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










Publication year range
2.
bioRxiv ; 2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38645173

ABSTRACT

Alcohol use disorders (AUDs) impose an enormous societal and financial burden, and world-wide, alcohol misuse is the 7th leading cause of premature death1. Despite this, there are currently only 3 FDA approved pharmacological treatments for the treatment of AUDs in the United States. The neurotensin (Nts) system has long been implicated in modulating behaviors associated with alcohol misuse. Recently, a novel compound, SBI-553, that biases the action of Nts receptor 1 (NTSR1) activation, has shown promise in preclinical models of psychostimulant misuse. Here we investigate the efficacy of this compound to alter ethanol-mediated behaviors in a comprehensive battery of experiments assessing ethanol consumption, behavioral responses to ethanol, sensitivity to ethanol, and ethanol metabolism. Additionally, we investigated behavior in avoidance and cognitive assays to monitor potential side effects of SBI-553. We find that SBI-553 reduces binge-like ethanol consumption in mice without altering avoidance behavior or novel object recognition. We also observe sex-dependent differences in physiological responses to sequential ethanol injections in mice. In rats, we show that SBI-553 attenuates sensitivity to the interoceptive effects of ethanol (using a Pavlovian drug discrimination task). Our data suggest that targeting NTSR1 signaling may be promising to attenuate alcohol misuse, and adds to a body of literature that suggests NTSR1 may be a common downstream target involved in the psychoactive effects of multiple reinforcing substances.

3.
Neuropharmacology ; 234: 109544, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37055008

ABSTRACT

Strong expression of the G protein-coupled receptor (GPCR) neurotensin receptor 1 (NTR1) in ventral tegmental area (VTA) dopamine (DA) neurons and terminals makes it an attractive target to modulate DA neuron activity and normalize DA-related pathologies. Recent studies have identified a novel class of NTR1 ligand that shows promising effects in preclinical models of addiction. A lead molecule, SBI-0654553 (SBI-553), can act as a positive allosteric modulator of NTR1 ß-arrestin recruitment while simultaneously antagonizing NTR1 Gq protein signaling. Using cell-attached recordings from mouse VTA DA neurons we discovered that, unlike neurotensin (NT), SBI-553 did not independently increase spontaneous firing. Instead, SBI-553 blocked the NT-mediated increase in firing. SBI-553 also antagonized the effects of NT on dopamine D2 auto-receptor signaling, potentially through its inhibitory effects on G-protein signaling. We also measured DA release directly, using fast-scan cyclic voltammetry in the nucleus accumbens and observed antagonist effects of SBI-553 on an NT-induced increase in DA release. Further, in vivo administration of SBI-553 did not notably change basal or cocaine-evoked DA release measured in NAc using fiber photometry. Overall, these results indicate that SBI-553 blunts NT's effects on spontaneous DA neuron firing, D2 auto-receptor function, and DA release, without independently affecting these measures. In the presence of NT, SBI-553 has an inhibitory effect on mesolimbic DA activity, which could contribute to its efficacy in animal models of psychostimulant use.


Subject(s)
Dopamine D2 Receptor Antagonists , Dopamine , Dopaminergic Neurons , Neurotensin , Nucleus Accumbens , Receptors, Neurotensin , Ventral Tegmental Area , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/physiology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Nucleus Accumbens/metabolism , Dopamine/metabolism , Male , Female , Animals , Mice , Mice, Inbred C57BL , Presynaptic Terminals/metabolism , Presynaptic Terminals/physiology , Action Potentials/drug effects , Receptors, Neurotensin/antagonists & inhibitors , Receptors, Neurotensin/metabolism , Neurotensin/metabolism , Neurotensin/pharmacology , Ligands , Dopamine D2 Receptor Antagonists/metabolism , Dopamine D2 Receptor Antagonists/pharmacology
4.
Biochemistry ; 62(7): 1233-1248, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36917754

ABSTRACT

The NTSR1 neurotensin receptor (NTSR1) is a G protein-coupled receptor (GPCR) found in the brain and peripheral tissues with neurotensin (NTS) being its endogenous peptide ligand. In the brain, NTS modulates dopamine neuronal activity, induces opioid-independent analgesia, and regulates food intake. Recent studies indicate that biasing NTSR1 toward ß-arrestin signaling can attenuate the actions of psychostimulants and other drugs of abuse. Here, we provide the cryoEM structures of NTSR1 ternary complexes with heterotrimeric Gq and GoA with and without the brain-penetrant small-molecule SBI-553. In functional studies, we discovered that SBI-553 displays complex allosteric actions exemplified by negative allosteric modulation for G proteins that are Gα subunit selective and positive allosteric modulation and agonism for ß-arrestin translocation at NTSR1. Detailed structural analysis of the allosteric binding site illuminated the structural determinants for biased allosteric modulation of SBI-553 on NTSR1.


