRNA expression. Depression-like behavior was induced in CUMS rats, as indicated by a decreased movement distance, frequency of rearing and grooming in the OFT, and sucrose preference index and increased immobility in the FST. Moreover, the AS rats showed increased plasma concentration and hypothalamic mRNA expression of nesfatin-1, which were positively correlated with the plasma corticosterone concentration and hypothalamic CRH expression, respectively. These results indicated that acute stress, but not chronic stress, increased the plasma concentration and hypothalamic mRNA expression of NUCB2/nesfatin-1 in rats.
Collapse
Affiliation(s)
- Ya-Yun Xu
- School of Pharmacy, Anhui Medical University, Hefei, 230032 Anhui, China
| | - Jin-Fang Ge
- School of Pharmacy, Anhui Medical University, Hefei, 230032 Anhui, China.
| | - Gan Qin
- School of Pharmacy, Anhui Medical University, Hefei, 230032 Anhui, China
| | - Yao-Nan Peng
- The First Clinical College of Anhui Medical University, Hefei, 230032 Anhui, China
| | - Chao-Feng Zhang
- The First Clinical College of Anhui Medical University, Hefei, 230032 Anhui, China
| | - Xing-Rui Liu
- The First Clinical College of Anhui Medical University, Hefei, 230032 Anhui, China
| | - Li-Chuan Liang
- The First Clinical College of Anhui Medical University, Hefei, 230032 Anhui, China
| | - Zhong-Zheng Wang
- The First Clinical College of Anhui Medical University, Hefei, 230032 Anhui, China
| | - Fei-Hu Chen
- School of Pharmacy, Anhui Medical University, Hefei, 230032 Anhui, China.
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei, 230032 Anhui, China
| |
Collapse
|
36
|
Nozu T, Kumei S, Miyagishi S, Takakusaki K, Okumura T. Colorectal distention induces acute and delayed visceral hypersensitivity: role of peripheral corticotropin-releasing factor and interleukin-1 in rats. J Gastroenterol 2015; 50:1153-61. [PMID: 25808230 DOI: 10.1007/s00535-015-1070-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/13/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Most studies evaluating visceral sensation measure visceromotor response (VMR) to colorectal distention (CRD). However, CRD itself induces visceral sensitization, and little is known about the detailed characteristics of this response. The present study tried to clarify this question. METHODS VMR was determined by measuring abdominal muscle contractions as a response to CRD in rats. The CRD set consisted of two isobaric distentions (60 mmHg for 10 min twice, with a 30-min rest), and the CRD set was performed on two separate days, i.e., days 1 and 3, 8. RESULTS On day 1, VMR to the second CRD was increased as compared with that to the first CRD, which is the acute sensitization. VMR to the first CRD on day 3 returned to the same level as that to the first CRD on day 1, and total VMR, i.e., the whole response to the CRD set, was not different between day 1 and day 3. However, total VMR was significantly increased on day 8 as compared with that on day 1, suggesting CRD induced the delayed sensitization. Intraperitoneally administered astressin (200 µg/kg), a corticotropin-releasing factor receptor antagonist, at the end of the first CRD blocked the acute sensitization, but anakinra (20 mg/kg, intraperitoneally), an interleukin-1 receptor antagonist, did not modify it. Astressin (200 µg/kg, twice before CRD on day 8) did not alter the delayed sensitization, but anakinra (20 mg/kg, twice) abolished it. CONCLUSIONS CRD induced both acute sensitization and delayed sensitization, which were mediated through peripheral corticotropin-releasing factor and interleukin-1 pathways, respectively.
Collapse
Affiliation(s)
- Tsukasa Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, Midorigaoka Higashi 2-1-1-1, Asahikawa, 078-8510, Japan.
