Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters










Publication year range
1.
Horm Behav ; 158: 105469, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38091929

ABSTRACT

Testosterone may reduce pain in cisgender women and transgender men. Rodents can provide a useful model for investigating physiological effects of hormone therapy. To this end, continuous-release testosterone or blank (placebo) capsules were implanted s.c. into young adult female rats, and three weeks later rats were either ovariectomized or sham-ovariectomized. Testosterone treatment that mimicked previously reported endogenous levels in males eliminated estrous cycling and decreased uterine weight. Testosterone also significantly increased body weight and suppressed the increases in daily wheel running observed in placebo controls over time. Subsequent ovariectomy or sham-ovariectomy decreased wheel running in all groups, but testosterone-treated rats recovered significantly more quickly than did placebo-treated rats. Neither testosterone nor ovariectomy significantly altered hindpaw mechanical threshold. Two weeks after sham/ovariectomy surgery, injection of Complete Freund Adjuvant (CFA) into one hindpaw reduced wheel running and mechanical threshold in all groups; running significantly decreased from the first to second day after CFA in testosterone- but not in placebo-treated rats. Morphine 1.0 but not 3.2 mg/kg increased CFA-suppressed wheel running similarly in all groups, whereas both doses of morphine increased CFA-suppressed mechanical threshold. These data suggest that weeks-long testosterone treatment with or without ovariectomy may provide a useful physiological model of testosterone therapy as used in human gender transition. Although testosterone administered at levels similar to those in gonadally intact males tended to hasten female rats' recovery from surgery, it did not decrease maximal pain-related behaviors after surgery or hindpaw inflammatory insult, nor did it alter opioid antinociception.


Subject(s)
Motor Activity , Testosterone , Animals , Female , Rats , Morphine/pharmacology , Ovariectomy , Pain/drug therapy , Testosterone/pharmacology
2.
Physiol Behav ; 272: 114376, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37820887

ABSTRACT

Fentanyl is a potent analgesic with a rapid onset and short half-life that make it a useful treatment for pain and a lethal drug of abuse. The present study used voluntary home cage wheel running to assess the effect of hindpaw inflammation, fentanyl administration, and spontaneous fentanyl withdrawal. Fentanyl (0.32 or 1.0 mg/kg/day) or placebo osmotic pumps were implanted subcutaneously and rats received an intraplantar injection of Complete Freund's Adjuvant (CFA) or saline. Rats with hindpaw inflammation caused by CFA administration were less active than rats injected with saline into the hindpaw. The antinociceptive effect of 0.32 mg/kg/day of fentanyl was evident as a recovery of wheel running in these rats. Administration of 1 mg/kg/day of fentanyl almost completely inhibited wheel running during the first day in rats with and without hindpaw inflammation. Wheel running increased each subsequent day until the pumps were surgically removed after day 3. Withdrawal from 0.32 or 1 mg/kg/day of fentanyl caused a decrease in wheel running that lasted 2 days in rats without hindpaw inflammation. In contrast, withdrawal was only evident following termination of 1 mg/kg/day of fentanyl in rats with hindpaw inflammation. This decrease in running seemed to persist beyond the 3 days of assessment. These data demonstrate that fentanyl can either depress or restore activity depending on the dose and pain condition. Moreover, termination of 3 days of continuous fentanyl administration resulted in a dose and time dependent decrease in wheel running consistent with opioid withdrawal.


Subject(s)
Fentanyl , Motor Activity , Animals , Rats , Fentanyl/administration & dosage , Freund's Adjuvant/toxicity , Inflammation/complications , Pain/drug therapy
3.
Cannabis Cannabinoid Res ; 8(4): 603-607, 2023 08.
Article in English | MEDLINE | ID: mdl-36862519

ABSTRACT

Introduction: The analgesic effects of delta-9-tetrahydrocannabinol (THC), the primary psychoactive compound in cannabis, have been widely promoted. Unfortunately, animal research is limited by the use of high doses and pain-evoked tests. Motor and psychoactive effects of THC may suppress evoked responses in the absence of antinociceptive effects. Materials and Methods: This study overcomes these problems by assessing the antinociceptive effect of low doses of subcutaneous THC on depression of home cage wheel running caused by hindpaw inflammation. Female and male Long-Evans rats were individually housed in a cage with a running wheel. Results: Female rats ran significantly more than male rats. Administration of Complete Freund's Adjuvant into the right hindpaw produced inflammatory pain that significantly depressed wheel running in female and male rats. Administration of a low dose of THC (0.32, but not 0.56 or 1.0 mg/kg) restored wheel running in the hour after administration in female rats. Administration of these doses had no effect on pain-depressed wheel running in male rats. Conclusions: These data are consistent with previous studies showing greater antinociceptive effects of THC in female compared with male rats. These data extend previous findings by showing that low doses of THC can restore pain-depressed behaviors.