Subject(s)
Neurotensin , Receptors, Neurotensin , Receptors, Neurotensin/chemistry , Receptors, Neurotensin/metabolism , Neurotensin/metabolism , Signal Transduction , Peptides/metabolism , beta-Arrestins/metabolism
5.
NMR Biomed ; 36(2): e4842, 2023 02.
Article in English | MEDLINE | ID: mdl-36259728

ABSTRACT

The United States is experiencing a dramatic increase in maternal opioid misuse and, consequently, the number of individuals exposed to opioids in utero. Prenatal opioid exposure has both acute and long-lasting effects on health and wellbeing. Effects on the brain, often identified at school age, manifest as cognitive impairment, attention deficit, and reduced scholastic achievement. The neurobiological basis for these effects is poorly understood. Here, we examine how in utero exposure to heroin affects brain development into early adolescence in a mouse model. Pregnant C57BL/6J mice received escalating doses of heroin twice daily on gestational days 4-18. The brains of offspring were assessed on postnatal day 28 using 9.4 T diffusion MRI of postmortem specimens at 36 µm resolution. Whole-brain volumes and the volumes of 166 bilateral regions were compared between heroin-exposed and control offspring. We identified a reduction in whole-brain volume in heroin-exposed offspring and heroin-associated volume changes in 29 regions after standardizing for whole-brain volume. Regions with bilaterally reduced standardized volumes in heroin-exposed offspring relative to controls include the ectorhinal and insular cortices. Regions with bilaterally increased standardized volumes in heroin-exposed offspring relative to controls include the periaqueductal gray, septal region, striatum, and hypothalamus. Leveraging microscopic resolution diffusion tensor imaging and precise regional parcellation, we generated whole-brain structural MRI diffusion connectomes. Using a dimension reduction approach with multivariate analysis of variance to assess group differences in the connectome, we found that in utero heroin exposure altered structure-based connectivity of the left septal region and the region that acts as a hub for limbic regulatory actions. Consistent with clinical evidence, our findings suggest that prenatal opioid exposure may have effects on brain morphology, connectivity, and, consequently, function that persist into adolescence. This work expands our understanding of the risks associated with opioid misuse during pregnancy and identifies biomarkers that may facilitate diagnosis and treatment.


Subject(s)
Opioid-Related Disorders , Prenatal Exposure Delayed Effects , Humans , Pregnancy , Female , Animals , Mice , Heroin/adverse effects , Diffusion Tensor Imaging/methods , Analgesics, Opioid/pharmacology , Mice, Inbred C57BL , Brain
6.
Sci Rep ; 12(1): 21422, 2022 Dec 11.
Article in English | MEDLINE | ID: mdl-36503898

ABSTRACT

Genetically tractable animal models provide needed strategies to resolve the biological basis of drug addiction. Intravenous self-administration (IVSA) is the gold standard for modeling psychostimulant and opioid addiction in animals, but technical limitations have precluded the widespread use of IVSA in mice. Here, we describe IVSA paradigms for mice that capture the multi-stage nature of the disorder and permit predictive modeling. In these paradigms, C57BL/6J mice with long-standing indwelling jugular catheters engaged in cocaine- or remifentanil-associated lever responding that was fixed ratio-dependent, dose-dependent, extinguished by withholding the drug, and reinstated by the presentation of drug-paired cues. The application of multivariate analysis suggested that drug taking in both paradigms was a function of two latent variables we termed incentive motivation and discriminative control. Machine learning revealed that vulnerability to drug seeking and relapse were predicted by a mouse's a priori response to novelty, sensitivity to drug-induced locomotion, and drug-taking behavior. The application of these behavioral and statistical-analysis approaches to genetically-engineered mice will facilitate the identification of neural circuits driving addiction susceptibility and relapse and focused therapeutic development.