| | - Shima Kumei
- Department of General Medicine, Asahikawa Medical University, Midorigaoka Higashi 2-1-1-1, Asahikawa, 078-8510, Japan
| | - Saori Miyagishi
- Department of General Medicine, Asahikawa Medical University, Midorigaoka Higashi 2-1-1-1, Asahikawa, 078-8510, Japan
| | - Kaoru Takakusaki
- Research Center for Brain Function and Medical Engineering, Asahikawa Medical University, Midorigaoka Higashi 2-1-1-1, Asahikawa, 078-8510, Japan
| | - Toshikatsu Okumura
- Department of General Medicine, Asahikawa Medical University, Midorigaoka Higashi 2-1-1-1, Asahikawa, 078-8510, Japan
| |
Collapse
|
37
|
Chung SH, Lee KJ, Kim JY, Im SG, Kim E, Yang MJ, Ryu SH. Association of the Extent of Atrophic Gastritis With Specific Dyspeptic Symptoms. J Neurogastroenterol Motil 2015; 21:528-36. [PMID: 26424039 PMCID: PMC4622135 DOI: 10.5056/jnm15074] [Citation(s) in RCA: 5] [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: 05/05/2015] [Revised: 06/18/2015] [Accepted: 07/15/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND/AIMS It remains unclear whether atrophic gastritis can affect dyspeptic symptoms. We aimed to investigate whether the extent of atrophic gastritis is associated with specific dyspeptic symptoms. METHODS Consecutive adults in a routine health-checkup program were enrolled in the study. The extent of atrophic gastritis was classified into 3 groups based on the Kimura-Takemoto criteria; the gastritis with no or little atrophy (group A: C0), the gastritis with atrophy mainly in the antrum (group B: C1 and C2), and the gastritis with atrophy in the large area of the corpus (group C: C3 and O). Upper gastrointestinal symptoms were categorized into "typical reflux symptoms," "epigastric pain syndrome (EPS)-related symptoms," and "postprandial distress syndrome (PDS)-related symptoms." RESULTS A total of 1827 patients (1009 males, mean age 45.1 years) were included in the analysis. The subgroups of atrophic gastritis were as follows: group A (n = 1218, 66.7%), group B (n = 392, 21.4%), and group C (n = 217, 11.9%). Typical reflux, EPS-related, and PDS-related symptoms were present in 10.5%, 19.8%, and 16.2% of the subjects, respectively. PDS-related and EPS-related symptoms were significantly more prevalent in the group C of male patients and the group B of female patients, respectively, compared with other groups. PDS-related and EPS-related symptoms were independently associated with the group C in males (OR, 2.123; 95% CI, 1.090-4.136) and the group B in females (OR, 2.571; 95% CI, 1.319-5.025), respectively. CONCLUSIONS The extent of atrophic gastritis appears to affect the generation of specific dyspeptic symptoms in a gender-dependent manner.
Collapse
Affiliation(s)
- Sook Hee Chung
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Gyeonggido, Korea
| | - Kwang Jae Lee
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Gyeonggido, Korea
| | - Ja Yeon Kim
- Office of Biostatistics, Ajou University School of Medicine, Suwon, Gyeonggido, Korea
| | - Seon Gyo Im
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Gyeonggido, Korea
| | - Eunkyung Kim
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Gyeonggido, Korea
| | - Min Jae Yang
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Gyeonggido, Korea
| | - Seo Hee Ryu
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Gyeonggido, Korea
| |
Collapse
|
38
|
Jain P, Hassan AM, Koyani CN, Mayerhofer R, Reichmann F, Farzi A, Schuligoi R, Malle E, Holzer P. Behavioral and molecular processing of visceral pain in the brain of mice: impact of colitis and psychological stress. Front Behav Neurosci 2015. [PMID: 26217204 PMCID: PMC4498125 DOI: 10.3389/fnbeh.2015.00177] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Gastrointestinal disorders with abdominal pain are associated with central sensitization and psychopathologies that are often exacerbated by stress. Here we investigated the impact of colitis induced by dextran sulfate sodium (DSS) and repeated water avoidance stress (WAS) on spontaneous and nociception-related behavior and molecular signaling in the mouse brain. DSS increased the mechanical pain sensitivity of the abdominal skin while both WAS and DSS enhanced the mechanical and thermal pain sensitivity of the plantar skin. These manifestations of central sensitization were associated with augmented c-Fos expression in spinal cord, thalamus, hypothalamus, amygdala and prefrontal cortex. While WAS stimulated phosphorylation of mitogen-activated protein kinase (MAPK) p42/44, DSS activated another signaling pathway, both of which converged on c-Fos. The DSS- and WAS-induced hyperalgesia in the abdominal and plantar skin and c-Fos expression in the brain disappeared when the mice were subjected to WAS+DSS treatment. Intrarectal allyl isothiocyanate (AITC) evoked aversive behavior (freezing, reduction of locomotion and exploration) in association with p42/44 MAPK and c-Fos activation in spinal cord and brain. These effects were inhibited by morphine, which attests to their relationship with nociception. DSS and WAS exerted opposite effects on AITC-evoked p42/44 MAPK and c-Fos activation, which indicates that these transduction pathways subserve different aspects of visceral pain processing in the brain. In summary, behavioral perturbations caused by colitis and psychological stress are associated with distinct alterations in cerebral signaling. These findings provide novel perspectives on central sensitization and the sensory and emotional processing of visceral pain stimuli in the brain.