Subject(s)
Motor Activity , Pain , Rats , Female , Animals , Rats, Sprague-Dawley , Rats, Long-Evans , Pain/drug therapy , Analgesics/pharmacology , Analgesics/therapeutic use
4.
Pharmacol Biochem Behav ; 219: 173451, 2022 09.
Article in English | MEDLINE | ID: mdl-35995262

ABSTRACT

The disruptive effects of pain on quality of life are greater in men than in women, but the disruptive effects of opioid administration and withdrawal tend to be greater in women. These sex differences in pain, acute opioid effects, and opioid withdrawal tend to be opposite to sex differences reported in laboratory rats. We hypothesized that sex differences in humans and rats would more closely align if animal research measured quality of life as opposed to traditional evoked behaviors of pain (e.g., nociceptive reflexes) and opioid withdrawal (e.g., wet dog shakes). The present study assessed quality of life in adult female and male rats by measuring voluntary wheel running in the rat's home cage. Hindpaw inflammation induced by administration of Complete Freund's Adjuvant (CFA) into the right hindpaw caused a greater depression of wheel running in male compared to female rats. Twice daily injections of high morphine doses (5-20 mg/kg) and the subsequent morphine withdrawal caused a greater depression of wheel running in female compared to male rats. These sex differences are consistent with human data that shows the impact of pain on quality of life is greater in men than women, but the negative effects of opioid administration and withdrawal are greater in women. The present data indicate that the clinical significance of animal research would be enhanced by shifting the endpoint from pain and opioid evoked behaviors to measures of quality of life such as voluntary wheel running.


Subject(s)
Morphine , Substance Withdrawal Syndrome , Analgesics, Opioid/adverse effects , Animals , Female , Humans , Male , Morphine/adverse effects , Motor Activity , Narcotics/pharmacology , Pain , Quality of Life , Rats , Rats, Sprague-Dawley , Sex Characteristics
5.
Omega (Westport) ; : 302228221101277, 2022 May 12.
Article in English | MEDLINE | ID: mdl-35549588

ABSTRACT

The overarching mission of Death Cafes is to raise people's consciousness about life's temporality so that they make the most of their finite lives. Death Cafés create a space for people to gather and have an open dialogue about death and dying. The current qualitative research study explores the experience of nine participants who engage in Death Cafes as facilitators or attendees. Findings including themes of "The Setting", "The Players", and "The Conversation" explore the experiences of Death Cafe participation. Discussion of findings and implications include death rituals, commercialization of death experiences, and the supportive community of the Death Cafe.

6.
Pharmacol Biochem Behav ; 209: 173251, 2021 10.
Article in English | MEDLINE | ID: mdl-34363827

ABSTRACT

Opioids such as morphine are the most effective treatment for pain, but termination of opioid use can produce severe withdrawal symptoms. The present study models this process by using home cage wheel running to assess well-being as a result of pain, morphine analgesia, and opioid withdrawal. Injection of CFA into the right hindpaw caused a dramatic decrease in wheel running and body weight. Implantation of two morphine pellets (75 mg each) resulted in an increase in body weight on Day 1 of administration and a more gradual restoration of wheel running that was only evident during the dark phase of the circadian cycle on Days 3 and 4 of morphine administration. Continuous morphine administration decreased wheel running during the relatively inactive light phase. These findings are consistent with the clinical goal of pain therapeutics to restore normal activity during the day and facilitate sleep at night. Administration of naloxone (1 mg/kg) on Day 5 of morphine administration depressed wheel running for approximately 4 h and caused an increase in wet dog shakes. Naloxone-precipitated changes were no longer evident 6 h after administration. These findings demonstrate that the use of morphine to treat pain does not protect against opioid withdrawal. Moreover, this study provides additional support for the use of home cage wheel running as a method to assess changes in well-being as a result of pain, analgesia, and opioid withdrawal.


