1
|
Calvillo-Montoya DL, Martínez-Magaña CJ, Oviedo N, Murbartián J. The Estrogen Receptor Alpha Regulates the Sex-dependent Expression and Pronociceptive Role of Bestrophin-1 in Neuropathic Rats. THE JOURNAL OF PAIN 2024; 25:104513. [PMID: 38521145 DOI: 10.1016/j.jpain.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/21/2024] [Accepted: 03/14/2024] [Indexed: 03/25/2024]
Abstract
Bestrophin-1, a calcium-activated chloride channel (CaCC), is involved in neuropathic pain; however, it is unclear whether it has a dimorphic role in female and male neuropathic rats. This study investigated if 17β-estradiol and estrogen receptor alpha (ERα) activation regulate bestrophin-1 activity and expression in neuropathic rats. Neuropathic pain was induced by L5-spinal nerve transection (SNT). Intrathecal administration of CaCCinh-A01 (.1-1 µg), a CaCC blocker, reversed tactile allodynia induced by SNT in female but not male rats. In contrast, T16Ainh-A01, a selective anoctamin-1 blocker, had an equal antiallodynic effect in both sexes. SNT increased bestrophin-1 protein expression in injured L5 dorsal root ganglia (DRG) in female rats but decreased bestrophin-1 protein in L5 DRG in male rats. Ovariectomy prevented the antiallodynic effect of CaCCinh-A01, but 17β-estradiol replacement restored it. The effect of CaCCinh-A01 was prevented by intrathecal administration of MPP, a selective ERα antagonist, in rats with and without prior hormonal manipulation. In female rats with neuropathy, ovariectomy prevented the increase in bestrophin-1 and ERα protein expression, while 17β-estradiol replacement allowed for an increase in both proteins in L5 DRG. Furthermore, ERα antagonism (with MPP) prevented the increase in bestrophin-1 and ERα protein expression. Finally, ERα activation with PPT, an ERα selective activator, induced the antiallodynic effect of CaCCinh-A01 in neuropathic male rats and prevented the reduction in bestrophin-1 protein expression in L5 DRG. In summary, data suggest ERα activation is necessary for bestrophin-1's pronociceptive action to maintain neuropathic pain in female rats. PERSPECTIVE: The mechanisms involved in neuropathic pain differ between male and female animals. Our data suggest that ERα is necessary for expression and function of bestrophin-1 in neuropathic female but not male rats. Data support the idea that a therapeutic approach to relieving neuropathic pain must be based on patient's gender.
Collapse
Affiliation(s)
| | | | - Norma Oviedo
- Unidad de Investigación Médica en Inmunología e Infectología, Centro Médico Nacional, La Raza, IMSS, Mexico City, Mexico
| | - Janet Murbartián
- Department of Pharmacobiologý, Cinvestav, South Campus, Mexico City, Mexico.
| |
Collapse
|
2
|
Mota-Carrillo E, Juárez-Contreras R, González-Ramírez R, Luis E, Morales-Lázaro SL. The Influence of Sex Steroid Hormone Fluctuations on Capsaicin-Induced Pain and TRPV1 Expression. Int J Mol Sci 2024; 25:8040. [PMID: 39125611 PMCID: PMC11312332 DOI: 10.3390/ijms25158040] [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: 06/29/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
Sexual dimorphism among mammals includes variations in the pain threshold. These differences are influenced by hormonal fluctuations in females during the estrous and menstrual cycles of rodents and humans, respectively. These physiological conditions display various phases, including proestrus and diestrus in rodents and follicular and luteal phases in humans, distinctly characterized by varying estrogen levels. In this study, we evaluated the capsaicin responses in male and female mice at different estrous cycle phases, using two murine acute pain models. Our findings indicate that the capsaicin-induced pain threshold was lower in the proestrus phase than in the other three phases in both pain assays. We also found that male mice exhibited a higher pain threshold than females in the proestrus phase, although it was similar to females in the other cycle phases. We also assessed the mRNA and protein levels of TRPV1 in the dorsal root and trigeminal ganglia of mice. Our results showed higher TRPV1 protein levels during proestrus compared to diestrus and male mice. Unexpectedly, we observed that the diestrus phase was associated with higher TRPV1 mRNA levels than those in both proestrus and male mice. These results underscore the hormonal influence on TRPV1 expression regulation and highlight the role of sex steroids in capsaicin-induced pain.
Collapse
Affiliation(s)
- Edgardo Mota-Carrillo
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (E.M.-C.); (R.J.-C.)
- Programa de Doctorado en Ciencias Biomédicas, Unidad de Posgrado, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Rebeca Juárez-Contreras
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (E.M.-C.); (R.J.-C.)
- Programa de Posgrado en Ciencias Biológicas, Unidad de Posgrado, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Ricardo González-Ramírez
- Departamento de Biología Molecular e Histocompatibilidad, Hospital General “Dr. Manuel Gea González”, Ciudad de México 14080, Mexico;
- Centro de Investigación sobre el Envejecimiento, CINVESTAV, Ciudad de México 14390, Mexico
| | - Enoch Luis
- Investigador por México—Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
- Laboratorio Nacional de Canalopatías, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Sara Luz Morales-Lázaro
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (E.M.-C.); (R.J.-C.)
- Centro de Investigación sobre el Envejecimiento, CINVESTAV, Ciudad de México 14390, Mexico
| |
Collapse
|
3
|
Rodríguez-Palma EJ, Islas-Espinoza AM, Ramos-Rodríguez II, Pizaña-Encarnación JM, Gutiérrez-Agredano MÁ, Morales-Moreno C, Fernández-Guasti A, Granados-Soto V. Estradiol modulates the role of the spinal α 6-subunit containing GABA A receptors in female rats with neuropathic pain. Eur J Pharmacol 2024; 974:176616. [PMID: 38679122 DOI: 10.1016/j.ejphar.2024.176616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
The purpose of this study was to investigate the mechanisms underlying sex differences in the role of spinal α6-subunit containing GABAA (α6GABAA) receptors in rats with neuropathic pain. Intrathecal 2,5-dihydro-7-methoxy-2-(4-methoxyphenyl)-3H-pyrazolo [4,3-c] quinoline-3-one (PZ-II-029, positive allosteric modulator of α6GABAA receptors) reduced tactile allodynia in female but not in male rats with neuropathic pain. PZ-II-029 was also more effective in females than males in inflammatory and nociplastic pain. Ovariectomy abated the antiallodynic effect of PZ-II-029 in neuropathic rats, whereas 17β-estradiol or 4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl) trisphenol (PPT), estradiol receptor-α agonist, restored the effect of PZ-II-029 in ovariectomized rats. Blockade of estradiol receptor-α, using MPP (1,3-bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy) phenol]-1H-pyrazole dihydrochloride), prevented the effect of 17β-estradiol on PZ-II-029-induced antiallodynia in ovariectomized neuropathic females. Nerve injury reduced α6GABAA receptor protein expression at the dorsal root ganglia (DRG) and spinal cord of intact and ovariectomized female rats. In this last group, reconstitution with 17β-estradiol fully restored its expression in DRG and spinal cord. In male rats, nerve injury reduced α6GABAA receptor protein expression only at the spinal cord. Nerve injury enhanced estradiol receptor-α protein expression at the DRG in intact non-ovariectomized rats. However, ovariectomy decreased estradiol receptor-α protein expression at the DRG. In the spinal cord there were no changes in estradiol receptor-α protein expression. 17β-estradiol restored estradiol receptor-α protein expression at the DRG and increased it at the spinal cord of neuropathic rats. These data suggest that 17β-estradiol modulates the expression and function of the α6GABAA receptor through its interaction with estradiol receptor-α in female rats.
Collapse
Affiliation(s)
- Erick J Rodríguez-Palma
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Ana M Islas-Espinoza
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Itzel I Ramos-Rodríguez
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Tabasco, Mexico
| | | | - Miguel Á Gutiérrez-Agredano
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | | | | | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico.
| |
Collapse
|
4
|
Hullugundi SK, Dolkas J, Chernov AV, Yaksh TL, Eddinger KA, Angert M, Catroli GF, Strongin AY, Dougherty PM, Li Y, Quehenberger O, Armando A, Shubayev VI. Cholesterol-dependent LXR transcription factor activity represses pronociceptive effects of estrogen in sensory neurons and pain induced by myelin basic protein fragments. Brain Behav Immun Health 2024; 38:100757. [PMID: 38590761 PMCID: PMC10999831 DOI: 10.1016/j.bbih.2024.100757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/17/2024] [Indexed: 04/10/2024] Open
Abstract
Background A bioactive myelin basic protein (MBP) fragment, comprising MBP84-104, is released in sciatic nerve after chronic constriction injury (CCI). Intraneural injection (IN) of MBP84-104 in an intact sciatic nerve is sufficient to induce persistent neuropathic pain-like behavior via robust transcriptional remodeling at the injection site and ipsilateral dorsal root ganglia (DRG) and spinal cord. The sex (female)-specific pronociceptive activity of MBP84-104 associates with sex-specific changes in cholesterol metabolism and activation of estrogen receptor (ESR)1 signaling. Methods In male and female normal and post-CCI rat sciatic nerves, we assessed: (i) cholesterol precursor and metabolite levels by lipidomics; (ii) MBP84-104 interactors by mass spectrometry of MBP84-104 pull-down; and (iii) liver X receptor (LXR)α protein expression by immunoblotting. To test the effect of LXRα stimulation on IN MBP84-104-induced mechanical hypersensitivity, the LXRα expression was confirmed along the segmental neuraxis, in DRG and spinal cord, followed by von Frey testing of the effect of intrathecally administered synthetic LXR agonist, GW3965. In cultured male and female rat DRGs exposed to MBP84-104 and/or estrogen treatments, transcriptional effect of LXR stimulation by GW3965 was assessed on downstream cholesterol transporter Abc, interleukin (IL)-6, and pronociceptive Cacna2d1 gene expression. Results CCI regulated LXRα ligand and receptor levels in nerves of both sexes, with cholesterol precursors, desmosterol and 7-DHC, and oxysterol elevated in females relative to males. MBP84-104 interacted with nuclear receptor coactivator (Ncoa)1, known to activate LXRα, injury-specific in nerves of both sexes. LXR stimulation suppressed ESR1-induced IL-6 and Cacna2d1 expression in cultured DRGs of both sexes and attenuated MBP84-104-induced pain in females. Conclusion The injury-released bioactive MBP fragments induce pronociceptive changes by selective inactivation of nuclear transcription factors, including LXRα. By Ncoa1 sequestration, bioactive MBP fragments render LXRα function to counteract pronociceptive activity of estrogen/ESR1 in sensory neurons. This effect of MBP fragments is prevalent in females due to high circulating estrogen levels in females relative to males. Restoring LXR activity presents a promising therapeutic strategy in management of neuropathic pain induced by bioactive MBP.