Collapse
Affiliation(s)
- Piyush Jain
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz Graz, Austria
| | - Ahmed M Hassan
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz Graz, Austria
| | - Chintan N Koyani
- Institute of Molecular Biology and Biochemistry, Medical University of Graz Graz, Austria
| | - Raphaela Mayerhofer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz Graz, Austria
| | - Florian Reichmann
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz Graz, Austria
| | - Aitak Farzi
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz Graz, Austria
| | - Rufina Schuligoi
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz Graz, Austria
| | - Ernst Malle
- Institute of Molecular Biology and Biochemistry, Medical University of Graz Graz, Austria
| | - Peter Holzer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz Graz, Austria
| |
Collapse
|
39
|
Greenwood-Van Meerveld B, Prusator DK, Johnson AC. Animal models of gastrointestinal and liver diseases. Animal models of visceral pain: pathophysiology, translational relevance, and challenges. Am J Physiol Gastrointest Liver Physiol 2015; 308:G885-903. [PMID: 25767262 DOI: 10.1152/ajpgi.00463.2014] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/11/2015] [Indexed: 02/08/2023]
Abstract
Visceral pain describes pain emanating from the thoracic, pelvic, or abdominal organs. In contrast to somatic pain, visceral pain is generally vague, poorly localized, and characterized by hypersensitivity to a stimulus such as organ distension. Animal models have played a pivotal role in our understanding of the mechanisms underlying the pathophysiology of visceral pain. This review focuses on animal models of visceral pain and their translational relevance. In addition, the challenges of using animal models to develop novel therapeutic approaches to treat visceral pain will be discussed.
Collapse
Affiliation(s)
- Beverley Greenwood-Van Meerveld
- Veterans Affairs Medical Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Dawn K Prusator
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Anthony C Johnson
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
40
|
Stress and opioids: role of opioids in modulating stress-related behavior and effect of stress on morphine conditioned place preference. Neurosci Biobehav Rev 2015; 51:138-50. [PMID: 25636946 DOI: 10.1016/j.neubiorev.2014.12.018] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 12/24/2014] [Accepted: 12/31/2014] [Indexed: 11/21/2022]
Abstract
Research studies have defined the important role of endogenous opioids in modulating stress-associated behavior. The release of β-endorphins in the amygdala in response to stress helps to cope with a stressor by inhibiting the over-activation of HPA axis. Administration of mu opioid agonists reduces the risk of developing post-traumatic stress disorder (PTSD) following a traumatic event by inhibiting fear-related memory consolidation. Similarly, the release of endogenous enkephalin and nociceptin in the basolateral amygdala and the nucleus accumbens tends to produce the anti-stress effects. An increase in dynorphin levels during prolonged exposure to stress may produce learned helplessness, dysphoria and depression. Stress also influences morphine-induced conditioned place preference (CPP) depending upon the intensity and duration of the stressor. Acute stress inhibits morphine CPP, while chronic stress potentiates CPP. The development of dysphoria due to increased dynorphin levels may contribute to chronic stress-induced potentiation of morphine CPP. The activation of ERK/cyclic AMP responsive element-binding (CREB) signaling in the mesocorticolimbic area, glucocorticoid receptors in the basolateral amygdala, and norepinephrine and galanin system in the nucleus accumbens may decrease the acute stress-induced inhibition of morphine CPP. The increase in dopamine levels in the nucleus accumbens and augmentation of GABAergic transmission in the median prefrontal cortex may contribute in potentiating morphine CPP. Stress exposure reinstates the extinct morphine CPP by activating the orexin receptors in the nucleus accumbens, decreasing the oxytocin levels in the lateral septum and amygdala, and altering the GABAergic transmission (activation of GABAA and inactivation of GABAB receptors). The present review describes these varied interactions between opioids and stress along with the possible mechanism.