Subject(s)
Inflammation/drug therapy , Morphine/pharmacology , Motor Activity/drug effects , Naloxone/pharmacology , Substance Withdrawal Syndrome/drug therapy , Analgesics, Opioid/pharmacology , Animals , Behavior, Animal/drug effects , Male , Narcotic Antagonists/pharmacology , Opioid-Related Disorders/drug therapy , Pain/drug therapy , Rats , Rats, Sprague-Dawley
7.
Psychopharmacology (Berl) ; 238(10): 2895-2903, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34247265

ABSTRACT

RATIONALE: Social support and opioid replacement therapy are commonly used to treat opioid withdrawal. OBJECTIVE: The present study tested the hypothesis that social housing and buprenorphine administration can restore wheel running depressed by morphine withdrawal in rats. RESULTS: Experiment 1 assessed disruptive side effects of buprenorphine and found that administration of low doses (3.2, 10, & 32 µg/kg, s.c.) had no impact on voluntary wheel running. Experiment 2 assessed the impact of social housing and acute buprenorphine administration (10 µg/kg) on morphine withdrawal. Two 75 mg morphine pellets were implanted for 3 days to induce dependence. Removal of the morphine pellets caused a decrease in body weight, increase in wet dog shakes, and depression of wheel running during the normally active dark phase of the circadian cycle. Social housing restored wheel running and reduced the number of wet dog shakes but did not affect body weight. Administration of buprenorphine restored wheel running depressed by morphine withdrawal for 2 days in individually housed rats and produced time-dependent changes in socially housed rats: Depression of wheel running in the 3 h following administration and restoration of running subsequently compared to saline-treated controls. CONCLUSIONS: The impact of buprenorphine and social housing to reduce the effect of morphine withdrawal in rats is consistent with the use of opioid substitution therapy and psychotherapy/social support to treat opioid withdrawal in humans. These data provide further validation for the clinical relevance for the use of wheel running to assess spontaneous opioid withdrawal.


Subject(s)
Buprenorphine , Motor Activity , Substance Withdrawal Syndrome , Animals , Housing , Morphine/pharmacology , Motor Activity/drug effects , Narcotics/pharmacology , Rats , Substance Withdrawal Syndrome/drug therapy
8.
J Pain ; 22(9): 1040-1047, 2021 09.
Article in English | MEDLINE | ID: mdl-33727159

ABSTRACT

Inflammatory Bowel Disease (IBD) is a life-long disorder that often begins between the ages of 15 and 30. Anecdotal reports suggest cannabinoids may be an effective treatment. This study sought to determine whether home cage wheel running is an effective method to assess IBD, and whether Tetrahydrocannabinol (THC), the primary psychoactive compound in cannabis, can restore wheel running depressed by IBD. Adolescent and adult female Sprague-Dawley rats were individually housed in a cage with a running wheel. Rats were injected with trinitrobenzene sulphonic acid (TNBS) into the rectum to induce IBD-like symptoms. One day later, both vehicle and TNBS treated rats were injected with a low dose of THC (0.32 mg/kg, s.c.) or vehicle. Administration of TNBS depressed wheel running in adolescent and adult rats. No antinociceptive effect of THC was evident when administered 1 day after TNBS. In fact, administration of THC prolonged TNBS-induced depression of wheel running for over 5 days in adolescent and adult rats. These results show that home cage wheel running is depressed by TNBS-induced IBD, making it a useful tool to evaluate the behavioral consequences of IBD, and that administration of THC, instead of producing antinociception, exacerbates TNBS-induced IBD. PERSPECTIVE: This article advances research on inflammatory bowel disease in two important ways: 1) Home cage wheel running is a new and sensitive tool to assess the behavioral consequences of IBD in adolescent and adult rats; and 2) Administration of the cannabinoid THC exacerbates the negative behavioral effects of IBD.


Subject(s)
Behavior, Animal/drug effects , Behavior, Animal/physiology , Cannabinoid Receptor Agonists/pharmacology , Dronabinol/pharmacology , Inflammatory Bowel Diseases/physiopathology , Running/physiology , Age Factors , Animals , Cannabinoid Receptor Agonists/administration & dosage , Cannabinoid Receptor Agonists/adverse effects , Disease Models, Animal , Dronabinol/administration & dosage , Dronabinol/adverse effects , Female , Inflammatory Bowel Diseases/complications , Rats , Rats, Sprague-Dawley , Symptom Flare Up
9.
Behav Brain Res ; 396: 112912, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32949642