Collapse
Affiliation(s)
- Swathi K. Hullugundi
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, La Jolla, CA, USA
| | - Jennifer Dolkas
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, La Jolla, CA, USA
| | - Andrei V. Chernov
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, La Jolla, CA, USA
| | - Tony L. Yaksh
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, USA
| | - Kelly A. Eddinger
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, USA
| | - Mila Angert
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, La Jolla, CA, USA
| | - Glaucilene Ferreira Catroli
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, La Jolla, CA, USA
| | - Alex Y. Strongin
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Patrick M. Dougherty
- Department of Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yan Li
- Department of Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Aaron Armando
- Lipidomics Core, University of California, San Diego, La Jolla, CA, USA
| | - Veronica I. Shubayev
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, La Jolla, CA, USA
| |
Collapse
|
5
|
Keita-Alassane S, Otis C, Bouet E, Guillot M, Frezier M, Delsart A, Moreau M, Bédard A, Gaumond I, Pelletier JP, Martel-Pelletier J, Beaudry F, Lussier B, Lecomte R, Marchand S, Troncy E. Estrogenic impregnation alters pain expression: analysis through functional neuropeptidomics in a surgical rat model of osteoarthritis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:703-715. [PMID: 35318491 DOI: 10.1007/s00210-022-02231-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/11/2022] [Indexed: 12/20/2022]
Abstract
PURPOSE Several observational studies suggest that estrogens could bias pain perception. To evaluate the influence of estrogenic impregnation on pain expression, a prospective, randomized, controlled, blinded study was conducted in a Sprague-Dawley rat model of surgically induced osteoarthritis (OA). METHODS Female rats were ovariectomized and pre-emptive 17β-estradiol (0.025 mg, 90-day release time) or placebo pellets were installed subcutaneously during the OVX procedures. Thirty-five days after, OA was surgically induced on both 17β-estradiol (OA-E) and placebo (OA-P) groups. Mechanical hypersensitivity was assessed by static weight-bearing (SWB) and paw withdrawal threshold (PWT) tests. Mass spectrometry coupled with high-performance liquid chromatography (HPLC-MS) was performed to quantify the spinal pronociceptive neuropeptides substance P (SP), calcitonin gene-related peptide (CGRP), bradykinin (BK), somatostatin (SST), and dynorphin-A (Dyn-A). RESULTS Compared to control, ovariectomized rats presented higher SP (P = 0.009) and CGRP (P = 0.017) concentrations. OA induction increased the spinal level of SP (+ 33%, P < 0.020) and decreased the release of BK (- 20%, (P < 0.037)). The OA-E rats at functional assessment put more % body weight on the affected hind limb than OA-P rats at D7 (P = 0.027) and D56 (P = 0.033), and showed higher PWT at D56 (P = 0.009), suggesting an analgesic and anti-allodynic effect of 17β-estradiol. Interestingly, the 17β-estradiol treatment counteracted the increase of spinal concentration of Dyn-A (P < 0.016) and CGRP (P < 0.018). CONCLUSION These results clearly indicate that 17β-estradiol interfers with the development of central sensitization and confirm that gender dimorphism should be considered when looking at pain evaluation.
Collapse
Affiliation(s)
- Sokhna Keita-Alassane
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
| | - Colombe Otis
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Emilie Bouet
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
| | - Martin Guillot
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
- Charles River Laboratories Montreal ULC, Senneville, QC, Canada
| | - Marilyn Frezier
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
| | - Aliénor Delsart
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
| | - Maxim Moreau
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Agathe Bédard
- Charles River Laboratories Montreal ULC, Senneville, QC, Canada
| | - Isabelle Gaumond
- Département de Chirurgie, Département d'anesthésie, Faculté de Médecine Et Des Sciences de La Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jean-Pierre Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Johanne Martel-Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Francis Beaudry
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Bertrand Lussier
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Roger Lecomte
- Sherbrooke Molecular Imaging Center, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada
| | - Serge Marchand
- Département de Chirurgie, Département d'anesthésie, Faculté de Médecine Et Des Sciences de La Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada
| | - Eric Troncy
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada.
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada.
| |
Collapse
|
6
|
Estrogens influence female itch sensitivity via the spinal gastrin-releasing peptide receptor neurons. Proc Natl Acad Sci U S A 2021; 118:2103536118. [PMID: 34312228 PMCID: PMC8346901 DOI: 10.1073/pnas.2103536118] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Many women exhibit a dramatic increase in itch during pregnancy, but the underlying mechanism is unknown. Here, we demonstrate that the female sex steroid hormone estradiol, but not progesterone, enhances itch-related scratching behavior in female rats elicited by histamine, the prototypical itch mediator in humans. This is associated with an enhancement in histamine-evoked activity of a subset of spinal dorsal horn neurons that express a neuropeptide receptor, gastrin-releasing peptide receptor (GRPR), that was previously shown to be involved in spinal cord processing of itch. These findings may account for why itch sensation varies with estrogen levels and provide a basis for treating histamine-related itch diseases in females by targeting GRPR. There are sex differences in somatosensory sensitivity. Circulating estrogens appear to have a pronociceptive effect that explains why females are reported to be more sensitive to pain than males. Although itch symptoms develop during pregnancy in many women, the underlying mechanism of female-specific pruritus is unknown. Here, we demonstrate that estradiol, but not progesterone, enhances histamine-evoked scratching behavior indicative of itch in female rats. Estradiol increased the expression of the spinal itch mediator, gastrin-releasing peptide (GRP), and increased the histamine-evoked activity of itch-processing neurons that express the GRP receptor (GRPR) in the spinal dorsal horn. The enhancement of itch behavior by estradiol was suppressed by intrathecal administration of a GRPR blocker. In vivo electrophysiological analysis showed that estradiol increased the histamine-evoked firing frequency and prolonged the response of spinal GRP-sensitive neurons in female rats. On the other hand, estradiol did not affect the threshold of noxious thermal pain and decreased touch sensitivity, indicating that estradiol separately affects itch, pain, and touch modalities. Thus, estrogens selectively enhance histamine-evoked itch in females via the spinal GRP/GRPR system. This may explain why itch sensation varies with estrogen levels and provides a basis for treating itch in females by targeting GRPR.
Collapse
|
7
|
Delay L, Gonçalves Dos Santos G, Dias EV, Yaksh TL, Corr M. Sexual Dimorphism in the Expression of Pain Phenotype in Preclinical Models of Rheumatoid Arthritis. Rheum Dis Clin North Am 2021; 47:245-264. [PMID: 33781493 DOI: 10.1016/j.rdc.2020.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Rheumatoid arthritis is one of most frequent rheumatic diseases, affecting around 1% of the population worldwide. Pain impacting the quality of life for the patient with rheumatoid arthritis, is often the primary factor leading them to seek medical care. Although sex-related differences in humans and animal models of rheumatoid arthritis are described, the correlation between pain and sex in rheumatoid arthritis has only recently been directly examined. Here we review the literature and explore the mechanisms underlying the expression of the pain phenotype in females and males in preclinical models of rheumatoid arthritis.
Collapse
Affiliation(s)
- Lauriane Delay
- Department of Anesthesiology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA.
| | | | - Elayne Vieira Dias
- Department of Anesthesiology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Tony L Yaksh
- Department of Anesthesiology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Maripat Corr
- Division of Rheumatology, Allergy and Immunology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| |
Collapse
|
8
|
Sakamoto T, Ishio Y, Ishida Y, Mogi K, Kikusui T. Low maternal licking/grooming stimulation increases pain sensitivity in male mouse offspring. Exp Anim 2021; 70:13-21. [PMID: 32741955 PMCID: PMC7887629 DOI: 10.1538/expanim.20-0030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/03/2020] [Indexed: 12/21/2022] Open
Abstract
Deprivation of maternal care has been associated with higher pain sensitivity in offspring. In the present study, we hypothesized that the maternal licking/grooming behavior was an important factor for the development of the pain regulatory system. To test this hypothesis, we used male F2 offspring of early-weaned (EW) F1 mother mice that exhibit lower frequency of licking/grooming behavior. The formalin test revealed that F2 offspring of EW F1 dams showed significantly higher pain behavior than F2 offspring of normally-weaned (NW) F1 dams. We found that the mRNA levels of transient receptor potential vanilloid 1 (TRPV1), a nociceptor, were higher in the lumbosacral dorsal root ganglion (DRG) of F2 offspring of EW F1 dams than those of F2 offspring of NW F1 dams, suggesting that the higher pain sensitivity may be attributed to low licking/grooming, which may result in developmental changes in nociceptive neurons. In the DRG, mRNA levels of Mas-related G-protein coupled receptor B4 (MrgprB4), a marker of sensory neurons that detect gentle stroking, was also up-regulated in the F2 offspring of EW F1 dams. Considering that gentle touch alleviates pain, Mrgprb4 up-regulation may reflect a compensatory change. The present findings indicate important implications of maternal licking/grooming behavior in the development of the pain regulatory system.
Collapse
Affiliation(s)
- Takashi Sakamoto
- Department of Animal Science and Biotechnology, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa 252-5201, Japan
| | - Yukino Ishio
- Department of Animal Science and Biotechnology, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa 252-5201, Japan
| | - Yuiko Ishida
- Department of Animal Science and Biotechnology, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa 252-5201, Japan
| | - Kazutaka Mogi
- Department of Animal Science and Biotechnology, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa 252-5201, Japan
| | - Takefumi Kikusui
- Department of Animal Science and Biotechnology, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa 252-5201, Japan
| |
Collapse
|
9
|
So SY, Savidge TC. Sex-Bias in Irritable Bowel Syndrome: Linking Steroids to the Gut-Brain Axis. Front Endocrinol (Lausanne) 2021; 12:684096. [PMID: 34093447 PMCID: PMC8170482 DOI: 10.3389/fendo.2021.684096] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder that is more common in females. Despite its high global incidence, the disease mechanism is still unclear and therapeutic options remain limited. The sexual dimorphism in IBS incidence suggests that sex steroids play a role in disease onset and symptoms severity. This review considers sex steroids and their involvement in IBS symptoms and the underlying disease mechanisms. Estrogens and androgens play important regulatory roles in IBS symptomology, including visceral sensitivity, gut motility and psychological conditions, possibly through modulating the gut-brain axis. Steroids are regulators of hypothalamic-pituitary-adrenal activity and autonomic nervous system function. They also modulate gut microbiota and enteric nervous systems, impacting serotonin and mast cell signaling. Sex steroids also facilitate bidirectional cross-talk between the microbiota and host following bacterial transformation and recycling of steroids by the intestine. The sex-specific interplay between sex steroids and the host provides neuroendocrinology insight into the pathophysiology, epigenetics and treatment of IBS patients.
Collapse
Affiliation(s)
- Sik Yu So
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Microbiome Center, Department of Pathology, Texas Children’s Hospital, Houston, TX, United States
| | - Tor C. Savidge
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Microbiome Center, Department of Pathology, Texas Children’s Hospital, Houston, TX, United States
- *Correspondence: Tor C. Savidge,
| |
Collapse
|
10
|
Franco-Enzástiga Ú, García G, Murbartián J, González-Barrios R, Salinas-Abarca AB, Sánchez-Hernández B, Tavares-Ferreira D, Herrera LA, Barragán-Iglesias P, Delgado-Lezama R, Price TJ, Granados-Soto V. Sex-dependent pronociceptive role of spinal α 5 -GABA A receptor and its epigenetic regulation in neuropathic rodents. J Neurochem 2020; 156:897-916. [PMID: 32750173 DOI: 10.1111/jnc.15140] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/26/2020] [Accepted: 07/22/2020] [Indexed: 12/23/2022]
Abstract
Extrasynaptic α5 -subunit containing GABAA (α5 -GABAA ) receptors participate in chronic pain. Previously, we reported a sex difference in the action of α5 -GABAA receptors in dysfunctional pain. However, the underlying mechanisms remain unknown. The aim of this study was to examine this sexual dimorphism in neuropathic rodents and the mechanisms involved. Female and male Wistar rats or ICR mice were subjected to nerve injury followed by α5 -GABAA receptor inverse agonist intrathecal administration, L-655,708. The drug produced an antiallodynic effect in nerve-injured female rats and mice, and a lower effect in males. We hypothesized that changes in α5 -GABAA receptor, probably influenced by hormonal and epigenetic status, might underlie this sex difference. Thus, we performed qPCR and western blot. Nerve injury increased α5 -GABAA mRNA and protein in female dorsal root ganglia (DRG) and decreased them in DRG and spinal cord of males. To investigate the hormonal influence over α5 -GABAA receptor actions, we performed nerve injury to ovariectomized rats and reconstituted them with 17β-estradiol (E2). Ovariectomy abrogated L-655,708 antiallodynic effect and E2 restored it. Ovariectomy decreased α5 -GABAA receptor and estrogen receptor α protein in DRG of neuropathic female rats, while E2 enhanced them. Since DNA methylation might contribute to α5 -GABAA receptor down-regulation in males, we examined CpG island DNA methylation of α5 -GABAA receptor coding gene through pyrosequencing. Nerve injury increased methylation in male, but not female rats. Pharmacological inhibition of DNA methyltransferases increased α5 -GABAA receptor and enabled L-655,708 antinociceptive effect in male rats. These results suggest that α5 -GABAA receptor is a suitable target to treat chronic pain in females.