Collapse
|
41
|
Moloney RD, O'Mahony SM, Dinan TG, Cryan JF. Stress-induced visceral pain: toward animal models of irritable-bowel syndrome and associated comorbidities. Front Psychiatry 2015; 6:15. [PMID: 25762939 PMCID: PMC4329736 DOI: 10.3389/fpsyt.2015.00015] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/28/2015] [Indexed: 12/12/2022] Open
Abstract
Visceral pain is a global term used to describe pain originating from the internal organs, which is distinct from somatic pain. It is a hallmark of functional gastrointestinal disorders such as irritable-bowel syndrome (IBS). Currently, the treatment strategies targeting visceral pain are unsatisfactory, with development of novel therapeutics hindered by a lack of detailed knowledge of the underlying mechanisms. Stress has long been implicated in the pathophysiology of visceral pain in both preclinical and clinical studies. Here, we discuss the complex etiology of visceral pain reviewing our current understanding in the context of the role of stress, gender, gut microbiota alterations, and immune functioning. Furthermore, we review the role of glutamate, GABA, and epigenetic mechanisms as possible therapeutic strategies for the treatment of visceral pain for which there is an unmet medical need. Moreover, we discuss the most widely described rodent models used to model visceral pain in the preclinical setting. The theory behind, and application of, animal models is key for both the understanding of underlying mechanisms and design of future therapeutic interventions. Taken together, it is apparent that stress-induced visceral pain and its psychiatric comorbidities, as typified by IBS, has a multifaceted etiology. Moreover, treatment strategies still lag far behind when compared to other pain modalities. The development of novel, effective, and specific therapeutics for the treatment of visceral pain has never been more pertinent.
Collapse
Affiliation(s)
- Rachel D Moloney
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork , Cork , Ireland
| | - Siobhain M O'Mahony
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork , Cork , Ireland ; Department of Anatomy and Neuroscience, University College Cork , Cork , Ireland
| | - Timothy G Dinan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork , Cork , Ireland ; Department of Psychiatry, University College Cork , Cork , Ireland
| | - John F Cryan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork , Cork , Ireland ; Department of Anatomy and Neuroscience, University College Cork , Cork , Ireland
| |
Collapse
|
42
|
Traub RJ, Cao DY, Karpowicz J, Pandya S, Ji Y, Dorsey SG, Dessem D. A clinically relevant animal model of temporomandibular disorder and irritable bowel syndrome comorbidity. THE JOURNAL OF PAIN 2014; 15:956-66. [PMID: 24981128 DOI: 10.1016/j.jpain.2014.06.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 06/18/2014] [Accepted: 06/19/2014] [Indexed: 12/28/2022]
Abstract
UNLABELLED Temporomandibular disorder and irritable bowel syndrome are comorbid functional chronic pain disorders of unknown etiology that are triggered/exacerbated by stress. Here we present baseline phenotypic characterization of a novel animal model to gain insight into the underlying mechanisms that contribute to such comorbid pain conditions. In this model, chronic visceral hypersensitivity, a defining symptom of irritable bowel syndrome, is dependent on 3 factors: estradiol, existing chronic somatic pain, and stress. In ovariectomized rats, estradiol replacement followed by craniofacial muscle injury and stress induced visceral hypersensitivity that persisted for months. Omission of any 1 factor resulted in a transient (1 week) visceral hypersensitivity from stress alone or no hypersensitivity (no inflammation or estradiol). Maintenance of visceral hypersensitivity was estradiol dependent, resolving when estradiol replacement ceased. Referred cutaneous hypersensitivity was concurrent with visceral hypersensitivity. Increased spinal Fos expression suggests induction of central sensitization. These data demonstrate the development and maintenance of visceral hypersensitivity in estradiol-replaced animals following distal somatic injury and stress that mimics some characteristics reported in patients with temporomandibular disorder and comorbid irritable bowel syndrome. This new animal model is a powerful experimental tool that can be employed to gain further mechanistic insight into overlapping pain conditions. PERSPECTIVE The majority of patients with temporomandibular disorder report symptoms consistent with irritable bowel syndrome. Stress and female prevalence are common to both conditions. In a new experimental paradigm in ovariectomized rats with estradiol replacement, masseter inflammation followed by stress induces visceral hypersensitivity that persists for months, modeling these comorbid pain conditions.