ABSTRACT

The increased use of opioids to treat pain has led to a dramatic increase in opioid abuse. Our previous data indicate that pain may facilitate the development of opioid abuse by increasing the magnitude and duration of opioid withdrawal. The present study tested the hypothesis that social housing would facilitate recovery of activity depressed by pain and opioid withdrawal. Male Sprague Dawley rats were housed either in pairs or alone and then moved to a cage with a running wheel for 6 h daily to assess pain- and opioid withdrawal-induced depression of wheel running. Rats were implanted with two morphine (75 mg each) or placebo pellets to induce opioid dependence and were simultaneously injected with Complete Freund's Adjuvant or saline into the right hind paw to induce persistent inflammatory pain. Hind paw inflammation depressed wheel running whether rats were implanted with a morphine or placebo pellet. Pair-housed rats showed greater recovery of wheel running than individually housed rats. Spontaneous morphine withdrawal precipitated by removal of the morphine pellets caused a reduction in wheel running that was greater in rats with hind paw inflammation compared to pain free rats. Social housing facilitated recovery from withdrawal in rats with hind paw inflammation, but slowed recovery in pain free rats. These data suggest that social housing facilitates recovery by reducing pain both before and during opioid withdrawal. Our findings are consistent with previous studies showing that social buffering reduces pain-evoked responses.


Subject(s)
Analgesics, Opioid/administration & dosage , Depression/physiopathology , Housing, Animal , Locomotion/physiology , Morphine/administration & dosage , Nociceptive Pain/physiopathology , Opioid-Related Disorders/physiopathology , Social Behavior , Substance Withdrawal Syndrome/physiopathology , Animals , Behavior, Animal/physiology , Depression/rehabilitation , Freund's Adjuvant , Inflammation/chemically induced , Inflammation/complications , Male , Nociceptive Pain/etiology , Nociceptive Pain/rehabilitation , Rats , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/rehabilitation
10.
Behav Brain Res ; 397: 112953, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33031872

ABSTRACT

Opioid abuse is a major health problem. The objective of the present study was to evaluate the potentially disruptive side effects and therapeutic potential of a novel antagonist (D24M) of the mu-/delta-opioid receptor (MOR/DOR) heterodimer in male rats. Administration of high doses of D24M (1 & 10 nmol) into the lateral ventricle did not disrupt home cage wheel running. Repeated twice daily administration of increasing doses of morphine (5-20 mg/kg) over 5 days depressed wheel running and induced antinociceptive tolerance measured with the hot plate test. Administration of D24M had no effect on morphine tolerance, but tended to prolong morphine antinociception in non-tolerant rats. Spontaneous morphine withdrawal was evident as a decrease in body weight, a reduction in wheel running and an increase in sleep during the normally active dark phase of the circadian cycle, and an increase in wheel running and wakefulness in the normally inactive light phase. Administration of D24M during the dark phase on the third day of withdrawal had no effect on wheel running. These data provide additional evidence for the clinical relevance of home cage wheel running as a method to assess spontaneous opioid withdrawal in rats. These data also demonstrate that blocking the MOR/DOR heterodimer does not produce disruptive side effects or block the antinociceptive effects of morphine. Although administration of D24M had no effect on morphine withdrawal, additional studies are needed to evaluate withdrawal to continuous morphine administration and other opioids in rats with persistent pain.


Subject(s)
Behavior, Animal/drug effects , Morphine/pharmacology , Narcotic Antagonists/pharmacology , Narcotics/pharmacology , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, mu/antagonists & inhibitors , Sleep/drug effects , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/physiopathology , Animals , Disease Models, Animal , Male , Morphine/administration & dosage , Narcotic Antagonists/administration & dosage , Narcotics/administration & dosage , Rats , Rats, Sprague-Dawley , Running
11.
Behav Pharmacol ; 32(2&3): 142-152, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33079736

ABSTRACT

Chronic pain affects approximately one-third of the population worldwide. The primary goal of animal research is to understand the neural mechanisms underlying pain so better treatments can be developed. Despite an enormous investment in time and money, almost no novel treatments for pain have been developed. There are many factors that contribute to this lack of translation in drug development. The mismatch between the goals of drug development in animals (inhibition of pain-evoked responses) and treatment in humans (restoration of function) is a major problem. To solve this problem, a number of pain-depressed behavioral tests have been developed to assess changes in normal behavior in laboratory animals. The use of home cage wheel running as a pain assessment tool is especially useful in that it is easy to use, provides an objective measurement of the magnitude and duration of pain, and is a clinically relevant method to screen novel drugs. Pain depresses activity in humans and animals, and effective analgesic treatments restore activity. Unlike traditional pain-evoked tests (e.g., hot plate, tail flick, von Frey test), restoration of home cage wheel running evaluates treatments for both antinociceptive efficacy and the absence of disruptive side effects (e.g., sedation, paralysis, nausea). This article reviews the literature using wheel running to assess pain and makes the case for home cage wheel running as an effective and clinically relevant method to screen novel analgesics for therapeutic potential.


