opioid sensitivity
Recently Published Documents


TOTAL DOCUMENTS

43
(FIVE YEARS 12)

H-INDEX

12
(FIVE YEARS 1)

2021 ◽  
Vol 35 (S1) ◽  
Author(s):  
Nicholas Burgraff ◽  
Nathan Baertsch ◽  
Nicholas Bush ◽  
Jan‐Marino Ramirez

2021 ◽  
Vol 35 (S1) ◽  
Author(s):  
Elizabeth Jaeckel ◽  
Alberto Perez‐Medina ◽  
Yoani Herrera ◽  
Erwin Arias‐Hervert ◽  
William Birdsong

2021 ◽  
Vol 35 (S1) ◽  
Author(s):  
Jordan Bateman ◽  
Erica Levitt
Keyword(s):  

2021 ◽  
Vol 11 (4) ◽  
pp. 315
Author(s):  
Lynn M. Oswald ◽  
Kelly E. Dunn ◽  
David A. Seminowicz ◽  
Carla L. Storr

A robust body of research has shown that traumatic experiences occurring during critical developmental periods of childhood when neuronal plasticity is high increase risks for a spectrum of physical and mental health problems in adulthood, including substance use disorders. However, until recently, relatively few studies had specifically examined the relationships between early life stress (ELS) and opioid use disorder (OUD). Associations with opioid use initiation, injection drug use, overdose, and poor treatment outcome have now been demonstrated. In rodents, ELS has also been shown to increase the euphoric and decrease antinociceptive effects of opioids, but little is known about these processes in humans or about the neurobiological mechanisms that may underlie these relationships. This review aims to establish a theoretical model that highlights the mechanisms by which ELS may alter opioid sensitivity, thereby contributing to future risks for OUD. Alterations induced by ELS in mesocorticolimbic brain circuits, and endogenous opioid and dopamine neurotransmitter systems are described. The limited but provocative evidence linking these alterations with opioid sensitivity and risks for OUD is presented. Overall, the findings suggest that better understanding of these mechanisms holds promise for reducing vulnerability, improving prevention strategies, and prescribing guidelines for high-risk individuals.


2020 ◽  
pp. bmjspcare-2020-002705
Author(s):  
Lana Ferguson ◽  
Stacey Hooper

This case report describes the use of dexmedetomidine for refractory cancer pain management in a patient with significant pelvic disease due to metastatic urothelial cancer. Specifically, the management of increased opioid sensitivity secondary to dexmedetomidine is discussed. Further, the phenomenon of dexmedetomidine withdrawal syndrome and our management of this is addressed.


2020 ◽  
Vol 40 (34) ◽  
pp. 6522-6535 ◽  
Author(s):  
Anibal Garza Carbajal ◽  
Alexis Bavencoffe ◽  
Edgar T. Walters ◽  
Carmen W. Dessauer

2020 ◽  
Vol 34 (S1) ◽  
pp. 1-1
Author(s):  
Nicholas J. Burgraff ◽  
Nathan A. Baertsch ◽  
Jan-Marino Ramirez

2020 ◽  
Vol 34 (S1) ◽  
pp. 1-1
Author(s):  
Jordan Bateman ◽  
Brandon Reid ◽  
Sebastian Maletz ◽  
Erica Levitt
Keyword(s):  

eLife ◽  
2020 ◽  
Vol 9 ◽  
Author(s):  
Eduardo Javier López Soto ◽  
Diane Lipscombe

Cell-specific alternative splicing modulates myriad cell functions and is disrupted in disease. The mechanisms governing alternative splicing are known for relatively few genes and typically focus on RNA splicing factors. In sensory neurons, cell-specific alternative splicing of the presynaptic CaV channel Cacna1b gene modulates opioid sensitivity. How this splicing is regulated is unknown. We find that cell and exon-specific DNA hypomethylation permits CTCF binding, the master regulator of mammalian chromatin structure, which, in turn, controls splicing in a DRG-derived cell line. In vivo, hypomethylation of an alternative exon specifically in nociceptors, likely permits CTCF binding and expression of CaV2.2 channel isoforms with increased opioid sensitivity in mice. Following nerve injury, exon methylation is increased, and splicing is disrupted. Our studies define the molecular mechanisms of cell-specific alternative splicing of a functionally validated exon in normal and disease states – and reveal a potential target for the treatment of chronic pain.


Author(s):  
Eduardo Javier Lopez Soto ◽  
Diane Lipscombe

SummaryCell-specific alternative splicing modulates myriad cell functions and this process is disrupted in disease. The mechanisms governing alternative splicing are known for relatively few genes and typically focus on RNA splicing factors. In sensory neurons, cell-specific alternative splicing of the presynaptic voltage-gated calcium channel Cacna1b gene modulates opioid sensitivity. How this splicing is regulated has remained unknown. We find that cell-specific exon DNA hypomethylation permits binding of CTCF, the master regulator of chromatin structure in mammals, which, in turn, controls splicing in noxious heat-sensing nociceptors.Hypomethylation of an alternative exon specifically in nociceptors allows for CTCF binding, and expression of CaV2.2 channels with increased opioid sensitivity. Following nerve injury, exon methylation is increased, and splicing is disrupted. Our studies define the molecular mechanisms of cell-specific alternative splicing of a functionally validated exon in normal and disease states – and reveal a potential target for the treatment of chronic pain.HighlightsThe molecular basis of cell-specific splicing of a synaptic calcium channel gene.Splicing controlled by cell-specific exon hypomethylation and CTCF binding.Peripheral nerve injury disrupts exon hypomethylation and splicing.Targeted demethylation of exon by dCAS9-TET modifies alternative splicing.GRAPHICAL ABSTRACTCell-specific epigenetic modifications in a synaptic calcium ion channel gene controls cell-specific splicing in normal and neuropathic pain.In naïve animals, in most neurons, Cacna1b e37a locus is hipermethylated (5-mC) and CTCF does not bind this locus. During splicing, e37a is skipped and Cacna1b mRNAs include e37b. In contrast, in Trpv1-lineage neurons, Cacna1b e37a locus is hypomethylated and is permissive for CTCF binding. CTCF promotes e37a inclusion and both Cacna1b e37a and e37b mRNAs are expressed. E37a confers strong sensitivity to the Cav2.2 channel to inhibition by μ-opioid receptors (μOR). Morphine is more effective at inhibiting e37a-containing Cav2.2 channels. After peripheral nerve injury that results in pathological pain, methylation level of Cacna1b e37a locus is increased, CTCF binding is impaired, and Cacna1b e37a mRNA levels are decreased. This disrupted splicing pattern is associated with reduced efficacy of morphine in vivo.


Sign in / Sign up

Export Citation Format

Share Document