Collapse
Affiliation(s)
- Úrzula Franco-Enzástiga
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Guadalupe García
- Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Janet Murbartián
- Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | | | - Ana B Salinas-Abarca
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Beatriz Sánchez-Hernández
- Departamento de Genética, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Diana Tavares-Ferreira
- School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Luis A Herrera
- Cancer Biomedical Research Unit, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Paulino Barragán-Iglesias
- School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA.,Department of Physiology and Pharmacology, Center for Basic Sciences, Autonomous University of Aguascalientes, Aguascalientes, Mexico
| | - Rodolfo Delgado-Lezama
- Departamento de Fisiología, Biofísica y Neurociencias, Cinvestav, Zacatenco, Mexico City, Mexico
| | - Theodore J Price
- School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| |
Collapse
|
11
|
Meester I, Rivera-Silva GF, González-Salazar F. Immune System Sex Differences May Bridge the Gap Between Sex and Gender in Fibromyalgia. Front Neurosci 2020; 13:1414. [PMID: 32009888 PMCID: PMC6978848 DOI: 10.3389/fnins.2019.01414] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 12/16/2019] [Indexed: 12/19/2022] Open
Abstract
The fibromyalgia syndrome (FMS) is characterized by chronic widespread pain, sleep disturbances, fatigue, and cognitive alterations. A limited efficacy of targeted treatment and a high FMS prevalence (2–5% of the adult population) sums up to high morbidity. Although, altered nociception has been explained with the central sensitization hypothesis, which may occur after neuropathy, its molecular mechanism is not understood. The marked female predominance among FMS patients is often attributed to a psychosocial predisposition of the female gender, but here we will focus on sex differences in neurobiological processes, specifically those of the immune system, as various immunological biomarkers are altered in FMS. The activation of innate immune sensors is compatible with a neuropathy or virus-induced autoimmune diseases. Considering sex differences in the immune system and the clustering of FMS with autoimmune diseases, we hypothesize that the female predominance in FMS is due to a neuropathy-induced autoimmune pathophysiology. We invite the scientific community to verify the autoimmune hypothesis for FMS.
Collapse
Affiliation(s)
- Irene Meester
- Laboratory of Tissue Engineering and Regenerative Medicine, Basic Sciences Department, University of Monterrey, San Pedro Garza García, Mexico
| | - Gerardo Francisco Rivera-Silva
- Laboratory of Tissue Engineering and Regenerative Medicine, Basic Sciences Department, University of Monterrey, San Pedro Garza García, Mexico
| | - Francisco González-Salazar
- Laboratory of Tissue Engineering and Regenerative Medicine, Basic Sciences Department, University of Monterrey, San Pedro Garza García, Mexico.,Laboratory of Cellular Physiology, Northeast Center of Research, Mexican Institute of Social Security, Monterrey, Mexico
| |
Collapse
|
12
|
Zhang W, Wu H, Xu Q, Chen S, Sun L, Jiao C, Wang L, Fu F, Feng Y, Qian X, Chen X. Estrogen modulation of pain perception with a novel 17β-estradiol pretreatment regime in ovariectomized rats. Biol Sex Differ 2020; 11:2. [PMID: 31918752 PMCID: PMC6953313 DOI: 10.1186/s13293-019-0271-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 11/21/2019] [Indexed: 11/10/2022] Open
Abstract
Estrogen plays substantial roles in pain modulation; however, studies concerning sex hormones and nociception often yield confusing results. The discrepancy could be a result of lack of consensus to regard estrogen as a variable when working with animal models; thus, the influence of hormones’ fluctuations on nociception has continually been neglected. In the present study, we designed a novel hormone substitution model to aid us to evaluate the effects of estrogen’s long-term alterations on ovariectomy (OVX)-induced mechanical hyperalgesia and the expression of estrogen receptors(ERs). OVX rats were implanted with slow-release estrogen pellets at differently arranged time points and doses, such that a gradual elevation or decrease of serum estrogen levels following a relatively stable period of estrogen replacement was achieved in rats. Our results demonstrated that gradual estrogen depletion rather than elevation following the stable period of estrogen substitution in OVX rats alleviated OVX-induced mechanical hyperalgesia in a dose-independent manner, and the opposite estrogen increase or decrease paradigms differently regulate the expression of spinal ERs. Specifically, in rats rendered to continuously increased serum estrogen, the early phase estrogen-induced anti-nociception effect in OVX rats was eliminated, which was accompanied by an over-activation of ERα and a strong depression of ERβ, while in the OVX rats subject to gradual decrease of estrogen replacement, both ERα and ERβ increased modestly compared with the OVX group. Thus, the present study demonstrated that estrogen increase or decrease modulate nociception differently through change of spinal ERs.
Collapse
Affiliation(s)
- Wenxin Zhang
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Hui Wu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Qi Xu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Sheng Chen
- Zhejiang University School of Medicine, 866th Yuhangtang Road, Hangzhou, 310058, China
| | - Lihong Sun
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Cuicui Jiao
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Luyang Wang
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Feng Fu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Ying Feng
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Xiaowei Qian
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China
| | - Xinzhong Chen
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road 1, Hangzhou, 310006, China.
| |
Collapse
|
13
|
Up-regulation of ASIC3 expression by β-estradiol. Neurosci Lett 2018; 684:200-204. [DOI: 10.1016/j.neulet.2018.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/08/2018] [Accepted: 08/11/2018] [Indexed: 02/06/2023]
|
14
|
Ito A, Yoshimura M. Mechanisms of the analgesic effect of calcitonin on chronic pain by alteration of receptor or channel expression. Mol Pain 2018; 13:1744806917720316. [PMID: 28726540 PMCID: PMC5524232 DOI: 10.1177/1744806917720316] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The polypeptide hormone calcitonin is well known clinically for its ability to relieve osteoporotic back pain and neuropathic pain such as spinal canal stenosis, diabetic neuropathy, chemotherapy-induced neuropathy, and complex regional pain syndrome. Because the analgesic effects of calcitonin have a broad range, the underlying mechanisms of pain relief by calcitonin are largely unknown. However, recent studies using several types of chronic pain models combined with various methods have been gradually clarifying the mechanism. Here, we review the mechanisms of the analgesic action of calcitonin on ovariectomy-induced osteoporotic and neuropathic pain. The analgesic action of calcitonin may be mediated by restoration of serotonin receptors that control selective glutamate release from C-afferent fibers in ovariectomized rats and by normalization of sodium channel expression in damaged peripheral nerves. Serotonin receptors are reduced or eliminated by the relatively rapid reduction in estrogen during the postmenopausal period, and damaged nerves exhibit hyperexcitability due to abnormal expression of Na+ channel subtypes. In addition, in chemotherapy-induced peripheral neuropathy, inhibition of signals related to transient receptor potential ankyrin-1 and melastatin-8 is proposed to participate in the anti-allodynic action of calcitonin. Further, an unknown calcitonin-dependent signal appears to be present in peripheral nervous tissues and may be activated by nerve injury, resulting in regulation of the excitability of primary afferents by control of sodium channel transcription in dorsal root ganglion neurons. The calcitonin signal in normal conditions may be non-functional because no target is present, and ovariectomy or nerve injury may induce a target. Moreover, it has been reported that calcitonin reduces serotonin transporter but increases serotonin receptor expression in the thalamus in ovariectomized rats. These data suggest that calcitonin could alleviate lower back pain in patients with osteoporosis or neuropathic pain by the alteration in receptor or channel expression.
Collapse
Affiliation(s)
- Akitoshi Ito
- 1 Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Co. Ltd., Shizuoka, Japan
| | - Megumu Yoshimura
- 2 Graduate School of Health Sciences, Kumamoto Health Science University, Kumamoto, Japan
| |
Collapse
|
15
|
Traish AM, Vignozzi L, Simon JA, Goldstein I, Kim NN. Role of Androgens in Female Genitourinary Tissue Structure and Function: Implications in the Genitourinary Syndrome of Menopause. Sex Med Rev 2018; 6:558-571. [PMID: 29631981 DOI: 10.1016/j.sxmr.2018.03.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/13/2018] [Accepted: 03/18/2018] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Genitourinary conditions in women increase in prevalence with age. Androgens are prerequisite hormones of estrogen biosynthesis, are produced in larger amounts than estrogens in women, and decrease throughout adulthood. However, research and treatment for genitourinary complaints have traditionally focused on estrogens to the exclusion of other potential hormonal influences. AIM To summarize and evaluate the evidence that androgens are important for maintaining genitourinary health in women and that lack of androgenic activity can contribute to the development of symptoms of the genitourinary syndrome of menopause. METHODS The role of androgens in the pathophysiology, diagnosis, and treatment of genitourinary syndrome of menopause was discussed by an international and multidisciplinary panel during a consensus conference organized by the International Society for the Study of Women's Sexual Health. A subgroup further examined publications from the PubMed database, giving preference to clinical studies or to basic science studies in human tissues. MAIN OUTCOME MEASURES Expert opinion evaluating trophic and functional effects of androgens, their differences from estrogenic effects, and regulation of androgen and estrogen receptor expression in female genitourinary tissues. RESULTS Androgen receptors have been detected throughout the genitourinary system using immunohistochemical, western blot, ligand binding, and gene expression analyses. Lower circulating testosterone and estradiol concentrations and various genitourinary conditions have been associated with differential expression of androgen and estrogen receptors. Supplementation of androgen and/or estrogen in postmenopausal women (local administration) or in ovariectomized animals (systemic administration) induces tissue-specific responses that include changes in androgen and estrogen receptor expression, cell growth, mucin production, collagen turnover, increased perfusion, and neurotransmitter synthesis. CONCLUSION Androgens contribute to the maintenance of genitourinary tissue structure and function. The effects of androgens can be distinct from those of estrogens or can complement estrogenic action. Androgen-mediated processes might be involved in the full or partial resolution of genitourinary syndrome of menopause symptoms in women. Traish AM, Vignozzi L, Simon JA, et al. Role of Androgens in Female Genitourinary Tissue Structure and Function: Implications in the Genitourinary Syndrome of Menopause. Sex Med Rev 2018;6:558-571.
Collapse
Affiliation(s)
- Abdulmaged M Traish
- Department of Urology, Boston University School of Medicine, Boston, MA, USA
| | - Linda Vignozzi
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - James A Simon
- Women's Health & Research Consultants, Department of Obstetrics and Gynecology, George Washington University, Washington, DC, USA
| | | | - Noel N Kim
- Institute for Sexual Medicine, San Diego, CA, USA.
| |
Collapse
|
16
|
Hashem NM, El-Azrak KM, Nour El-Din ANM, Sallam SM, Taha TA, Salem MH. Effects of Trifolium alexandrinum phytoestrogens on oestrous behaviour, ovarian activity and reproductive performance of ewes during the non-breeding season. Anim Reprod Sci 2018; 196:1-8. [PMID: 29548568 DOI: 10.1016/j.anireprosci.2018.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 02/26/2018] [Accepted: 03/06/2018] [Indexed: 11/25/2022]
Abstract
Phytoestrogens are classified as naturally occurring endocrine disrupting chemicals that may affect reproductive performance of farm animals. To investigate the effects of Berseem clover phytoestrogens on reproductive performance of seasonal anoestrus ewes, twenty four late pregnant Rahmani ewes were fed either Berseem clover or maize silage (n = 12/treatment). Treatment started 2 months prepartum and continued until oestrous induction (week 8 postpartum), using the CIDR-eCG based protocol, and early pregnancy. Throughout the 2-8 weeks postpartum, oestrous rate and ovarian activity were not affected by treatment. After oestrous induction, ewes in both groups expressed comparable oestrous rates; however feeding Berseem clover extended (P < 0.05) interval to oestrus (57.00 compared with 42.54 h) and shortened (P < 0.05) oestrous duration (20.0 compared with 34.90 h). Feeding Berseem clover did not affect follicular activity except the number of medium follicles, which was less (P < 0.05) on day of oestrus (Day 0). Feeding maize silage increased (P < 0.05) the total number of follicles and number of small and medium follicles the day before oestrus (Day -1). On Day 0, the greater total number of follicles was due to the greater (P < 0.05) number of medium follicles that was associated with less number of small follicles. Although, the number and diameter of corpora lutea (CLs) were not affected by treatment, serum P4 concentration was greater (P < 0.05) for ewes fed maize silage than for those fed Berseem clover. Fecundity and litter size tended to be greater (about 35%; P = 0.132 and 0.085, respectively) in the maize silage fed ewes. In conclusion, feeding Berseem clover throughout seasonal anoestrus disrupted aspects of behavioural oestrus and there was less luteal P4 synthesis and fecundity of ewes.