Subject(s)
Analgesics/pharmacology , Drug Development/methods , Pain/drug therapy , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Humans , Pain/physiopathology , Pain Measurement , Running/physiology
12.
Eur J Pain ; 24(3): 617-624, 2020 03.
Article in English | MEDLINE | ID: mdl-31785128

ABSTRACT

BACKGROUND: Morphine and fentanyl are two of the most commonly used opioids to treat pain. Although both opioids produce antinociception by binding to mu-opioid receptors (MOR), they appear to act via distinct signalling pathways. OBJECTIVE: This study will reveal whether differences in morphine and fentanyl antinociception are the result of selective activation of G-protein signalling and/or selective activation of pre- or postsynaptic MORs. METHODS: The contribution of each mechanism to morphine and fentanyl antinociception was assessed by microinjecting drugs to alter G-protein signalling or block potassium channels linked to pre- and postsynaptic MORs in the ventrolateral periaqueductal gray (PAG) of male Sprague-Dawley rats. RESULTS: Both morphine and fentanyl produced a dose-dependent antinociception when microinjected into the PAG. Enhancement of intracellular G-protein signalling by microinjection of the Regulator of G-protein Signalling 4 antagonist CCG-63802 into the PAG enhanced the antinociceptive potency of morphine, but not fentanyl. Microinjection of α-dendrotoxin into the PAG to block MOR activation of presynaptic Kv + channels caused a significant rightward shift in the dose-response curve of both morphine and fentanyl. Microinjection of tertiapin-Q to block MOR activation of postsynaptic GIRK channels caused a larger shift in the dose-response curve for fentanyl than morphine antinociception. CONCLUSIONS: These findings reveal different PAG signalling mechanisms for morphine and fentanyl antinociception. In contrast with fentanyl, the antinociceptive effects of morphine are mediated by G-protein signalling primarily activated by presynaptic MORs. SIGNIFICANCE: Microinjection of the opioids morphine and fentanyl into the periaqueductal gray (PAG) produce antinociception via mu-opioid receptor signalling. This study reveals differences in the signalling mechanisms underlying morphine and fentanyl antinociception in the PAG. In contrast with fentanyl, morphine antinociception is primarily mediated by presynaptic opioid receptors and is enhanced by blocking RGS proteins.


Subject(s)
Morphine , Periaqueductal Gray , Analgesics, Opioid/pharmacology , Animals , Drug Tolerance , Fentanyl/pharmacology , Male , Microinjections , Morphine/pharmacology , Rats , Rats, Sprague-Dawley
13.
J Pain ; 20(9): 1040-1047, 2019 09.
Article in English | MEDLINE | ID: mdl-30853505

ABSTRACT

Tolerance to the antinociceptive effect of mu-opioid receptor agonists, such as morphine and fentanyl, greatly limits their effectiveness for long-term use to treat pain. Clinical studies have shown that combination therapy and opioid rotation can be used to enhance opioid-induced antinociception once tolerance has developed. The mechanism and brain regions involved in these processes are unknown. The purpose of this study was to evaluate the contribution of the ventrolateral periaqueductal gray (vlPAG) to antinociceptive tolerance and cross-tolerance between administration and co-administration of morphine and fentanyl. Tolerance was induced by pretreating rats with morphine or fentanyl or low-dose combination of morphine and fentanyl into the vlPAG followed by an assessment of the cross-tolerance to the other opioid. In addition, tolerance to the combined treatment was assessed. Cross-tolerance did not develop between repeated vlPAG microinjections of morphine and fentanyl. Likewise, there was no evidence of cross-tolerance from morphine or fentanyl to the co-administration of morphine and fentanyl. Co-administration did not cause cross-tolerance to fentanyl. Cross-tolerance was only evident to morphine or morphine and fentanyl combined in rats pretreated with co-administration of low doses of morphine and fentanyl. This finding is consistent with the functionally selective signaling that has been reported for antinociception and tolerance after morphine and fentanyl binding to the mu-opioid receptor. This research supports the notion that combination therapy and opioid rotation may be useful clinical practices to decrease opioid tolerance and other side effects. PERSPECTIVE: This preclinical study shows that there is a decrease in cross-tolerance between morphine and fentanyl within the periaqueductal gray, which is a key brain region in opioid antinociception and tolerance.