Collapse
Affiliation(s)
- Richard J Traub
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, Maryland; UMB Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland.
| | - Dong-Yuan Cao
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, Maryland
| | - Jane Karpowicz
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, Maryland
| | - Sangeeta Pandya
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, Maryland
| | - Yaping Ji
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, Maryland
| | - Susan G Dorsey
- Department of Organizational Systems and Adult Health, School of Nursing, University of Maryland, Baltimore, Maryland; UMB Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland
| | - Dean Dessem
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, Maryland; UMB Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland
| |
Collapse
|
43
|
Meleine M, Matricon J. Gender-related differences in irritable bowel syndrome: Potential mechanisms of sex hormones. World J Gastroenterol 2014; 20:6725-6743. [PMID: 24944465 PMCID: PMC4051914 DOI: 10.3748/wjg.v20.i22.6725] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 02/08/2014] [Accepted: 03/19/2014] [Indexed: 02/06/2023] Open
Abstract
According to epidemiological studies, twice as many women as men are affected by irritable bowel syndrome (IBS) in western countries, suggesting a role for sex hormones in IBS pathophysiology. Despite growing evidence about the implications of sex hormones in IBS symptom modulation, data on mechanisms by which they influence disease development are sparse. This review aims to determine the state of knowledge about the role of sex hormones in sensorimotor dysfunctions and to address the possible interplay of sex hormones with common risk factors associated with IBS. The scientific bibliography was searched using the following keywords: irritable bowel syndrome, sex, gender, ovarian hormone, estradiol, progesterone, testosterone, symptoms, pain, sensitivity, motility, permeability, stress, immune system, brain activity, spinal, supraspinal, imaging. Ovarian hormones variations along the menstrual cycle affect sensorimotor gastrointestinal function in both healthy and IBS populations. They can modulate pain processing by interacting with neuromodulator systems and the emotional system responsible for visceral pain perception. These hormones can also modulate the susceptibility to stress, which is a pivotal factor in IBS occurrence and symptom severity. For instance, estrogen-dependent hyper-responsiveness to stress can promote immune activation or impairments of gut barrier function. In conclusion, whereas it is important to keep in mind that ovarian hormones cannot be considered as a causal factor of IBS, they arguably modulate IBS onset and symptomatology. However, our understanding of the underlying mechanisms remains limited and studies assessing the link between IBS symptoms and ovarian hormone levels are needed to improve our knowledge of the disease evolution with regard to gender. Further studies assessing the role of male hormones are also needed to understand fully the role of sex hormones in IBS. Finally, investigation of brain-gut interactions is critical to decipher how stress, ovarian hormones, and female brain processing of pain can translate into gut dysfunctions.
Collapse
|
44
|
Abstract
This paper is the thirty-fifth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2012 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
45
|
Traub RJ, Ji Y. Sex differences and hormonal modulation of deep tissue pain. Front Neuroendocrinol 2013; 34:350-66. [PMID: 23872333 PMCID: PMC3830473 DOI: 10.1016/j.yfrne.2013.07.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 07/08/2013] [Accepted: 07/09/2013] [Indexed: 12/11/2022]
Abstract
Women disproportionately suffer from many deep tissue pain conditions. Experimental studies show that women have lower pain thresholds, higher pain ratings and less tolerance to a range of painful stimuli. Most clinical and epidemiological reports suggest female gonadal hormones modulate pain for some, but not all, conditions. Similarly, animal studies support greater nociceptive sensitivity in females in many deep tissue pain models. Gonadal hormones modulate responses in primary afferents, dorsal horn neurons and supraspinal sites, but the direction of modulation is variable. This review will examine sex differences in deep tissue pain in humans and animals focusing on the role of gonadal hormones (mainly estradiol) as an underlying component of the modulation of pain sensitivity.
Collapse
Affiliation(s)
- Richard J Traub
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland Baltimore, 650 W. Baltimore St., 8 South, Baltimore, MD 21201, USA; Program in Neuroscience, University of Maryland Baltimore, Baltimore, MD 21201, USA; Center for Pain Studies, University of Maryland Baltimore, Baltimore, MD 21201, USA.