Subject(s)
Drug-Seeking Behavior , Mice , Animals , Mice, Inbred C57BL , Administration, Intravenous , Self Administration , Models, Animal
7.
Trends Pharmacol Sci ; 42(4): 283-299, 2021 04.
Article in English | MEDLINE | ID: mdl-33581873

ABSTRACT

G protein-coupled receptors (GPCRs) are the largest class of cell surface receptors in the genome and the most successful family of targets of FDA-approved drugs. New frontiers in GPCR drug discovery remain, however, as achieving receptor subtype selectivity and controlling off- and on-target side effects are not always possible with classic agonist and antagonist ligands. These challenges may be overcome by focusing development efforts on allosteric ligands that confer signaling bias. Biased allosteric modulators (BAMs) are an emerging class of GPCR ligands that engage less well-conserved regulatory motifs outside the orthosteric pocket and exert pathway-specific effects on receptor signaling. The unique ways that BAMs texturize receptor signaling present opportunities to fine-tune physiology and develop safer, more selective therapeutics. Here, we provide a conceptual framework for understanding the pharmacology of BAMs, explore their therapeutic potential, and discuss strategies for their discovery.


Subject(s)
Drug Discovery , Receptors, G-Protein-Coupled , Allosteric Regulation , Ligands , Signal Transduction
8.
Cell ; 181(6): 1364-1379.e14, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32470395

ABSTRACT

Small molecule neurotensin receptor 1 (NTSR1) agonists have been pursued for more than 40 years as potential therapeutics for psychiatric disorders, including drug addiction. Clinical development of NTSR1 agonists has, however, been precluded by their severe side effects. NTSR1, a G protein-coupled receptor (GPCR), signals through the canonical activation of G proteins and engages ß-arrestins to mediate distinct cellular signaling events. Here, we characterize the allosteric NTSR1 modulator SBI-553. This small molecule not only acts as a ß-arrestin-biased agonist but also extends profound ß-arrestin bias to the endogenous ligand by selectively antagonizing G protein signaling. SBI-553 shows efficacy in animal models of psychostimulant abuse, including cocaine self-administration, without the side effects characteristic of balanced NTSR1 agonism. These findings indicate that NTSR1 G protein and ß-arrestin activation produce discrete and separable physiological effects, thus providing a strategy to develop safer GPCR-targeting therapeutics with more directed pharmacological action.


Subject(s)
Behavior, Addictive/metabolism , Receptors, Neurotensin/metabolism , beta-Arrestins/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Behavior, Addictive/drug therapy , Cell Line , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Models, Animal , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Small Molecule Libraries/pharmacology
9.
J Med Chem ; 62(17): 8357-8363, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31390201

ABSTRACT

Neurotensin receptor 1 (NTR1) is a G protein coupled receptor that is widely expressed throughout the central nervous system where it acts as a neuromodulator. Neurotensin receptors have been implicated in a wide variety of CNS disorders, but despite extensive efforts to develop small molecule ligands there are few reports of such compounds. Herein we describe the optimization of a quinazoline based lead to give 18 (SBI-553), a potent and brain penetrant NTR1 allosteric modulator.


Subject(s)
Central Nervous System Diseases/drug therapy , Drug Discovery , Quinazolines/pharmacology , Receptors, Neurotensin/antagonists & inhibitors , beta-Arrestins/pharmacology , Administration, Oral , Allosteric Regulation/drug effects , Animals , Biological Availability , Central Nervous System Diseases/metabolism , Dopamine Plasma Membrane Transport Proteins/deficiency , Dopamine Plasma Membrane Transport Proteins/metabolism , Dose-Response Relationship, Drug , Female , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Quinazolines/administration & dosage , Quinazolines/chemistry , Rats , Receptors, Neurotensin/metabolism , Structure-Activity Relationship , beta-Arrestins/administration & dosage , beta-Arrestins/chemistry
10.
ACS Pharmacol Transl Sci ; 2(4): 230-246, 2019 Aug 09.
Article in English | MEDLINE | ID: mdl-32259059