Subject(s)
Analgesics, Opioid/pharmacology , Fentanyl/pharmacology , Morphine/pharmacology , Nociception/drug effects , Pain Threshold/drug effects , Periaqueductal Gray/drug effects , Animals , Drug Tolerance , Male , Pain Measurement , Pain Perception/drug effects , Rats , Rats, Sprague-Dawley
14.
Neurosci Biobehav Rev ; 100: 335-343, 2019 05.
Article in English | MEDLINE | ID: mdl-30885811

ABSTRACT

The primary objective of preclinical pain research is to improve the treatment of pain. Decades of research using pain-evoked tests has revealed much about mechanisms but failed to deliver new treatments. Evoked pain-tests are often limited because they ignore spontaneous pain and motor or disruptive side effects confound interpretation of results. New tests have been developed to focus more closely on clinical goals such as reducing pathological pain and restoring function. The objective of this review is to describe and discuss several of these tests. We focus on: Grimace Scale, Operant Behavior, Wheel Running, Burrowing, Nesting, Home Cage Monitoring, Gait Analysis and Conditioned Place Preference/ Aversion. A brief description of each method is presented along with an analysis of the advantages and limitations. The pros and cons of each test will help researchers identify the assessment tool most appropriate to meet their particular objective to assess pain in rodents. These tests provide another tool to unravel the mechanisms underlying chronic pain and help overcome the translational gap in drug development.


Subject(s)
Disease Models, Animal , Pain Measurement/methods , Pain/psychology , Animals , Behavior, Animal , Conditioning, Classical , Conditioning, Operant , Gait Analysis , Mice , Motor Activity , Nesting Behavior , Nociception , Rats , Translational Research, Biomedical/methods
15.
Appl Environ Microbiol ; 85(3)2019 02 01.
Article in English | MEDLINE | ID: mdl-30446559

ABSTRACT

Diet is a major determinant of intestinal microbiome composition. While studies have evaluated microbiome responses to diet variation, less is understood about how the act of feeding influences the microbiome, independent of diet type. Here, we use the clownfish Premnas biaculeatus, a species reared commonly in ornamental marine aquaculture, to test how the diversity, predicted gene content, and gene transcription of the microbiome vary over a 2-day diurnal period with a single daily feeding event. This study used fish fed four times daily, once daily, or every 3 days prior to the diurnal period, allowing us also to test how feeding frequency affected microbiome diversity. The amount of time between feedings had no effect on baseline diversity of the microbiome. In contrast, the act of feeding itself caused a significant short-term change in the microbiome, with microbiome diversity, predicted gene content, and gene transcription varying significantly between time points immediately before and 1.5 hours postfeeding. Variation was driven by abundance shifts involving exact sequence variants (ESVs), with one ESV identified as Photobacterium sp. increasing from <0.5% of sequences immediately prefeeding to 34% at 1.5 h postfeeding. Other ESVs from a range of microbial groups also increased dramatically after feeding, with the majority also detected in the food. One ESV identified as Clostridium perfringens represented up to 55% of sequences but did not vary significantly over the diurnal period and was not detected in the food. Postfeeding samples were enriched in transcripts and predicted genes for social interactions, cell motility, and coping with foreign DNA, whereas time points farther from feeding were enriched in genes of diverse catabolic and biosynthetic functions. These results confirm feeding as a significant destabilizing force in clownfish intestinal microbiomes, likely due to both input of cells attached to food and stimulation of resident microbes. Microbes such as Photobacterium may episodically transition from environmental reservoirs to growth in the gut, likely in association with food particles. This transition may be facilitated by functions for navigating a new environment and interacting with neighboring microbes and host cells. Other taxa, such as Clostridium, are comparatively stable intestinal members and less likely to be affected by passing food. Conclusions about microbiome ecology may therefore differ based on when samples were collected relative to the last feeding.IMPORTANCE Despite extensive study of intestinal microbiome diversity and the role of diet type in structuring gut microbial communities, we know very little about short-term changes in the intestinal microbiome as a result of feeding alone. Sampling microbiomes over a feeding cycle will allow us to differentiate opportunistic, feeding-responsive microbes from resident, potentially commensal members of the gut community. Also, since feeding has the potential to alter microbiome structure, sampling at different points relative to the last feeding event will likely yield different conclusions about microbiome composition and function. This variation should be addressed in comparative microbiome studies. Our study contributes to knowledge of short-term changes in the gut microbiome associated with feeding events.