Collapse
Affiliation(s)
- N M Hashem
- Animal and Fish Production Department, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt.
| | - K M El-Azrak
- Animal and Fish Production Department, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt
| | - A N M Nour El-Din
- Animal and Fish Production Department, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt
| | - S M Sallam
- Animal and Fish Production Department, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt
| | - T A Taha
- Animal and Fish Production Department, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt
| | - M H Salem
- Animal and Fish Production Department, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt
| |
Collapse
|
17
|
Difference of acute dissociation and 1-day culture on the electrophysiological properties of rat dorsal root ganglion neurons. J Physiol Biochem 2018; 74:207-221. [DOI: 10.1007/s13105-017-0606-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 12/29/2017] [Indexed: 10/24/2022]
|
18
|
Jana B, Palus K, Meller K, Całka J. Porcine dorsal root ganglia ovarian neurons are affected by long lasting testosterone treatment. Physiol Res 2017; 65:1019-1030. [PMID: 27959574 DOI: 10.33549/physiolres.933342] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We studied the effect of testosterone overdose on the number, distribution and chemical coding of ovarian neurons in the dorsal root ganglia (DRGs) in pigs. On day 3 of the estrous cycle, the ovaries of both the control and experimental gilts were injected with retrograde tracer Fast Blue. From day 4 of the estrous cycle to the expected day 20 of the second studied cycle, the experimental gilts were injected with testosterone, while the control gilts received oil. After the completion of the protocol the Th16-L5 DRGs were collected. Injections of testosterone increased the testosterone (~3.5 fold) and estradiol-17beta (~1.6 fold) levels in the peripheral blood, and reduced the following in the DRGs: the total number of the Fast Blue-positive perikarya, the population of perikarya in the L2-L4 ganglia, and the numbers of SP(+)/CGRP(+), SP(+)/PACAP(+), SP(+)/nNOS(+) and SP(-)/nNOS(+) perikarya. In the testosterone-injected gilts, the populations of SP(+)CGRP(-), small and large androgen receptors-expressing perikarya were increased. These results suggest that elevated androgen levels during pathological states may regulate the transmission of sensory modalities from the ovary to the spinal cord, and antidromic regulation of the ovarian functions.
Collapse
Affiliation(s)
- B Jana
- Division of Reproductive Biology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Olsztyn, Poland.
| | | | | | | |
Collapse
|
19
|
Bisphenol A inhibits compound action potentials in the frog sciatic nerve in a manner independent of estrogen receptors. Biochem Biophys Rep 2017; 10:145-151. [PMID: 28955742 PMCID: PMC5614631 DOI: 10.1016/j.bbrep.2017.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 03/18/2017] [Accepted: 03/22/2017] [Indexed: 01/18/2023] Open
Abstract
Although the endocrine disruptor bisphenol A (BPA) is reported to inhibit nerve conduction, the underlying mechanisms are unclear. Therefore, in the present study, we examined the effect of BPA on compound action potentials (CAPs) recorded from the frog sciatic nerve using the air-gap method. Treatment of the sciatic nerve with BPA (0.5 mM) for 20 min reduced the peak amplitude of the CAP by approximately 60% in a partially reversible manner. The reduction in the CAP peak amplitude was concentration-dependent, with a half-maximal inhibitory concentration (IC50) value of 0.31 mM. This effect of BPA was unaffected by an estrogen-receptor antagonist, 4-hydroxytamoxifen, which by itself reduced CAP peak amplitude, with an IC50 value of 0.26 mM (comparable to that of BPA). The natural estrogen 17β-estradiol, at the highest dissolvable concentration (0.05 mM), had an effect similar to that of BPA. The IC50 value of BPA was comparable to those of some local anesthetics in inhibiting frog CAPs. Our findings suggest that BPA inhibits nerve conduction in a manner independent of estrogen receptors. This action of BPA may underlie, at least in part, the neurotoxicity of the compound. Bisphenol A acutely inhibits compound action potentials in nerve fibers. The effect of bisphenol A is not mediated by estrogen receptors. The effect of bisphenol A is comparable to those of local anesthetics.
Collapse
Key Words
- 17β-Estradiol
- 4-OHT, 4-hydroxytamoxifen
- BPA, bisphenol A
- Bisphenol A
- CAP, compound action potential
- DMSO, dimethyl sulfoxide
- DRG, dorsal root ganglion
- ERRγ, estrogen-related receptor γ
- ERα, estrogen receptor α
- ERβ, estrogen receptor β
- Frog
- IC50, half-maximal inhibitory concentration
- LA, local anesthetic
- Local anesthetic
- Nerve conduction inhibition
- Sciatic nerve
- TTX, tetrodotoxin
- nH, Hill coefficient
Collapse
|
20
|
Affiliation(s)
- Bruce Becker
- Department of Emergency Medicine, Division of Sex and Gender in Emergency Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Alyson J. McGregor
- Department of Emergency Medicine, Division of Sex and Gender in Emergency Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
21
|
Inhibitory effect of estrogen receptor beta on P2X3 receptors during inflammation in rats. Purinergic Signal 2016; 13:105-117. [PMID: 27817132 DOI: 10.1007/s11302-016-9540-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 09/21/2016] [Indexed: 10/20/2022] Open
Abstract
Estrogen receptor beta (ERβ) has been shown to play a therapeutic role in inflammatory bowel disease (IBD). However, the mechanism underlying how ERβ exerts therapeutic effects and its relationship with P2X3 receptors (P2X3R) in rats with inflammation is not known. In our study, animal behavior tests, visceromotor reflex recording, and Western blotting were used to determine whether the therapeutic effect of ERβ in rats with inflammation was related with P2X3R. In complete Freund adjuvant (CFA)-induced chronic inflammation in rats, paw withdrawal threshold was significantly decreased which were then reversed by systemic injection of ERβ agonists, DPN or ERB-041. In 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis in rats, weight loss, higher DAI scores, increased visceromotor responses, and inflammatory responses were reversed by application of DPN or ERB-041. The higher expressions of P2X3R in dorsal root ganglia (DRG) of CFA-treated rats and those in rectocolon and DRG of TNBS-treated rats were all decreased by injection of DPN or ERB-041. DPN application also inhibited P2X3R-evoked inward currents in DRG neurons from TNBS rats. Mechanical hyperalgesia and increased P2X3 expression in ovariectomized (OVX) CFA-treated rats were reversed by estrogen replacements. Furthermore, the expressions of extracellular signal-regulated kinase (ERK) in DRG and spinal cord dorsal horn (SCDH) and c-fos in SCDH were significantly decreased after estrogen replacement compared with those of OVX rats. The ERK antagonist U0126 significantly reversed mechanical hyperalgesia in the OVX rats. These results suggest that estrogen may play an important therapeutic role in inflammation through down-regulation of P2X3R in peripheral tissues and the nervous system, probably via ERβ, suggesting a novel therapeutic strategy for clinical treatment of inflammation.
Collapse
|
22
|
Mowa CN, Papka RE. The Role of Sensory Neurons in Cervical Ripening: Effects of Estrogen and Neuropeptides. J Histochem Cytochem 2016; 52:1249-58. [PMID: 15385571 DOI: 10.1177/002215540405201001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Central nervous system nuclei and circuits, such as the medial preoptic, ventromedial and paraventricular nuclei of the hypothalamus, play important roles in reproduction and parturition, and are influenced by estrogen. Peripheral autonomic and sensory neurons also play important roles in pregnancy and parturition. Moreover, the steroid hormone estrogen acts directly, not only on the reproductive tract organs (uterus and cervix), but also on the central and peripheral nerves by regulating expression of various neuronal genes. The peripheral primary afferent neurons innervating the uterine cervix relay mechanical and biochemical sensory information induced by local cervical events and by passage of fetuses, to the spinal cord and supraspinal centers. Consequently, the birth process in mammals is influenced by the combined action of neurons and hormones. Peripheral sensory stimuli, induced physiologically by fetal expulsion or mechanically by vaginocervical stimulation, alter behavior, as well as autonomic and neuroendocrine systems. Recent evidence indicates that primary afferent neurons innervating the cervix, in addition to their sensory effects, likely exert local “efferent” actions on the ripening cervix near term. These efferent effects may involve estrogen-regulated production of such neuropeptides as substance P and calcitonin gene-related peptide in lumbosacral dorsal root ganglia, and their release in the cervix. Collectively, these findings suggest an interrelationship among estrogen, cervix-related sensory neurons, and local cervical events near term.
Collapse
Affiliation(s)
- C N Mowa
- Northeastern Ohio Universities College of Medicine, Department of Neurobiology, 4209 State Rt. 44, P.O. Box 95, Rootstown, OH 44272, USA
| | | |
Collapse
|
23
|
Pardutz A, Hoyk Z, Varga H, Vecsei L, Schoenen J. Oestrogen-Modulated Increase of Calmodulin-Dependent Protein Kinase II (CamKII) in Rat Spinal Trigeminal Nucleus After Systemic Nitroglycerin. Cephalalgia 2016; 27:46-53. [PMID: 17212683 DOI: 10.1111/j.1468-2982.2006.01244.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Migraine can be triggered by systemic administration of the nitric oxide (NO) donor nitroglycerin (NTG) and by abrupt falls in plasma oestradiol. Calmodulin-dependent protein kinase II (CamKII) present in superficial dorsal horns is thought to play a role in sensitization of central nociceptors, a phenomen present in migraineurs. We therefore examined in rats the expression of CamKII in the caudal trigeminal nucleus (TNC) after subcutaneous NTG (10 mg/kg) and its modulation by oestrogen. In male rats and in ovariectomized females, after 4 h NTG increased significantly CamKII expression in the superficial layers of TNC, but not in the upper thoracic spinal cord. NTG had no effect on CamKII expression in oestradiol-treated ovariectomized animals. Thus NTG, i.e. NO, selectively enhances CamKII in the rat TNC and oestradiol blocks this effect. These data may help to understand the mechanisms by which NO triggers migraine attacks and oestrogens influence migraine severity.