ABSTRACT

G-protein-coupled receptors (GPCRs) can bias signaling through distinct biochemical pathways that originate from G-protein/receptor and ß-arrestin/receptor complexes. Receptor conformations supporting ß-arrestin engagement depend on multiple receptor determinants. Using ghrelin receptor GHR1a, we demonstrate by bioluminescence resonance energy transfer and fluorescence microscopy a critical role for its second intracellular loop 2 (ICL2) domain in stabilizing ß-arrestin/GHSR1a core interactions and determining receptor trafficking fate. We validate our findings in ICL2 gain- and loss-of-function experiments assessing ß-arrestin and ubiquitin-dependent internalization of the CC chemokine receptor, CCR1. Like all CC and CXC subfamily chemokine receptors, CCR1 lacks a critical proline residue found in the ICL2 consensus domain of rhodopsin-family GPCRs. Our study indicates that ICL2, C-tail determinants, and the orthosteric binding pocket that regulates ß-arrestin/receptor complex stability are sufficient to encode a broad repertoire of the trafficking fates observed for rhodopsin-family GPCRs, suggesting they provide the essential elements for regulating a large fraction of ß-arrestin signaling bias.

11.
Synapse ; 72(1)2018 01.
Article in English | MEDLINE | ID: mdl-28941296

ABSTRACT

The "brain-gut" peptide ghrelin, which mediates food-seeking behaviors, is recognized as a very strong endogenous modulator of dopamine (DA) signaling. Ghrelin binds the G protein-coupled receptor GHSR1a, and administration of ghrelin increases the rewarding properties of psychostimulants while ghrelin receptor antagonists decrease them. In addition, the GHSR1a signals through ßarrestin-2 to regulate actin/stress fiber rearrangement, suggesting ßarrestin-2 participation in the regulation of actin-mediated synaptic plasticity for addictive substances like cocaine. The effects of ghrelin receptor ligands on reward strongly suggest that modulation of ghrelin signaling could provide an effective strategy to ameliorate undesirable behaviors arising from addiction. To investigate this possibility, we tested the effects of ghrelin receptor antagonism in a cocaine behavioral sensitization paradigm using DA neuron-specific ßarrestin-2 KO mice. Our results show that these mice sensitize to cocaine as well as wild-type littermates. The ßarrestin-2 KO mice, however, no longer respond to the locomotor attenuating effects of the GHSR1a antagonist YIL781. The data presented here suggest that the separate stages of addictive behavior differ in their requirements for ßarrestin-2 and show that pharmacological inhibition of ßarrestin-2 function through GHSR1a antagonism is not equivalent to the loss of ßarrestin-2 function achieved by genetic ablation. These data support targeting GHSR1a signaling in addiction therapy but indicate that using signaling biased compounds that modulate ßarrestin-2 activity differentially from G protein activity may be required.


Subject(s)
Cocaine-Related Disorders/metabolism , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Motor Activity/drug effects , Receptors, Ghrelin/antagonists & inhibitors , beta-Arrestin 2/metabolism , Animals , Cell Line, Tumor , Central Nervous System Agents , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Female , Ghrelin/metabolism , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Piperidines/pharmacology , Quinazolinones/pharmacology , Receptors, Ghrelin/metabolism , beta-Arrestin 2/genetics
12.
Pain ; 158(9): 1733-1742, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28570482

ABSTRACT

Metastatic bone pain is the single most common form of cancer pain and persists as a result of peripheral and central inflammatory, as well as neuropathic mechanisms. Here, we provide the first characterization of sphingolipid metabolism alterations in the spinal cord occurring during cancer-induced bone pain (CIBP). Following femoral arthrotomy and syngenic tumor implantation in mice, ceramides decreased with corresponding increases in sphingosine and the bioactive sphingolipid metabolite, sphingosine 1-phosphate (S1P). Intriguingly, de novo sphingolipid biosynthesis was increased as shown by the elevations of dihydro-ceramides and dihydro-S1P. We next identified the S1P receptor subtype 1 (S1PR1) as a novel target for therapeutic intervention. Intrathecal or systemic administration of the competitive and functional S1PR1 antagonists, TASP0277308 and FTY720/Fingolimod, respectively, attenuated cancer-induced spontaneous flinching and guarding. Inhibiting CIBP by systemic delivery of FTY720 did not result in antinociceptive tolerance over 7 days. FTY720 administration enhanced IL-10 in the lumbar ipsilateral spinal cord of CIBP animals and intrathecal injection of an IL-10 neutralizing antibody mitigated the ability of systemic FTY720 to reverse CIBP. FTY720 treatment was not associated with alterations in bone metabolism in vivo. Studies here identify a novel mechanism to inhibit bone cancer pain by blocking the actions of the bioactive metabolites S1P and dihydro-S1P in lumbar spinal cord induced by bone cancer and support potential fast-track clinical application of the FDA-approved drug, FTY720, as a therapeutic avenue for CIBP.