Subject(s)
Bacteria/genetics , Bacterial Proteins/genetics , Fishes/microbiology , Intestines/microbiology , Microbiota , Animal Feed/analysis , Animals , Bacteria/classification , Bacteria/isolation & purification , Bacterial Proteins/metabolism , Feeding Behavior , Fishes/metabolism , Transcription, Genetic
16.
Pain Rep ; 3(4): e664, 2018.
Article in English | MEDLINE | ID: mdl-30123857

ABSTRACT

INTRODUCTION: Corneal nerves mediate pain from the ocular surface, lacrimation, and blinking, all of which protect corneal surface homeostasis and help preserve vision. Because pain, lacrimation and blinking are rarely assessed at the same time, it is not known whether these responses and their underlying mechanisms have similar temporal dynamics after acute corneal injury. METHODS: We examined changes in corneal nerve density, evoked and spontaneous pain, and ocular homeostasis in Sprague-Dawley male rats after a superficial epithelial injury with heptanol. We also measured changes in calcitonin gene-related peptide (CGRP), which has been implicated in both pain and epithelial repair. RESULTS: Hyperalgesia was seen 24 hours after abrasion injury, while basal tear production was normal. One week after abrasion injury, pain responses had returned to baseline levels and dry eye symptoms emerged. There was no correlation between epithelial nerve density and pain responses. Expression of both ATF3 (a nerve injury marker) and CGRP increased in trigeminal ganglia 24 hours after injury when hyperalgesia was seen, and returned to normal one week later when pain behavior was normal. These molecular changes were absent in the contralateral ganglion, despite reductions in corneal epithelial nerve density in the uninjured eye. By contrast, CGRP was upregulated in peripheral corneal endings 1 week after injury, when dry eye symptoms emerged. CONCLUSION: Our results demonstrate dynamic trafficking of CGRP within trigeminal sensory nerves following corneal injury, with elevations in the ganglion correlated with pain behaviors and elevations in peripheral endings correlated with dry eye symptoms.

17.
Behav Pharmacol ; 29(5): 469-472, 2018 08.
Article in English | MEDLINE | ID: mdl-29462111

ABSTRACT

The potential of [INCREMENT]-tetrahydrocannabinol (THC) as a treatment for migraine depends on antinociceptive efficacy with repeated administration. Although morphine has good antinociceptive efficacy, repeated administration causes medication overuse headache (MOH) - a condition in which the intensity/frequency of migraine increases. The present study compared the effect of repeated morphine or THC administration on the magnitude and duration of migraine-like pain induced by a microinjection of allyl isothiocyanate (AITC) onto the dura mater of female rats. Acute administration of THC or morphine prevented AITC-induced depression of wheel running. This antinociception was maintained in rats treated repeatedly with THC, but not following repeated administration of morphine. Moreover, repeated morphine, but not THC administration, extended the duration of AITC-induced depression of wheel running. These data indicate that tolerance and MOH develop rapidly to morphine administration. The lack of tolerance and MOH to THC indicates that THC may be an especially effective long-term treatment against migraine.


Subject(s)
Dronabinol/pharmacology , Migraine Disorders/drug therapy , Analgesics, Opioid/pharmacology , Animals , Dronabinol/metabolism , Drug Tolerance , Dura Mater/drug effects , Female , Headache Disorders, Secondary/metabolism , Headache Disorders, Secondary/physiopathology , Injections, Intraperitoneal , Isothiocyanates , Migraine Disorders/metabolism , Models, Animal , Morphine/metabolism , Morphine/pharmacology , Motor Activity/drug effects , Pain/drug therapy , Pain Measurement/drug effects , Rats , Rats, Sprague-Dawley
18.
Eur J Pharmacol ; 818: 271-277, 2018 Jan 05.
Article in English | MEDLINE | ID: mdl-29111112