Collapse
Affiliation(s)
- A Pardutz
- Research Centre for Cellular & Molecular Neurobiology, University of Liège, Liège, Belgium
| | | | | | | | | |
Collapse
|
24
|
Robarge JD, Duarte DB, Shariati B, Wang R, Flockhart DA, Vasko MR. Aromatase inhibitors augment nociceptive behaviors in rats and enhance the excitability of sensory neurons. Exp Neurol 2016; 281:53-65. [PMID: 27072527 DOI: 10.1016/j.expneurol.2016.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/28/2016] [Accepted: 04/05/2016] [Indexed: 12/22/2022]
Abstract
Although aromatase inhibitors (AIs) are commonly used therapies for breast cancer, their use is limited because they produce arthralgia in a large number of patients. To determine whether AIs produce hypersensitivity in animal models of pain, we examined the effects of the AI, letrozole, on mechanical, thermal, and chemical sensitivity in rats. In ovariectomized (OVX) rats, administering a single dose of 1 or 5mg/kg letrozole significantly reduced mechanical paw withdrawal thresholds, without altering thermal sensitivity. Repeated injection of 5mg/kg letrozole in male rats produced mechanical, but not thermal, hypersensitivity that extinguished when drug dosing was stopped. A single dose of 5mg/kg letrozole or daily dosing of letrozole or exemestane in male rats also augmented flinching behavior induced by intraplantar injection of 1000nmol of adenosine 5'-triphosphate (ATP). To determine whether sensitization of sensory neurons contributed to AI-induced hypersensitivity, we evaluated the excitability of neurons isolated from dorsal root ganglia of male rats chronically treated with letrozole. Both small and medium-diameter sensory neurons isolated from letrozole-treated rats were more excitable, as reflected by increased action potential firing in response to a ramp of depolarizing current, a lower resting membrane potential, and a lower rheobase. However, systemic letrozole treatment did not augment the stimulus-evoked release of the neuropeptide calcitonin gene-related peptide (CGRP) from spinal cord slices, suggesting that the enhanced nociceptive responses were not secondary to an increase in peptide release from sensory endings in the spinal cord. These results provide the first evidence that AIs modulate the excitability of sensory neurons, which may be a primary mechanism for the effect of these drugs to augment pain behaviors in rats.
Collapse
Affiliation(s)
- Jason D Robarge
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, United States.
| | - Djane B Duarte
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States; Laboratório de Farmacologia Molecular, Faculdade de Ciências da Saúde, Universidade de Brasília, Brazil.
| | - Behzad Shariati
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States.
| | - Ruizhong Wang
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States.
| | - David A Flockhart
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, United States.
| | - Michael R Vasko
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
25
|
Liang Y, Yao S. Potential role of estrogen in maintaining the imbalanced sympathetic and sensory innervation in endometriosis. Mol Cell Endocrinol 2016; 424:42-9. [PMID: 26777300 DOI: 10.1016/j.mce.2016.01.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 11/02/2015] [Accepted: 01/13/2016] [Indexed: 02/07/2023]
Abstract
Endometriosis, one of the most common benign gynecological diseases, affects millions of women of childbearing age. Endometriosis-associated pain is a major cause of disability and compromised quality of life in women. Neuropathic mechanisms are believed to play an important role. An imbalanced sympathetic and sensory innervation (reduced sympathetic innervation, with unchanged or increased sensory innervation in endometriotic lesions) has been demonstrated in endometriosis in recent studies. And it is believed to contribute to the pathogenesis of endometriosis-associated pain. It is primarily considered to be a natural adaptive program to endometriosis-associated inflammation. However, it is important to further clarify whether other potential modulating factors are involved in this dysregulation. It is generally accepted that endometriosis is an estrogen dependent disease. Higher estrogen biosynthesis and lower estrogen inactivation in endometriosis can lead to an excess of local estrogen in endometriotic lesions. In addition to its proliferative and anti-inflammatory actions, local estrogen in endometriosis also exerts potential neuromodulatory effects on the innervation in endometriosis. The aim of this review is to highlight the role of estrogen in mediating this imbalanced sympathetic and sensory innervation in endometriosis, through direct and indirect mechanisms on sympathetic and sensory nerves. Theoretical elaboration of the underlying mechanisms provides new insights in supporting the therapeutic role of estrogen in endometriosis-associated pain.
Collapse
Affiliation(s)
- Yanchun Liang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuzhong Yao
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
26
|
Vacca V, Marinelli S, Pieroni L, Urbani A, Luvisetto S, Pavone F. 17beta-estradiol counteracts neuropathic pain: a behavioural, immunohistochemical, and proteomic investigation on sex-related differences in mice. Sci Rep 2016; 6:18980. [PMID: 26742647 PMCID: PMC4705539 DOI: 10.1038/srep18980] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 09/02/2015] [Indexed: 01/31/2023] Open
Abstract
Sex differences play a role in pain sensitivity, efficacy of analgesic drugs and prevalence of neuropathic pain, even if the underlying mechanisms are far from being understood. We demonstrate that male and female mice react differently to structural and functional changes induced by sciatic nerve ligature, used as model of neuropathic pain. Male mice show a gradual decrease of allodynia and a complete recovery while, in females, allodynia and gliosis are still present four months after neuropathy induction. Administration of 17β-estradiol is able to significantly attenuate this difference, reducing allodynia and inducing a complete recovery also in female mice. Parallel to pain attenuation, 17β-estradiol treated-mice show a functional improvement of the injured limb, a faster regenerative process of the peripheral nerve and a decreased neuropathy-induced gliosis. These results indicate beneficial effects of 17β-estradiol on neuropathic pain and neuronal regeneration and focuses on the importance of considering gonadal hormones also in clinical studies.
Collapse
Affiliation(s)
- Valentina Vacca
- CNR-National Research Council, Institute of Cell Biology and Neurobiology, 00143 Roma, Italy.,IRCCS Fondazione Santa Lucia, 00143 Roma, Italy
| | - Sara Marinelli
- CNR-National Research Council, Institute of Cell Biology and Neurobiology, 00143 Roma, Italy.,IRCCS Fondazione Santa Lucia, 00143 Roma, Italy
| | - Luisa Pieroni
- IRCCS Fondazione Santa Lucia, 00143 Roma, Italy.,Department of Experimental Medicine and Surgery, Division of Biochemistry, University of "Tor Vergata", 00133 Roma, Italy
| | - Andrea Urbani
- IRCCS Fondazione Santa Lucia, 00143 Roma, Italy.,Department of Experimental Medicine and Surgery, Division of Biochemistry, University of "Tor Vergata", 00133 Roma, Italy
| | - Siro Luvisetto
- CNR-National Research Council, Institute of Cell Biology and Neurobiology, 00143 Roma, Italy.,IRCCS Fondazione Santa Lucia, 00143 Roma, Italy
| | - Flaminia Pavone
- CNR-National Research Council, Institute of Cell Biology and Neurobiology, 00143 Roma, Italy.,IRCCS Fondazione Santa Lucia, 00143 Roma, Italy
| |
Collapse
|
27
|
Ovarian hormones and chronic pain: A comprehensive review. Pain 2014; 155:2448-2460. [DOI: 10.1016/j.pain.2014.08.027] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 08/15/2014] [Accepted: 08/20/2014] [Indexed: 01/19/2023]
|
28
|
Effects of estradiol on voltage-gated potassium channels in mouse dorsal root ganglion neurons. J Membr Biol 2014; 247:541-8. [PMID: 24838692 DOI: 10.1007/s00232-014-9670-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 04/29/2014] [Indexed: 01/22/2023]
Abstract
Voltage-gated potassium channels are regulators of membrane potentials, action potential shape, firing adaptation, and neuronal excitability in excitable tissues including in the primary sensory neurons of dorsal root ganglion (DRG). In this study, using the whole-cell patch-clamp technique, the effect of estradiol (E2) on voltage-gated total outward potassium currents, the component currents transient "A-type" current (I A) currents, and "delayed rectifier type" (I KDR) currents in isolated mouse DRG neurons was examined. We found that the extracellularly applied 17β-E2 inhibited voltage-gated total outward potassium currents; the effects were rapid, reversible, and concentration-dependent. Moreover, the membrane impermeable E2-BSA was as efficacious as 17β-E2, whereas 17α-E2 had no effect. 17β-E2-stimulated decrease in the potassium current was unaffected by treatment with ICI 182780 (classic estrogen receptor antagonist), actinomycin D (RNA synthesis inhibitor), or cycloheximide (protein synthesis inhibitor). We also found that I A and I KDR were decreased after 17β-E2 application. 17β-E2 significantly shifted the activation curve for I A and I KDR channels in the hyperpolarizing direction. In conclusion, our results demonstrate that E2 inhibited voltage-gated K(+) channels in mouse DRG neurons through a membrane ER-activated non-genomic pathway.
Collapse
|
29
|
Pan XQ, Malykhina AP. Estrous cycle dependent fluctuations of regulatory neuropeptides in the lower urinary tract of female rats upon colon-bladder cross-sensitization. PLoS One 2014; 9:e94872. [PMID: 24788240 PMCID: PMC4006778 DOI: 10.1371/journal.pone.0094872] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 03/20/2014] [Indexed: 12/30/2022] Open
Abstract
Co-morbidity of bladder, bowel, and non-specific pelvic pain symptoms is highly prevalent in women. Little evidence is present on modulation of pelvic pain syndromes by sex hormones, therefore, the objective of this study was to clarify the effects of hormonal fluctuations within the estrous cycle on regulatory neuropeptides in female rats using a model of neurogenic bladder dysfunction. The estrous cycle in female rats (Sprague-Dawley, 230-250 g) was assessed by vaginal smears and weight of uterine horns. Neurogenic bladder dysfunction was induced by a single inflammatory insult to the distal colon. Protein expression of calcitonin gene related peptide (CGRP), substance P (SP), nerve growth factor (NGF), and brain derived neurotrophic factor (BDNF) in the pelvic organs, sensory ganglia and lumbosacral spinal cord was compared in rats in proestrus (high estrogen) vs diestrus (low estrogen). Under normal physiological conditions, concentration of SP and CGRP was similar in the distal colon and urinary bladder during all phases of the estrous cycle, however, acute colitis induced a significant up-regulation of CGRP content in the colon (by 63%) and urinary bladder (by 54%, p≤0.05 to control) of rats in proestrus. These changes were accompanied by a significant diminution of CGRP content in L6-S2 DRG after colonic treatment, likely associated with its release in the periphery. In rats with high estrogen at the time of testing (proestrus), experimental colitis caused a significant up-regulation of BDNF colonic content from 26.1±8.5 pg/ml to 83.4±32.5 pg/ml (N = 7, p≤0.05 to control) and also induced similar effects on BDNF in the urinary bladder which was also up-regulated by 5-fold in rats in proestrus (p≤0.05 to respective control). Our results demonstrate estrous cycle dependent fluctuations of regulatory neuropeptides in the lower urinary tract upon colon-bladder cross-sensitization, which may contribute to pain fluctuations in female patients with neurogenic bladder pain.
Collapse
Affiliation(s)
- Xiao-Qing Pan
- Division of Urology, Department of Surgery, University of Pennsylvania, Glenolden, Pennsylvania, United States of America
| | - Anna P. Malykhina
- Division of Urology, Department of Surgery, University of Pennsylvania, Glenolden, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
30
|
Blockade of substance P receptor attenuates osteoporotic pain, but not bone loss, in ovariectomized mice. Menopause 2014; 20:1074-83. [PMID: 23549442 DOI: 10.1097/gme.0b013e31828837a6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the effect of a substance P (SP) receptor (NK1 receptor [NK1-R]) antagonist on hyperalgesia and bone metabolism in ovariectomized mice. METHODS Thirty-six 9-week-old mice were subjected to either bilateral ovariectomy or sham surgery. Three weeks after the operation, the mice were treated with either a single-dose injection or 2-week repeated daily administration of L-703606, an NK1-R antagonist. Behavioral tests were performed for pain assessment; tibiae and the third lumbar vertebrae were dissected and assessed for microarchitectural or biomechanical properties. The expressions of SP and NK1-R in the dorsal root ganglia and spinal cord were also evaluated. RESULTS Both single-dose injection and 2-week repeated injections of L-703606 led to a significant increase in nociceptive threshold in ovariectomized mice. However, the antihyperalgesic effect faded at 2 hours and almost disappeared at 5 hours after a single-dose injection. With the 14-day repeated treatment of ovariectomized mice, the effect was not detectable at 24 hours after the first injection but was obvious at 24 hours after 1-week and 2-week administrations and still existed at 48 hours after the last injection. Ovariectomized mice at the hyperalgesic state had enhanced SP immunoreactivity in the dorsal root ganglia and up-regulated SP and NK1-R expressions in the spinal cord. However, no significant change in serum SP level was detected. Two-week treatment with L-703606 could down-regulate these expressions but failed to salvage the deteriorated trabecular microstructure and reduced compressive strength in ovariectomized mice. CONCLUSIONS Estrogen deficiency-induced hyperalgesia is achieved through up-regulation of SP and NK1-R expressions. Blockade of SP receptor can alleviate pain but cannot ameliorate bone loss. NK1-R antagonist is not recommended for the treatment of estrogen deficiency osteoporosis.