Subject(s)
Cancer Pain/etiology , Cancer Pain/metabolism , Neurogenic Inflammation/etiology , Neurogenic Inflammation/metabolism , Proprotein Convertases/metabolism , Receptors, Lysosphingolipid/metabolism , Serine Endopeptidases/metabolism , Animals , Breast Neoplasms/complications , Breast Neoplasms/pathology , Cancer Pain/drug therapy , Cell Line, Tumor , Disease Models, Animal , Female , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Neurogenic Inflammation/drug therapy , Proprotein Convertases/antagonists & inhibitors , Receptors, Lysosphingolipid/antagonists & inhibitors , Signal Transduction/drug effects , Sphingosine-1-Phosphate Receptors , Spinal Cord/drug effects , Spinal Cord/metabolism , Sulfones/pharmacology , Sulfones/therapeutic use , Triazoles/pharmacology , Triazoles/therapeutic use
13.
Pain ; 157(11): 2605-2616, 2016 11.
Article in English | MEDLINE | ID: mdl-27482630

ABSTRACT

Bone is one of the leading sites of metastasis for frequently diagnosed malignancies, including those arising in the breast, prostate and lung. Although these cancers develop unnoticed and are painless in their primary sites, bone metastases result in debilitating pain. Deeper investigation of this pain may reveal etiology and lead to early cancer detection. Cancer-induced bone pain (CIBP) is inadequately managed with current standard-of-care analgesics and dramatically diminishes patient quality of life. While CIBP etiology is multifaceted, elevated levels of glutamate, an excitatory neurotransmitter, in the bone-tumor microenvironment may drive maladaptive nociceptive signaling. Here, we establish a relationship between the reactive nitrogen species peroxynitrite, tumor-derived glutamate, and CIBP. In vitro and in a syngeneic in vivo model of breast CIBP, murine mammary adenocarcinoma cells significantly elevated glutamate via the cystine/glutamate antiporter system xc. The well-known system xc inhibitor sulfasalazine significantly reduced levels of glutamate and attenuated CIBP-associated flinching and guarding behaviors. Peroxynitrite, a highly reactive species produced in tumors, significantly increased system xc functional expression and tumor cell glutamate release. Scavenging peroxynitrite with the iron and mangano-based porphyrins, FeTMPyP and SRI10, significantly diminished tumor cell system xc functional expression, reduced femur glutamate levels and mitigated CIBP. In sum, we demonstrate how breast cancer bone metastases upregulate a cystine/glutamate co-transporter to elevate extracellular glutamate. Pharmacological manipulation of peroxynitrite or system xc attenuates CIBP, supporting a role for tumor-derived glutamate in CIBP and validating the targeting of system xc as a novel therapeutic strategy for the management of metastatic bone pain.


Subject(s)
Adenocarcinoma/complications , Bone Neoplasms/complications , Breast Neoplasms/metabolism , Cancer Pain/metabolism , Glutamic Acid/metabolism , Sulfasalazine/pharmacology , Adenocarcinoma/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antiporters/pharmacology , Bone Neoplasms/pathology , Breast Neoplasms/secondary , Calcium-Binding Proteins/metabolism , Cancer Pain/drug therapy , Cancer Pain/etiology , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Metalloporphyrins/pharmacology , Mice , Mice, Inbred BALB C , Peroxynitrous Acid/metabolism , Time Factors
14.
Pain ; 157(12): 2709-2721, 2016 12.
Article in English | MEDLINE | ID: mdl-27541850