ABSTRACT

Current anti-migraine treatments have limited efficacy and many side effects. Although anecdotal evidence suggests that marijuana is useful for migraine, this hypothesis has not been tested in a controlled experiment. Thus, the present study tested whether administration of ∆9-tetrahydrocannabinol (THC) produces anti-migraine effects in the female rat. Microinjection of the TRPA1 agonist allyl isothiocyanate (AITC) onto the dura mater produced migraine-like pain for 3h as measured by depression of home cage wheel running. Concurrent systemic administration of 0.32 but not 0.1mg/kg of THC prevented AITC-induced depression of wheel running. However, 0.32mg/kg was ineffective when administered 90min after AITC. Administration of a higher dose of THC (1.0mg/kg) depressed wheel running whether rats were injected with AITC or not. Administration of a CB1, but not a CB2, receptor antagonist attenuated the anti-migraine effect of THC. These data suggest that: 1) THC reduces migraine-like pain when administered at the right dose (0.32mg/kg) and time (immediately after AITC); 2) THC's anti-migraine effect is mediated by CB1 receptors; and 3) Wheel running is an effective method to assess migraine treatments because only treatments producing antinociception without disruptive side effects will restore normal activity. These findings support anecdotal evidence for the use of cannabinoids as a treatment for migraine in humans and implicate the CB1 receptor as a therapeutic target for migraine.


Subject(s)
Dronabinol/pharmacology , Migraine Disorders/drug therapy , Animals , Dronabinol/therapeutic use , Female , Migraine Disorders/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/metabolism , Time Factors
19.
Behav Pharmacol ; 29(2 and 3-Spec Issue): 234-240, 2018 04.
Article in English | MEDLINE | ID: mdl-29256893

ABSTRACT

Dopamine neurons in the ventrolateral periaqueductal gray (PAG) have been reported to contribute to antinociception. The objective of this study was to determine how this dopamine-mediated antinociception differs from what is known about morphine-induced antinociception. Microinjection of the dopamine receptor agonist apomorphine into the PAG produced a dose-dependent increase in hot plate latency and a decrease in open field activity that was greater in male than in female rats. The peak antinociceptive effect occurred 5 min after apomorphine administration. Surprisingly, the antinociceptive potency of apomorphine was enhanced following systemic administration of the opioid receptor antagonist naloxone in male, but not in female rats. The antinociceptive potency of microinjecting apomorphine into the ventrolateral PAG in male and female rats was also enhanced following twice-daily injections for 2 days. The characteristics of apomorphine-induced antinociception differ from previous reports of morphine antinociception following PAG microinjections in that morphine antinociception peaks at 15 min, is blocked by naloxone, and is susceptible to tolerance with repeated administration. These results indicate that apomorphine-induced antinociception is distinct from opioid-induced antinociception, and that dopamine receptor agonists may provide a novel approach to pain modulation.


Subject(s)
Apomorphine/pharmacology , Pain/physiopathology , Periaqueductal Gray/metabolism , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Animals , Dopamine , Dopamine Agonists/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Drug Tolerance , Female , Male , Microinjections , Morphine/pharmacology , Narcotic Antagonists , Pain/drug therapy , Rats , Rats, Sprague-Dawley , Receptors, Opioid/metabolism
20.
Pharmacol Biochem Behav ; 156: 10-15, 2017 05.
Article in English | MEDLINE | ID: mdl-28366799

ABSTRACT

Opioid withdrawal in humans is often subtle and almost always spontaneous. In contrast, most preclinical studies precipitate withdrawal by administration of an opioid receptor antagonist such as naloxone. These animal studies rely on measurement of physiological symptoms (e.g., wet dog shakes) in the period immediately following naloxone administration. To more closely model the human condition, we tested the hypothesis that depression of home cage wheel running will provide an objective method to measure the magnitude and duration of spontaneous morphine withdrawal. Rats were allowed access to a running wheel in their home cage for 8days prior to implantation of two 75mg morphine or placebo pellets. The pellets were removed 3 or 5days later to induce spontaneous withdrawal. In normal pain-free rats, removal of the morphine pellets depressed wheel running for 48h compared to rats that had placebo pellets removed. Morphine withdrawal-induced depression of wheel running was greatly enhanced in rats with persistent inflammatory pain induced by injection of Complete Freund's Adjuvant (CFA) into the hindpaw. Removal of the morphine pellets following 3days of treatment depressed wheel running in these rats for over 6days. These data demonstrate that home cage wheel running provides an objective and more clinically relevant method to assess spontaneous morphine withdrawal compared to precipitated withdrawal in laboratory rats. Moreover, the enhanced withdrawal in rats with persistent inflammatory pain suggests that pain patients may be especially susceptible to opioid withdrawal.


Subject(s)
Morphine/adverse effects , Pain/physiopathology , Physical Conditioning, Animal , Substance Withdrawal Syndrome , Animals , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...