Collapse
|
31
|
Aloisi AM, Sorda G. Relationship of female sex hormones with pain perception: focus on estrogens. Pain Manag 2014; 1:229-38. [PMID: 24646389 DOI: 10.2217/pmt.11.13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY The role of gonadal hormones has slowly gathered the right attention in the study of chronic pain mechanisms. The clear presence of sex differences in chronic pain and the number of studies showing the power of gonadal hormones to modify pain-induced behavioral responses appear to have convinced clinicians and researchers. Indeed, available data strongly indicate that more studies on gonadal hormones would certainly enhance the possibility of greatly increasing the knowledge of pain mechanisms and, thus, treatments. In the present article, old and new literature are summarized to evaluate data on pain and its modulation by gonadal hormones, particularly estrogens. Peripheral and central targets of these hormones are discussed with the aim of renewing interest in important aspects of estrogenic functions and their interactions with pain processes.
Collapse
Affiliation(s)
- Anna Maria Aloisi
- San Carlo Clinic, via dell'Ospedale 21, Paderno Dugnano Milano, Italy; Neurophysiology of Stress & Pain Laboratory, Department of Physiology, Polo Scientifico San Miniato, Via Aldo Moro 2, 53100 Siena, Italy
| | | |
Collapse
|
32
|
Jana B, Całka J, Bulc M, Czarzasta J. Long-term testosterone administration affects the number of paracervical ganglion ovary-projecting neurons in sexually mature gilts. Neurosci Res 2014; 83:89-96. [PMID: 24572298 DOI: 10.1016/j.neures.2014.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 02/10/2014] [Accepted: 02/13/2014] [Indexed: 12/15/2022]
Abstract
The influence of testosterone (T) overdose on the number and distribution of ovarian neurons in the paracervical ganglion (PCG) in pigs was examined. To identify the ovarian neurons, on day 3 of the estrous cycle, the ovaries of both the control and experimental gilts were injected with retrograde neuronal tracer Fast Blue. From next day to the expected day 20 of the second studied cycle, experimental gilts were injected with T, while control gilts received oil. The PCG was then collected and processed for double-labeling immunofluorescence. T injections increased the T (∼3.5-fold) and estradiol-17β (∼1.6-fold) levels in the peripheral blood, and reduced the following in the PCG: the total number of Fast Blue-positive neurons, the number of perikarya in the lateral part of the PCG, the numbers of VAChT(+)/SOM(+), VAChT(+)/VIP(+), VAChT(+)/nNOS(+), VAChT(+)/VIP(-), VAChT(+)/DβH(-), VAChT(-)/SOM(-), VAChT(-)/VIP(-), VAChT(-)/nNOS(-) and VAChT(-)/DβH(-) perikarya, In the T-affected PCG, the populations of ovarian perikarya coded VAChT(-)/SOM(+), VAChT(-)/VIP(+) and VAChT(-)/DβH(+), and expressing androgen receptor were increased. After T treatment within the PCG dropped the density of nerve fibers expressing VAChT and/or SOM, VIP, DβH. Obtained data suggest that elevated androgen levels occurring during pathological processes may regulate ovary function(s) by affecting the PCG gonad-supplying neurons.
Collapse
Affiliation(s)
- B Jana
- Division of Reproductive Biology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland.
| | - J Całka
- Division of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, 10-718 Olsztyn, Poland.
| | - M Bulc
- Division of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, 10-718 Olsztyn, Poland.
| | - J Czarzasta
- Division of Reproductive Biology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland.
| |
Collapse
|
33
|
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: 5.5] [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
|
34
|
Wibowo E, Wassersug RJ. The effect of estrogen on the sexual interest of castrated males: Implications to prostate cancer patients on androgen-deprivation therapy. Crit Rev Oncol Hematol 2013; 87:224-38. [DOI: 10.1016/j.critrevonc.2013.01.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 12/09/2012] [Accepted: 01/16/2013] [Indexed: 11/30/2022] Open
|
35
|
Jana B, Rytel L, Czarzasta J, Całka J. Reduction of the number of neurones in the caudal mesenteric ganglion innervating the ovary in sexually mature gilts following testosterone administration. J Neuroendocrinol 2013; 25:826-38. [PMID: 23763306 DOI: 10.1111/jne.12057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 06/05/2013] [Accepted: 06/09/2013] [Indexed: 12/22/2022]
Abstract
The effect of testosterone on the morphological and chemical plasticity of the porcine caudal mesenteric ganglion (CaMG) ovary-projecting neurones was investigated. To identify the neurones on day 3 of the oestrous cycle, the ovaries of both the control and experimental gilts were injected with Fast Blue retrograde neuronal tracer. From next day until day 20 of the anticipated second studied cycle, experimental gilts were injected with testosterone, whereas control gilts received oil. Testosterone injections increased testosterone (by approximately 3.5-fold) and 17β-oestradiol (by approximately 1.6-fold) levels in the peripheral blood and decreased the following in the CaMG: the total number of Fast Blue-positive perikarya (including small ones); the population of small perikarya in the caudal, ventral and dorsal ganglional regions; the numbers of dopamine-β-hydroxylase (DβH) and/or neuropeptide Y (NPY), somatostatin (SOM), galanin (GAL) small and large perikarya; the numbers of small perikarya containing DβH (but not NPY, SOM, GAL); and the density of DβH and/or NPY, SOM nerve fibres. A disappearance of small and large non-noradrenergic perikarya and an increase in the total number of androgen receptor-immunoreactive perikarya was noted. Our results suggest that elevated androgen levels occurring during pathological states may regulate ovary function(s) by affecting the CaMG gonad-supplying neurones.
Collapse
Affiliation(s)
- B Jana
- Division of Reproductive Biology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Olsztyn, Poland.
| | | | | | | |
Collapse
|
36
|
Lu Y, Jiang Q, Yu L, Lu ZY, Meng SP, Su D, Burnstock G, Ma B. 17β-estradiol rapidly attenuates P2X3 receptor-mediated peripheral pain signal transduction via ERα and GPR30. Endocrinology 2013; 154:2421-33. [PMID: 23610132 DOI: 10.1210/en.2012-2119] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Estrogen has been reported to affect pain perception, although the underlying mechanisms remain unclear. In this investigation, pain behavior testing, patch clamp recording, and immunohistochemistry were used on rats and transgenic mice to determine which estrogen receptors (ERs) and the related signaling pathway are involved in the rapid modulation of estrogen on P2X3 receptor-mediated events. The results showed that 17β-estradiol (E2) rapidly inhibited pain induced by α,β-methylene ATP (α,β-me-ATP), a P2X1 and P2X3 receptor agonist in ovariectomized rats and normal rats in diestrus. The ERα agonist 4,49,499-(4-propyl-[1H]-pyrazole-1,3,5-triyl) trisphenol (PPT) and G protein-coupled receptor 30 (GPR30) agonist G-1 mimicked the estrogen effect, whereas the ERβ agonist diarylpropionitrile (DPN) had no effect. In cultured rat dorsal root ganglion (DRG) neurons, PPT and G-1 but not DPN significantly attenuated α,β-me-ATP-mediated currents, with the dose-response curve of these currents shifted to the right. The inhibitory effect of E2 on P2X3 currents was blocked by G-15, a selective antagonist to the GPR30 estrogen receptor. E2 lacked this effect in DRG neurons from ERα-knockout mice but partly remained in those from ERβ-knockout mice. The P2X3 and GPR30 receptors were coexpressed in the rat DRG neurons. Furthermore, the ERK1/2 inhibitor U0126 reversed the inhibitory effect of E2 on α,β-me-ATP-induced pain and of PPT or G-1 on P2X3 receptor-mediated currents. The cAMP-protein kinase A (PKA) agonist forskolin, but not the PKC agonist phorbol-12-myristate-13-acetate (PMA), mimicked the estrogen-inhibitory effect on P2X3 receptor currents, which was blocked by another ERK1/2 inhibitor, PD98059. These results suggest that estrogen regulates P2X3-mediated peripheral pain by acting on ERα and GPR30 receptors expressed in primary afferent neurons, which probably involves the intracellular cAMP-PKA-ERK1/2 pathway.
Collapse
Affiliation(s)
- Yi Lu
- Department of Physiology, School of Pharmacy, Second Military Medical University, Shanghai 200433, People’s Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Long-term estradiol-17β administration changes the population of paracervical ganglion neurons supplying the ovary in adult gilts. J Mol Neurosci 2013; 50:424-33. [PMID: 23329259 DOI: 10.1007/s12031-012-9950-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Accepted: 12/28/2012] [Indexed: 12/22/2022]
Abstract
The aim of this study was to determine the influence of estradiol-17β (E(2)) overdose on the number and distribution of ovarian parasympathetic neurons in the paracervical ganglion (PCG) in adult pigs. To identify the neurons innervating gonads on day 3 of the estrous cycle, the ovaries of both the control and experimental gilts were injected with retrograde neuronal tracer Fast Blue. From next day to the expected day 20 of the second studied cycle, experimental gilts were injected with E(2), while control gilts received oil. The PCG were then collected and processed for double-labeling immunofluorescence. Injections of E(2) increased the E(2) level in the peripheral blood approximately four- to fivefold and reduced the following in the PCG: the total number of Fast Blue-positive neurons; the number of perikarya in the lateral part of the PCG; the numbers of vesicular acetylcholine transporter (VAChT)(+)/somatostatin(+), VAChT(+)/vasoactive intestinal polypeptide (VIP)(+), VAChT(+)/neuronal isoform of nitric oxide synthase(+), VAChT(+)/VIP(-), VAChT(+)/dopamine β-hydroxylase (DβH)(-), VAChT(-)/VIP(-), and VAChT(-)/DβH(-) perikarya; and the total number of perikarya expressing estrogen receptors (ERs) subtype α and/or β. In summary, long-term E(2) treatment of adult gilts downregulates the population of both cholinergic and ERs expressing the PCG ovary-projecting neurons. Our results suggest that elevated E(2) levels occurring during pathological states may regulate gonadal function(s) by affecting ovary-supplying neurons.
Collapse
|
38
|
Gintzler AR, Liu NJ. Importance of sex to pain and its amelioration; relevance of spinal estrogens and its membrane receptors. Front Neuroendocrinol 2012; 33:412-24. [PMID: 23036438 PMCID: PMC3778676 DOI: 10.1016/j.yfrne.2012.09.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 09/13/2012] [Accepted: 09/18/2012] [Indexed: 12/14/2022]
Abstract
Estrogens have a multitude of effects on opioid systems and are thought to play a key role in sexually dimorphic nociception and opioid antinociception. Heretofore, classical genomic actions of estrogens are largely thought to be responsible for the effects of these steroids on nociception and opioid antinociception. The recent discovery that estrogens can also activate estrogen receptors that are located in the plasma membrane, the effects of which are manifest in seconds to minutes instead of hours to days has revolutionized our thinking concerning the ways in which estrogens are likely to modulate pain responsiveness and the dynamic nature of that modulation. This review summarizes parameters of opioid functionality and nociception that are subject to modulation by estrogens, underscoring the added dimensions of such modulation that accrues from rapid membrane estrogen receptor signaling. Implications of this mode of signaling regarding putative sources of estrogens and its degradation are also discussed.