ABSTRACT

Many cancerous solid tumors metastasize to the bone and induce pain (cancer-induced bone pain [CIBP]). Cancer-induced bone pain is often severe because of enhanced inflammation, rapid bone degradation, and disease progression. Opioids are prescribed to manage this pain, but they may enhance bone loss and increase tumor proliferation, further compromising patient quality of life. Angiotensin-(1-7) (Ang-(1-7)) binds and activates the Mas receptor (MasR). Angiotensin-(1-7)/MasR activation modulates inflammatory signaling after acute tissue insult, yet no studies have investigated whether Ang-(1-7)/MasR play a role in CIBP. We hypothesized that Ang-(1-7) inhibits CIBP by targeting MasR in a murine model of breast CIBP. 66.1 breast cancer cells were implanted into the femur of BALB/cAnNHsd mice as a model of CIBP. Spontaneous and evoked pain behaviors were assessed before and after acute and chronic administration of Ang-(1-7). Tissues were collected from animals for ex vivo analyses of MasR expression, tumor burden, and bone integrity. Cancer inoculation increased spontaneous pain behaviors by day 7 that were significantly reduced after a single injection of Ang-(1-7) and after sustained administration. Preadministration of A-779 a selective MasR antagonist prevented this reduction, whereas pretreatment with the AT2 antagonist had no effect; an AT1 antagonist enhanced the antinociceptive activity of Ang-(1-7) in CIBP. Repeated Ang-(1-7) administration did not significantly change tumor burden or bone remodeling. Data here suggest that Ang-(1-7)/MasR activation significantly attenuates CIBP, while lacking many side effects seen with opioids. Thus, Ang-(1-7) may be an alternative therapeutic strategy for the nearly 90% of patients with advanced-stage cancer who experience excruciating pain.


Subject(s)
Analgesics/therapeutic use , Angiotensin I/therapeutic use , Bone Neoplasms/complications , Cancer Pain/drug therapy , Cancer Pain/etiology , Peptide Fragments/therapeutic use , Analysis of Variance , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Angiotensin II Type 2 Receptor Blockers/therapeutic use , Animals , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Imidazoles/therapeutic use , Losartan/therapeutic use , Mice , Mice, Inbred BALB C , Nesting Behavior/drug effects , Pyridines/therapeutic use , Rotarod Performance Test
15.
Cancer Growth Metastasis ; 8(Suppl 1): 47-62, 2015.
Article in English | MEDLINE | ID: mdl-26339191

ABSTRACT

Many common cancers have a propensity to metastasize to bone. Although malignancies often go undetected in their native tissues, bone metastases produce excruciating pain that severely compromises patient quality of life. Cancer-induced bone pain (CIBP) is poorly managed with existing medications, and its multifaceted etiology remains to be fully elucidated. Novel analgesic targets arise as more is learned about this complex and distinct pain state. Over the past two decades, multiple animal models have been developed to study CIBP's unique pathology and identify therapeutic targets. Here, we review animal models of CIBP and the mechanistic insights gained as these models evolve. Findings from immunocompromised and immunocompetent host systems are discussed separately to highlight the effect of model choice on outcome. Gaining an understanding of the unique neuromolecular profile of cancer pain through the use of appropriate animal models will aid in the development of more effective therapeutics for CIBP.

16.
Expert Opin Ther Pat ; 25(4): 443-66, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25576197

ABSTRACT

INTRODUCTION: Peroxynitrite is a cytotoxic oxidant species implicated in a host of pathologies, including inflammatory and neurodegenerative diseases, cancer, radiation injury and chronic pain. With the recognition of the role of peroxynitrite in disease, numerous experimental and therapeutic tools have arisen to probe peroxyntirite's pathophysiological contribution and attenuate its oxidative damage. Peroxynitrite decomposition catalysts (PNDCs) are redox-active compounds that detoxify peroxynitrite by catalyzing its isomerization or reduction to nitrate or nitrite. AREAS COVERED: This review discusses recent research articles and patents published 1995 - 2014 on the development and therapeutic use of PNDCs. Iron and manganese metalloporphyrin PNDCs attenuate the toxic effects of peroxynitrite and are currently being developed for clinical applications. Additionally, some Mn porphyrin-based PNDCs have optimized pharmaceutical properties such that they exhibit greater peroxynitrite selectivity. Other classes of PNDC agents, including bis(hydroxyphenyl)dipyrromethenes and metallocorroles, have demonstrated preclinical efficacy, oral availability and reduced toxicity risk. EXPERT OPINION: Interest in the drug-like properties of peroxynitrite-neutralizing agents has grown with the realization that PNDCs will be powerful tools in the treatment of disease. The design of compounds with enhanced oral availability and peroxynitrite selectivity is a critical step toward the availability of safe, effective and selective redox modulators for the treatment of peroxynitrite-associated pathologies.