| | | |
Collapse
|
46
|
He YQ, Chen Q, Ji L, Wang ZG, Bai ZH, Stephens RL, Yang M. PKCγ receptor mediates visceral nociception and hyperalgesia following exposure to PTSD-like stress in the spinal cord of rats. Mol Pain 2013; 9:35. [PMID: 23837410 PMCID: PMC3751645 DOI: 10.1186/1744-8069-9-35] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 07/04/2013] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Clinical studies indicate that patients with post-traumatic stress disorder (PTSD) frequently share comorbidity with numerous chronic pain conditions. However, the sustained effects of PTSD-like stress over time on visceral nociception and hyperalgesia have been rarely studied, and the underlying mechanisms of stress-induced modulation of visceral hyperalgesia remain elusive. The purpose of this study was to investigate the characterization of visceral nociception and hyperalgesia over time in rats exposed to PTSD-like stress, and to explore the potential role of protein kinase C gamma (PKCγ) in mediating visceral hyperalgesia following exposure to PTSD-like stress. RESULTS On day 1, the rats exposed to single-prolonged stress (SPS, an established animal model for PTSD) exhibited an analgesic response and its visceromotor response (VMR) to graded colorectal distention (CRD) at 40 and 60 mmHg was reduced compared with the control group (all P < 0.05). On day 6, the VMR returned to the baseline value. However, as early as 7 days after SPS, VMR dramatically increased compared with its baseline value and that in the controls (all P < 0.001) and this increase persisted for 28 days, with the peak on day 9. Abdominal withdrawal reflex (AWR) scores were higher in SPS rats than in controls on days 7, 9, 14, 21 and 28 (all P < 0.001). Intrathecal administration of GF109203X (an inhibitor of PKC gamma), attenuated the SPS-induced increase in both VMR and AWR scores on days 7, 14, 21 and 28 (all P < 0.05). PKCγ protein expression determined by immunofluorescence was reduced in the spinal cord within 3 days after the exposure to SPS (P < 0.01), which returned to normal levels between days 4 and 6, and significantly increased from day 7, and this increase was maintained on days 14, 21, and 28 (all P < 0.001), with the peak on day 9. In addition, Western blotting showed a consistent trend in the changes of PKCγ protein expression. CONCLUSIONS The modified SPS alters visceral sensitivity to CRD, and contributes to the maintenance of visceral hyperalgesia, which is associated with enhanced PKCγ expression in the spinal cord. Functional blockade of the PKCγ receptors attenuates SPS-induced visceral hyperalgesia. Thus, the present study identifies a specific molecular mechanism for visceral hyperalgesia which may pave the way for novel therapeutic strategies for PTSD-like conditions.
Collapse
|
47
|
Mulak A, Larauche M, Taché Y. Psychological Stress Induces Visceral Analgesic or Hyperalgesic Response in Rodents: A Role of Preconditions. ACTA ACUST UNITED AC 2012; 30:106-114. [PMID: 25400317 DOI: 10.1159/000338417] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A dual action of stress on pain modulation has been well characterized in the somatic pain studies, while much less is known in the visceral field. In the context of clinical observations that stress plays a critical role in the pathophysiology, symptoms presentation and clinical outcome of functional gastrointestinal disorders such as irritable bowel syndrome (IBS), a number of acute and chronic stress models have been developed in rodents. Recent data have demonstrated that the state of the animal tested (naïve vs. exposed to surgery), its social environment (group housing vs. single housing), the methods used to record visceromotor responses (EMG requiring surgery and antibiotic after surgery vs. manometry not requiring surgery/antibiotic) can significantly affect the analgesic response to exteroceptive stressors. Growing body of evidence indicates that a new noninvasive solid-state manometric method to monitor viscero motor response is valuable to unravel both analgesia and hyperalgesia without confounding factors. This is of critical importance regarding the recently recognized role of a compromised engagement of the inhibitory descending pain pathways in IBS patients. Better understanding of mechanisms of stress-related modulation of visceral pain leading to analgesia and hyperalgesia, along with the role of sex-dependent factors and complex interactions of the brain-gut-enteric microbiota axis may lead to new therapeutic targets in IBS.
Collapse
Affiliation(s)
- Agata Mulak
- CURE/Digestive Diseases Research Center, Oppenheimer Family Center for Neurobiology of Stress, Division of Digestive Diseases, Department of Medicine, University of California Los Angeles, and VA Greater Los Angeles Health Care System, Los Angeles, Calif., USA ; Department of Gastroenterology and Hepatology, Wroclaw Medical University, Wroclaw, Poland
| | - Muriel Larauche
- CURE/Digestive Diseases Research Center, Oppenheimer Family Center for Neurobiology of Stress, Division of Digestive Diseases, Department of Medicine, University of California Los Angeles, and VA Greater Los Angeles Health Care System, Los Angeles, Calif., USA
| | - Yvette Taché
- CURE/Digestive Diseases Research Center, Oppenheimer Family Center for Neurobiology of Stress, Division of Digestive Diseases, Department of Medicine, University of California Los Angeles, and VA Greater Los Angeles Health Care System, Los Angeles, Calif., USA
| |
Collapse
|