Collapse
Affiliation(s)
- Alan R Gintzler
- State University of New York, Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| | | |
Collapse
|
39
|
Estrogen and Visceral Nociception at the Level of Primary Sensory Neurons. PAIN RESEARCH AND TREATMENT 2012; 2012. [PMID: 21984961 PMCID: PMC3186056 DOI: 10.1155/2012/960780] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Clinical studies suggest the comorbidity of functional pain syndromes such as irritable bowel syndrome, painful bladder syndrome, chronic pelvic pain, and somatoform disorders approaches 40% to 60%. The incidence of episodic or persistent visceral pain associated with these “functional” disorders is two to three times higher in women than in men. One of the possible explanations for this phenomenon is estrogen modulation of viscerovisceral cross-sensitization. While a central site of this modulation has been shown previously, our studies suggest a peripheral site, the dorsal root ganglion (DRG). Estrogens have remarkably wide range of functions including modulation of voltage-gated calcium channels (VGCCs) and purinoreceptors (P2Xs). Significantly, inflammation dramatically alters purinoception by causing a several fold increase in ATP-activated current, alters the voltage dependence of P2X receptors, and enhances the expression of P2X receptors increasing neuronal hypersensitivity. Gonadal hormones are thought as indispensable cornerstones of the normal development and function, but it appears that no body region, no neuronal circuit, and virtually no cell is unaffected by them. Thus, increasing awareness toward estrogens appears to be obligatory.
Collapse
|
40
|
Jana B, Lata M, Bulc M, Całka J. Long term estradiol-17β administration changes population of the dorsal root ganglia neurons innervating the ovary in the sexually mature gilts. Neuropeptides 2012; 46:157-65. [PMID: 22677207 DOI: 10.1016/j.npep.2012.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 03/30/2012] [Accepted: 05/15/2012] [Indexed: 01/09/2023]
Abstract
The influence of estradiol-17β (E₂) overdose on the number and distribution of neurons in the dorsal root ganglia (DRGs) supplying the ovary of adult pigs was investigated. The numbers of ovarian substance P (SP)-, calcitonin gene-related peptide (CGRP)-, galanin (GAL)-, pituitary adenylate cyclase-activating polypeptide (PACAP)-, neuronal isoform of nitric oxide synthase (nNOS)- and estrogen receptors (ERs)-immunoreactive perikarya were also determined. On day 3 of the estrous cycle, the ovaries of both the control and experimental gilts were injected with retrograde tracer Fast Blue. From day 4 of the estrous cycle to the expected day 20 of the second studied cycle, the experimental gilts were injected with E₂, while the control gilts received oil. The DRGs Th16-L5 were then collected and processed for double-labelling immunofluorescence. Injections of E₂ increased the E₂ level in the peripheral blood ∼4-5-fold and reduced the following in the DRGs: the total number of Fast Blue-positive perikarya, the number of large perikarya, the population of perikarya in the L2 and L3 ganglia, the numbers of SP- and/or CGRP-, PACAP-, nNOS-immunoreactive perikarya and the number of large perikarya expressing ERs subtype α and β. These results show that long-term E₂ treatment of adult gilts affects both the spatial and neurochemical organization pattern of ovary sensory innervation. Our findings suggest that elevated E₂ levels occurring during pathological states may regulate the transmission of sensory modalities from the ovary to the spinal cord.
Collapse
Affiliation(s)
- Barbara Jana
- Division of Reproductive Biology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, 10-747 Olsztyn, Tuwima 10, Poland.
| | | | | | | |
Collapse
|
41
|
Ding H, Wang Q, Liu J, Qian W, Wang W, Wang J, Gao R, Xiao H. Alterations of gene expression of sodium channels in dorsal root ganglion neurons of estrogen receptor knockout (ERKO) mice induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Endocrine 2012; 42:118-24. [PMID: 22371119 DOI: 10.1007/s12020-012-9637-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 02/14/2012] [Indexed: 12/28/2022]
Abstract
Estrogen receptors (ERα and ERβ) mediate the neuroprotection of estrogens against MPTP-induced striatal dopamine (DA) depletion. Pain is an important and distressing symptom in Parkinson's disease (PD). Voltage-gated sodium channels in sensory neurons are involved in the development of neuropathic pain. In this study, MPTP caused changes in nociception and alterations of gene expression of voltage-gated sodium channels in dorsal root ganglion (DRG) neurons in ER knockout (ERKO) mice were investigated. We found that administration of MPTP (11 mg/kg) to WT mice led to an extensive depletion of DA and its two metabolites, αERKO mice were observed to be more susceptible to MPTP toxicity than βERKO or WT mice. In addition, we found that the mRNA levels of TTX-S and TTX-R sodium channel subtypes were differentially affected in MPTP-treated WT animals. The MPTP-induced up-regulation of Nav1.1 and Nav1.9, down-regulation of Nav1.6 in DRG neurons may be through ERβ, up-regulation of Nav1.7 and down-regulation of Nav1.8 are dependent on both ERα and ERβ. Therefore, the MPTP-induced alterations of gene expression of sodium channels in DRG neurons could be an important mechanism to affect excitability and nociceptive thresholds, and the ERs appear to play a role in nociception in PD.
Collapse
Affiliation(s)
- Haixia Ding
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Cao DY, Ji Y, Tang B, Traub RJ. Estrogen receptor β activation is antinociceptive in a model of visceral pain in the rat. THE JOURNAL OF PAIN 2012; 13:685-94. [PMID: 22698981 DOI: 10.1016/j.jpain.2012.04.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 04/19/2012] [Accepted: 04/27/2012] [Indexed: 01/31/2023]
Abstract
UNLABELLED The mechanism underlying estrogen modulation of visceral pain remains unclear. Our previous studies indicate that activation of estrogen receptor α (ERα) enhances visceral pain. The purpose of the present study was to investigate the role of estrogen receptor β (ERβ) activation in spinal processing of visceral stimuli. The effects of selective ERβ agonists on the visceromotor response (VMR) and dorsal horn neuronal responses to colorectal distention (CRD) were tested in ovariectomized and intact female rats. The magnitude of the VMR to CRD was significantly attenuated by ERβ agonists diarylpropionitrile (DPN) and WAY-200070 4 hours after subcutaneous injection. Pretreatment with the estrogen receptor antagonist ICI 182,780 obscured the DPN-evoked attenuation. There was no effect of DPN on the VMR at earlier time points. Subcutaneous and spinal administration of DPN attenuated the response of visceroceptive dorsal horn neurons with a comparable time course. DPN attenuated the VMR in intact rats regardless of estrous cycle stage. The time course of effect of ERβ activation on the visceromotor response and neuronal activity is consistent with transcriptional or translational modulation of neuronal activity. PERSPECTIVE Activation of ERβ is antinociceptive in the colorectal distention model of visceral pain, which may provide a therapeutic target to manage irritable bowel syndrome in the clinic.
Collapse
Affiliation(s)
- Dong-Yuan Cao
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, Maryland 21201, USA
| | | | | | | |
Collapse
|
43
|
Hu F, Wang Q, Wang P, Wang W, Qian W, Xiao H, Wang L. 17β-Estradiol regulates the gene expression of voltage-gated sodium channels: role of estrogen receptor α and estrogen receptor β. Endocrine 2012; 41:274-80. [PMID: 22169964 DOI: 10.1007/s12020-011-9573-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2011] [Accepted: 11/28/2011] [Indexed: 10/14/2022]
Abstract
Estradiol (E2) plays a key role in pain modulation, and the biological effects of E2 are transduced by binding estrogen receptors (ERs). Voltage-gated sodium (Nav) channels are responsible for the generation and propagation of action potentials in the membranes of most neurons and excitable cells. Adult dorsal root ganglion (DRG) neurons can express the ERs (ERα and ERβ), and Nav channels (TTX-S: Nav1.1, Nav1.6, and Nav1.7; and TTX-R: Nav1.8, and Nav1.9). Although E2 modulates Nav channel currents, little is known about the molecular mechanisms involved. In this study, we investigate the mRNA expressions of Nav channel subtypes mediated differentially by the ERs in the DRGs of wild-type (WT) and estrogen receptor knockout (αERKO and βERKO) mice. By means of quantitative real-time PCR, we found that the expressions of Nav1.1, Nav1.7, Nav1.8, and Nav1.9 subtypes were elevated in αERKO and βERKO mice, whereas Nav1.6 mRNA decreased in αERKO, but not in βERKO mice. The mRNA expressions of Nav subtypes were increased in E2-treated WT ovariectomized animals. We also found that E2-regulation of Nav1.1 and Nav1.9 mRNA expressions is dependent on ERα, ERβ, and another ER, whereas E2-regulation of Nav1.8 appears to be in an ERβ-dependent manner.
Collapse
Affiliation(s)
- Fang Hu
- Institute of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing 210029, China
| | | | | | | | | | | | | |
Collapse
|
44
|
Nation T, Buraundi S, Balic A, Southwell B, Newgreen D, Hutson J. Androgen and estrogen receptor expression in the spinal segments of the genitofemoral nerve during testicular descent. J Pediatr Surg 2011; 46:1539-43. [PMID: 21843721 DOI: 10.1016/j.jpedsurg.2010.11.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 11/21/2010] [Indexed: 11/25/2022]
Abstract
AIM During testicular descent (TD), the genitofemoral nerve (GFN) is masculinized by androgen. This study aimed to test whether androgen receptor (AR), estrogen receptor α (ERA), or estrogen receptor β (ERB) are expressed during TD in the GFN spinal segments and dorsal root ganglia (DRG) in normal and flutamide-treated rats. METHODS Time-mated Sprague-Dawley dams were injected with flutamide (75 mg/kg, subcutaneously (S/C) in sunflower oil) on embryonic (E) days 16 to 19. Embryonic and postnatal (P) male L1-2 spinal cord segments were collected (E16, E17, E19, P0, P2, and P4) in control and flutamide-treated groups (n = 5-10). Samples were fixed in 4% paraformaldehyde. Five-micrometer-thick sections were prepared immunohistochemically for AR, ERA, and ERB. RESULTS During TD, ERB was expressed in L1-2 DRG. Surprisingly, AR was not expressed in prenatal DRG, only after P2. There was no ERA expression. Flutamide had no effect on AR, ERB, or ERA expression in the L1-2 DRG during TD. CONCLUSION During the E window of androgen sensitivity, the GFN is not directly masculinized, with little AR expression and no change with flutamide over this period. Estrogen receptor β is expressed in the DRG during TD. However, its relevance is yet to be determined.
Collapse
Affiliation(s)
- Tamara Nation
- Douglas Stephens Surgical Research Laboratory, MCRI, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
45
|
Koszykowska M, Całka J, Gańko M, Jana B. Long-term estradiol-17β administration reduces population of neurons in the sympathetic chain ganglia supplying the ovary in adult gilts. Exp Mol Pathol 2011; 91:353-61. [PMID: 21545801 DOI: 10.1016/j.yexmp.2011.04.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 04/08/2011] [Accepted: 04/08/2011] [Indexed: 12/18/2022]
Abstract
Elevated levels of endogenous estrogens occurring in the course of pathological states of ovaries (follicular cysts, tumors) as well as xenoestrogens may result in hyperestrogenism. In rat, a close relationship between estrogens and sympathetic and sensory neurons supplying the genito-urinary system was reported. Recently, we have shown that long-term estradiol-17β (E(2)) administration affected morphological and immunochemical organization of the sympathetic ovarian neurons in the caudal mesenteric ganglion of adult gilts. In this study, the influence of E(2) overdose on the number and distribution of neurons in the sympathetic chain ganglia (SChG) projecting to the ovary of adult pigs was investigated. The numbers of ovarian dopamine-β-hydroxylase (DβH-), neuropeptide Y (NPY-), somatostatin (SOM-), galanin (GAL-) and estrogen receptors (ERs-) immunoreactive perikarya as well as the density of the intraganglionic nerve fibers containing DβH and/or NPY, SOM, GAL were also determined. On day 3 of the estrous cycle the ovaries of both the control and experimental gilts were injected with retrograde neuronal tracer Fast Blue, to identify the neurons innervating gonads. From day 4 of the estrous cycle to the expected day 20 of the second studied cycle, the experimental gilts were injected with E(2), while the control gilts were receiving oil. After the last E(2)/oil injection, the SChG Th16-S2 were collected and processed for double-labeling immunofluorescence. Injections of E(2): (1) increased the E(2) level in the peripheral blood ~4-5 fold, (2) reduced the total number of Fast Blue-positive postganglionic neurons in the ganglia under investigation, (3) decreased the number of perikarya in the L2-L4 ganglia, (4) reduced the number of perikarya in the ventral, dorsal and central regions of the SChG, (5) decreased the numbers of DβH(+)/NPY(+) and DβH(+)/GAL(+) perikarya and the numbers of DβH(+) but NPY(-), SOM(-) and GAL(-) perikarya in the SChG, (6) decreased the number of perikarya expressing ERs subtype α and β, and (7) decreased the total number of the intraganglionic nerve fibers containing DβH and/or NPY. These results show that long-term E(2) treatment of adult gilts down-regulates the population of both noradrenergic and ERs expressing the SChG ovary supplying neurons. Our findings suggest also that elevated E(2) levels that occur during pathological states may regulate gonadal function(s) by affecting ovary supplying neurons.