Subject(s)
Drug Design , Oxidants/metabolism , Peroxynitrous Acid/metabolism , Animals , Catalysis , Humans , Oxidation-Reduction/drug effects , Patents as Topic
17.
PLoS One ; 9(2): e88516, 2014.
Article in English | MEDLINE | ID: mdl-24520393

ABSTRACT

Our laboratory has previously demonstrated that peripheral inflammatory pain (PIP), induced by subcutaneous plantar injection of λ-carrageenan, results in increased expression and activity of the ATP-dependent efflux transporter P-glycoprotein (P-gp) that is endogenously expressed at the blood-brain barrier (BBB). The result of increased P-gp functional expression was a significant reduction in CNS uptake of morphine and, subsequently, reduced morphine analgesic efficacy. A major concern in the treatment of acute pain/inflammation is the potential for drug-drug interactions resulting from P-gp induction by therapeutic agents co-administered with opioids. Such effects on P-gp activity can profoundly modulate CNS distribution of opioid analgesics and alter analgesic efficacy. In this study, we examined the ability of diclofenac, a non-steroidal anti-inflammatory drug (NSAID) that is commonly administered in conjunction with the opioids during pain therapy, to alter BBB transport of morphine via P-gp and whether such changes in P-gp morphine transport could alter morphine analgesic efficacy. Administration of diclofenac reduced paw edema and thermal hyperalgesia in rats subjected to PIP, which is consistent with the known mechanism of action of this NSAID. Western blot analysis demonstrated an increase in P-gp expression in rat brain microvessels not only following PIP induction but also after diclofenac treatment alone. Additionally, in situ brain perfusion studies showed that both PIP and diclofenac treatment alone increased P-gp efflux activity resulting in decreased morphine brain uptake. Critically, morphine analgesia was significantly reduced in animals pretreated with diclofenac (3 h), as compared to animals administered diclofenac and morphine concurrently. These novel findings suggest that administration of diclofenac and P-gp substrate opioids during pain pharmacotherapy may result in a clinically significant drug-drug interaction.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Central Nervous System/metabolism , Diclofenac/pharmacology , Morphine/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biological Transport/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Carrageenan , Central Nervous System/drug effects , Central Nervous System/pathology , Diclofenac/administration & dosage , Diclofenac/therapeutic use , Edema/blood , Edema/complications , Edema/drug therapy , Edema/pathology , Female , Fluorescent Antibody Technique , Hyperalgesia/blood , Hyperalgesia/complications , Hyperalgesia/drug therapy , Hyperalgesia/pathology , Microvessels/drug effects , Microvessels/metabolism , Microvessels/pathology , Nociception/drug effects , Pain/blood , Pain/complications , Pain/drug therapy , Pain/pathology , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/blood
18.
J Cereb Blood Flow Metab ; 34(4): 699-707, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24473481

ABSTRACT

Cerebral hypoxia and subsequent reoxygenation stress (H/R) is a component of several diseases. One approach that may enable neural tissue rescue after H/R is central nervous system (CNS) delivery of drugs with brain protective effects such as 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors (i.e., statins). Our present in vivo data show that atorvastatin, a commonly prescribed statin, attenuates poly (ADP-ribose) polymerase (PARP) cleavage in the brain after H/R, suggesting neuroprotective efficacy. However, atorvastatin use as a CNS therapeutic is limited by poor blood-brain barrier (BBB) penetration. Therefore, we examined regulation and functional expression of the known statin transporter organic anion transporting polypeptide 1a4 (Oatp1a4) at the BBB under H/R conditions. In rat brain microvessels, H/R (6% O2, 60 minutes followed by 21% O2, 10 minutes) increased Oatp1a4 expression. Brain uptake of taurocholate (i.e., Oap1a4 probe substrate) and atorvastatin were reduced by Oatp inhibitors (i.e., estrone-3-sulfate and fexofenadine), suggesting involvement of Oatp1a4 in brain drug delivery. Pharmacological inhibition of transforming growth factor-ß (TGF-ß)/activin receptor-like kinase 5 (ALK5) signaling with the selective inhibitor SB431542 increased Oatp1a4 functional expression, suggesting a role for TGF-ß/ALK5 signaling in Oatp1a4 regulation. Taken together, our novel data show that targeting an endogenous BBB drug uptake transporter (i.e., Oatp1a4) may be a viable approach for optimizing CNS drug delivery for treatment of diseases with an H/R component.


Subject(s)
Blood-Brain Barrier/metabolism , Drug Delivery Systems , Hypoxia, Brain/metabolism , Organic Anion Transporters/metabolism , Reperfusion Injury/metabolism , Animals , Atorvastatin , Biological Transport , Blood Gas Analysis , Blood-Brain Barrier/drug effects , Blotting, Western , Carbon Dioxide/blood , Electrolytes/blood , Female , Heptanoic Acids/administration & dosage , Heptanoic Acids/pharmacokinetics , Heptanoic Acids/therapeutic use , Hypoxia, Brain/complications , Hypoxia, Brain/drug therapy , Microvessels/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/therapeutic use , Oxygen/blood , Pyrroles/administration & dosage , Pyrroles/pharmacokinetics , Pyrroles/therapeutic use , Rats , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Reperfusion Injury/etiology
19.
Curr Pharm Des ; 20(10): 1422-49, 2014.
Article in English | MEDLINE | ID: mdl-23789948

ABSTRACT

The blood-brain barrier (BBB) and blood-cerebrospinal fluid (BCSF) barriers are critical determinants of CNS homeostasis. Additionally, the BBB and BCSF barriers are formidable obstacles to effective CNS drug delivery. These brain barrier sites express putative influx and efflux transporters that precisely control permeation of circulating solutes including drugs. The study of transporters has enabled a shift away from "brute force" approaches to delivering drugs by physically circumventing brain barriers towards chemical approaches that can target specific compounds of the BBB and/or BCSF barrier. However, our understanding of transporters at the BBB and BCSF barriers has primarily focused on understanding efflux transporters that efficiently prevent drugs from attaining therapeutic concentrations in the CNS. Recently, through the characterization of multiple endogenously expressed uptake transporters, this paradigm has shifted to the study of brain transporter targets that can facilitate drug delivery (i.e., influx transporters). Additionally, signaling pathways and trafficking mechanisms have been identified for several endogenous BBB/BCSF transporters, thereby offering even more opportunities to understand how transporters can be exploited for optimization of CNS drug delivery. This review presents an overview of the BBB and BCSF barrier as well as the many families of transporters functionally expressed at these barrier sites. Furthermore, we present an overview of various strategies that have been designed and utilized to deliver therapeutic agents to the brain with a particular emphasis on those approaches that directly target endogenous BBB/BCSF barrier transporters.


Subject(s)
Blood-Brain Barrier/drug effects , Central Nervous System/drug effects , Drug Delivery Systems , Membrane Transport Proteins/chemistry , Pharmaceutical Preparations/administration & dosage , Animals , Blood-Brain Barrier/metabolism , Central Nervous System/metabolism , Humans , Membrane Transport Proteins/metabolism
20.
Mol Pharmacol ; 84(5): 774-86, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24019224

ABSTRACT

Effective pharmacologic treatment of pain with opioids requires that these drugs attain efficacious concentrations in the central nervous system (CNS). A primary determinant of CNS drug permeation is P-glycoprotein (P-gp), an endogenous blood-brain barrier (BBB) efflux transporter that is involved in brain-to-blood transport of opioid analgesics (i.e., morphine). Recently, the nuclear receptor constitutive androstane receptor (CAR) has been identified as a regulator of P-gp functional expression at the BBB. This is critical to pharmacotherapy of pain/inflammation, as patients are often administered acetaminophen (APAP), a CAR-activating ligand, in conjunction with an opioid. Our objective was to investigate, in vivo, the role of CAR in regulation of P-gp at the BBB. Following APAP treatment, P-gp protein expression was increased up to 1.4-1.6-fold in a concentration-dependent manner. Additionally, APAP increased P-gp transport of BODIPY-verapamil in freshly isolated rat brain capillaries. This APAP-induced increase in P-gp expression and activity was attenuated in the presence of CAR pathway inhibitor okadaic acid or transcriptional inhibitor actinomycin D, suggesting P-gp regulation is CAR-dependent. Furthermore, morphine brain accumulation was enhanced by P-gp inhibitors in APAP-treated animals, suggesting P-gp-mediated transport. A warm-water (50°C) tail-flick assay revealed a significant decrease in morphine analgesia in animals treated with morphine 3 or 6 hours after APAP treatment, as compared with animals treated concurrently. Taken together, our data imply that inclusion of APAP in a pain treatment regimen activates CAR at the BBB and increases P-gp functional expression, a clinically significant drug-drug interaction that modulates opioid analgesic efficacy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Blood-Brain Barrier/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Biological Transport/drug effects , Brain/blood supply , Constitutive Androstane Receptor , Female , Morphine/pharmacokinetics , Permeability , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...