Collapse
Affiliation(s)
- Marlena Koszykowska
- Division of Reproductive Endocrinology and Pathophysiology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, 10-747 Olsztyn, Tuwima 10, Poland
| | | | | | | |
Collapse
|
46
|
Inhibition of voltage-gated sodium channels by bisphenol A in mouse dorsal root ganglion neurons. Brain Res 2011; 1378:1-8. [PMID: 21241682 DOI: 10.1016/j.brainres.2011.01.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 12/14/2010] [Accepted: 01/08/2011] [Indexed: 11/22/2022]
Abstract
Bisphenol A (BPA), an estrogenic compound, is contained in cans, polycarbonate bottles, and some dental sealants. Exposure to BPA might have potential toxicological effects on the nervous system. Previous studies have demonstrated that BPA may affect ion channel function, but the effects of BPA on voltage-gated sodium channels are unknown. Herein, we report the effects of BPA on TTX-sensitive (TTX-S) and TTX-resistant (TTX-R) Na+ currents, using a conventional whole-cell patch clamp technique from acutely isolated mouse dorsal root ganglion neurons. BPA inhibited TTX-S Na+ currents and TTX-R Na+ currents, the effects of BPA were rapid, reversible and in a concentration-dependent manner. Moreover, BPA could shift the voltage-gated activation curve for TTX-S Na+ channel in the hyperpolarizing direction without changing that for TTX-R Na+ channel; shift the steady-state inactivation curve for TTX-S Na+ channel in the depolarizing direction without changing that for TTX-R Na+ channel; and lengthen the time course of recovery from inactivation for both TTX-S Na+ current and TTX-R Na+ current. We also found that PKC inhibitor GÖ-6983 and PKA inhibitor H-89 blocked the BPA-induced inhibition of Na+ currents. Considering its complex modulatory effects on voltage-gated sodium channels, BPA might have potential toxicological effects on the nervous system and lead to a change in excitability of nociceptive afferent fibers.
Collapse
|
47
|
Ma B, Yu LH, Fan J, Cong B, He P, Ni X, Burnstock G. Estrogen modulation of peripheral pain signal transduction: involvement of P2X(3) receptors. Purinergic Signal 2011; 7:73-83. [PMID: 21484099 DOI: 10.1007/s11302-010-9212-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Accepted: 12/16/2010] [Indexed: 11/27/2022] Open
Abstract
There is evidence that gonadal hormones may affect the perception of painful stimulation, although the underlying mechanisms remain unclear. This investigation was undertaken to determine whether the adenosine 5'-triphosphate (ATP) receptor subunit, P2X(3), is involved in the modulatory action of estrogen in peripheral pain signal transduction in dorsal root ganglion (DRG). The mechanical pain behavior test, real-time quantitative reverse transcription-polymerase chain reaction analysis, and Western blot methods were used to determine the mean relative concentrations and functions of P2X(3) receptors in DRG in sham, ovariectomized (OVX), and estradiol replacement (OVX+E(2)) female rats and in sham and orchiectomized male rats. The mechanical hyperalgesia appeared after ovariectomy, which was subsequently reversed after estradiol replacement, whereas it was not observed after orchiectomy in male rats. Plantar injection of 2'(3')-O-(2,4,6-trinitrophenyl) ATP (TNP-ATP), a P2X(3) and P2X(2/3) receptor antagonist, resulted in an increase of the pain threshold force in OVX rats while had no effect on sham rats. Furthermore, A-317491, a selective P2X(3)/P2X(2/3) receptor antagonist, significantly reversed the hyperalgesia of OVX rats. Injection of ATP into the plantars also caused a significant increase of the paw withdrawal duration in OVX rats compared with that seen in the sham group, which became substantially attenuated by TNP-ATP. P2X(3) receptors expressed in DRG were significantly increased in both mRNA and protein levels after ovariectomy and then reversed after estrogen replacement, while a similar increase was not observed after orchiectomy in male rats. Furthermore, P2X(3) mRNA was significantly decreased 24 h after the application of 17β-estradiol in a concentration-dependent manner in cultured DRG neurons. ICI 182,780, an estrogen receptor antagonist, blocked the reduction in the protein level. These results suggest that the female gonadal hormone, 17β-estradiol, might participate in the control of peripheral pain signal transduction by modulating P2X(3) receptor-mediated events in primary sensory neurons, probably through genomic mechanisms.
Collapse
Affiliation(s)
- Bei Ma
- Department of Physiology, Shanghai Second Military Medical University, Shanghai, 200433, People's Republic of China,
| | | | | | | | | | | | | |
Collapse
|
48
|
Zhong YQ, Li KC, Zhang X. Potentiation of excitatory transmission in substantia gelatinosa neurons of rat spinal cord by inhibition of estrogen receptor alpha. Mol Pain 2010; 6:92. [PMID: 21143988 PMCID: PMC3016347 DOI: 10.1186/1744-8069-6-92] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Accepted: 12/11/2010] [Indexed: 01/23/2023] Open
Abstract
Background It has been shown that estrogen is synthesized in the spinal dorsal horn and plays a role in modulating pain transmission. One of the estrogen receptor (ER) subtypes, estrogen receptor alpha (ERα), is expressed in the spinal laminae I-V, including substantia gelatinosa (SG, lamina II). However, it is unclear how ERs are involved in the modulation of nociceptive transmission. Results In the present study, a selective ERα antagonist, methyl-piperidino-pyrazole (MPP), was used to test the potential functional roles of spinal ERα in the nociceptive transmission. Using the whole-cell patch-clamp technique, we examined the effects of MPP on SG neurons in the dorsal root-attached spinal cord slice prepared from adult rats. We found that MPP increased glutamatergic excitatory postsynaptic currents (EPSCs) evoked by the stimulation of either Aδ- or C-afferent fibers. Further studies showed that MPP treatment dose-dependently increased spontaneous EPSCs frequency in SG neurons, while not affecting the amplitude. In addition, the PKC was involved in the MPP-induced enhancement of synaptic transmission. Conclusions These results suggest that the selective ERα antagonist MPP pre-synaptically facilitates the excitatory synaptic transmission to SG neurons. The nociceptive transmission evoked by Aδ- and C-fiber stimulation could be potentiated by blocking ERα in the spinal neurons. Thus, the spinal estrogen may negatively regulate the nociceptive transmission through the activation of ERα.
Collapse
Affiliation(s)
- Yan-Qing Zhong
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences
| | | | | |
Collapse
|
49
|
Koszykowska M, Calka J, Szwajca P, Jana B. Long-term estradiol-17β administration decreases the number of neurons in the caudal mesenteric ganglion innervating the ovary in sexually mature gilts. J Reprod Dev 2010; 57:62-71. [PMID: 20881351 DOI: 10.1262/jrd.10-061s] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The effect of estradiol-17β (E(2)) on the number and distribution of neurons in the caudal mesenteric ganglion (CaMG) supplying the ovary of adult pigs was investigated. Also, the numbers of ovarian dopamine-β-hydroxylase (DβH-), neuropeptide Y (NPY-), somatostatin (SOM-), galanin (GAL-) and estrogen receptor (ER)-immunoreactive perikarya as well as the density of the intraganglionic nerve fibers containing DβH and/or NPY, SOM, GAL were determined. E(2) was administered i.m. from day 4 of the first studied estrous cycle to the expected day 20 of the second studied cycle. Injections of E(2) (1) increased the E(2) level in the peripheral blood approximately 4-5 fold, (2) decreased the number of small-sized Fast Blue-positive postganglionic neurons in the CaMG, (3) decreased the number of small perikarya in the ventral, dorsal and central regions of the CaMG, (4) decreased the number of large perikarya in the dorsal and central regions, (5) decreased the number of small and large perikarya in the CaMG that were DβH(+)/NPY(+), (6) decreased the number of small DβH(+) but NPY(-) perikarya, (7) decreased the number of small perikarya coded DβH(+)/SOM(+) and DβH(+)/SOM(-), (8) decreased the number of small DβH(+)/GAL(-) perikarya, (9) decreased the number of small and large perikarya expressing ER subtypes α and β and (10) decreased the total number of nerve fibers in the CaMG containing DβH and/or NPY and DβH and/or GAL. These results show that long-term E(2) treatment of adult gilts downregulates the populations of both noradrenergic and ERs expressing ovarian neurons in the CaMG. Our findings suggest also that elevated E(2) levels that occur during pathological states may regulate gonadal function(s) by affecting ovary supplying neurons.
Collapse
Affiliation(s)
- Marlena Koszykowska
- Division of Reproductive Endocrinology and Pathophysiology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Olsztyn, Poland
| | | | | | | |
Collapse
|
50
|
Spary EJ, Maqbool A, Batten TFC. Changes in oestrogen receptor alpha expression in the nucleus of the solitary tract of the rat over the oestrous cycle and following ovariectomy. J Neuroendocrinol 2010; 22:492-502. [PMID: 20236229 DOI: 10.1111/j.1365-2826.2010.01977.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Oestrogen is capable of modulating autonomic outflow and baroreflex function via actions on groups of neurones in the brainstem. We investigated the presence of oestrogen receptor (ER) alpha in a part of the nucleus of the solitary tract (NTS) associated with central cardiovascular control, aiming to determine whether ERalpha mRNA and protein expression is correlated with levels of circulating oestrogen during the oestrous cycle. Polymerase chain reaction (PCR) detected ERalpha mRNA in the NTS at each stage of the oestrous cycle, from ovariectomised, sham-operated and male rats. Real-time PCR showed variations in ERalpha mRNA expression during the oestrous cycle, with the highest levels seen in oestrus, and lowest levels in metoestrus (P < 0.05 versus oestrus) and proestrus (P < 0.05 versus oestrus). Expression in males was lower than in dioestrus and oestrus females (P < 0.05). After ovariectomy, ERalpha mRNA levels were decreased compared to sham-operated animals (P < 0.01). Confocal fluorescence immunohistochemistry with stereological analysis showed that numbers of ERalpha immunoreactive cell nuclei per mm(3) of tissue in the caudal NTS were significantly greater in proestrus than in other groups of rats (P < 0.05). There were also differences among the groups in the extent of colocalisation of ERalpha in neurones immunoreactive for tyrosine hydroxylase and nitric oxide synthase. These results imply a complex pattern of region-specific oestrogen signalling in the NTS and suggest that ERalpha expression in this important autonomic nucleus may be related to circulating oestrogen levels. This may have consequences for the regulation of autonomic tone and baroreflex sensitivity when oestrogen levels decline, for example following menopause.
Collapse
Affiliation(s)
- E J Spary
- Division of Cardiovascular and Neuronal Remodelling, LIGHT Institute, University of Leeds, Leeds, UK.
| | | | | |
Collapse
|