Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Br J Pharmacol ; 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715413

ABSTRACT

BACKGROUND AND PURPOSE: The voltage-gated sodium channel isoform NaV1.7 is a high-interest target for the development of non-opioid analgesics due to its preferential expression in pain-sensing neurons. NaV1.7 is also expressed in autonomic neurons, yet its contribution to involuntary visceral reflexes has received limited attention. The small molecule inhibitor ST-2560 was advanced into pain behaviour and cardiovascular models to understand the pharmacodynamic effects of selective inhibition of NaV1.7. EXPERIMENTAL APPROACH: Potency of ST-2560 at NaV1.7 and off-target ion channels was evaluated by whole-cell patch-clamp electrophysiology. Effects on nocifensive reflexes were assessed in non-human primate (NHP) behavioural models, employing the chemical capsaicin and mechanical stimuli. Cardiovascular parameters were monitored continuously in freely-moving, telemetered NHPs following administration of vehicle and ST-2560. KEY RESULTS: ST-2560 is a potent inhibitor (IC50 = 39 nM) of NaV1.7 in primates with ≥1000-fold selectivity over other isoforms of the human NaV1.x family. Following systemic administration, ST-2560 (0.1-0.3 mg·kg-1, s.c.) suppressed noxious mechanical- and chemical-evoked reflexes at free plasma concentrations threefold to fivefold above NaV1.7 IC50. ST-2560 (0.1-1.0 mg·kg-1, s.c.) also produced changes in haemodynamic parameters, most notably a 10- to 20-mmHg reduction in systolic and diastolic arterial blood pressure, at similar exposures. CONCLUSIONS AND IMPLICATIONS: Acute pharmacological inhibition of NaV1.7 is antinociceptive, but also has the potential to impact the cardiovascular system. Further work is merited to understand the role of NaV1.7 in autonomic ganglia involved in the control of heart rate and blood pressure, and the effect of selective NaV1.7 inhibition on cardiovascular function.

2.
ACS Med Chem Lett ; 13(11): 1763-1768, 2022 Nov 10.
Article in English | MEDLINE | ID: mdl-36385936

ABSTRACT

The voltage-gated sodium channel isoform NaV1.7 has drawn widespread interest as a target for non-opioid, investigational new drugs to treat pain. Selectivity over homologous, off-target sodium channel isoforms, which are expressed in peripheral motor neurons, the central nervous system, skeletal muscle and the heart, poses a significant challenge to the development of small molecule inhibitors of NaV1.7. Most inhibitors of NaV1.7 disclosed to date belong to a class of aryl and acyl sulfonamides that preferentially bind to an inactivated conformation of the channel. By taking advantage of a sequence variation unique to primate NaV1.7 in the extracellular pore of the channel, a series of bis-guanidinium analogues of the natural product, saxitoxin, has been identified that are potent against the resting conformation of the channel. A compound of interest, 25, exhibits >600-fold selectivity over off-target sodium channel isoforms and is efficacious in a preclinical model of acute pain.

3.
Pain ; 162(4): 1250-1261, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33086288

ABSTRACT

ABSTRACT: The voltage-gated sodium channel Nav1.7 is highly expressed in nociceptive afferents and is critically involved in pain signal transmission. Nav1.7 is a genetically validated pain target in humans because loss-of-function mutations cause congenital insensitivity to pain and gain-of-function mutations cause severe pain syndromes. Consequently, pharmacological inhibition has been investigated as an analgesic therapeutic strategy. We describe a small molecule Nav1.7 inhibitor, ST-2530, that is an analog of the naturally occurring sodium channel blocker saxitoxin. When evaluated against human Nav1.7 by patch-clamp electrophysiology using a protocol that favors the resting state, the Kd of ST-2530 was 25 ± 7 nM. ST-2530 exhibited greater than 500-fold selectivity over human voltage-gated sodium channel isoforms Nav1.1-Nav1.6 and Nav1.8. Although ST-2530 had lower affinity against mouse Nav1.7 (Kd = 250 ± 40 nM), potency was sufficient to assess analgesic efficacy in mouse pain models. A 3-mg/kg dose administered subcutaneously was broadly analgesic in acute pain models using noxious thermal, mechanical, and chemical stimuli. ST-2530 also reversed thermal hypersensitivity after a surgical incision on the plantar surface of the hind paw. In the spared nerve injury model of neuropathic pain, ST-2530 transiently reversed mechanical allodynia. These analgesic effects were demonstrated at doses that did not affect locomotion, motor coordination, or olfaction. Collectively, results from this study indicate that pharmacological inhibition of Nav1.7 by a small molecule agent with affinity for the resting state of the channel is sufficient to produce analgesia in a range of preclinical pain models.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel , Saxitoxin , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Mice , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/genetics , Protein Isoforms , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use
4.
J Med Chem ; 62(19): 8695-8710, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31012583

ABSTRACT

Voltage-gated sodium ion channel subtype 1.7 (NaV1.7) is a high interest target for the discovery of non-opioid analgesics. Compelling evidence from human genetic data, particularly the finding that persons lacking functional NaV1.7 are insensitive to pain, has spurred considerable effort to develop selective inhibitors of this Na+ ion channel target as analgesic medicines. Recent clinical setbacks and disappointing performance of preclinical compounds in animal pain models, however, have led to skepticism around the potential of selective NaV1.7 inhibitors as human therapeutics. In this Perspective, we discuss the attributes and limitations of recently disclosed investigational drugs targeting NaV1.7 and review evidence that, by better understanding the requirements for selectivity and target engagement, the opportunity to deliver effective analgesic medicines targeting NaV1.7 endures.


Subject(s)
Analgesics/chemistry , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Sodium Channel Blockers/chemistry , Analgesics/metabolism , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Disease Models, Animal , Humans , NAV1.7 Voltage-Gated Sodium Channel/chemistry , Pain/drug therapy , Pain/pathology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Signal Transduction/drug effects , Sodium Channel Blockers/metabolism , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use , Sulfonamides/chemistry , Sulfonamides/metabolism
5.
Cell ; 172(4): 857-868.e15, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29336889

ABSTRACT

The mechanism by which the wild-type KRAS allele imparts a growth inhibitory effect to oncogenic KRAS in various cancers, including lung adenocarcinoma (LUAD), is poorly understood. Here, using a genetically inducible model of KRAS loss of heterozygosity (LOH), we show that KRAS dimerization mediates wild-type KRAS-dependent fitness of human and murine KRAS mutant LUAD tumor cells and underlies resistance to MEK inhibition. These effects are abrogated when wild-type KRAS is replaced by KRASD154Q, a mutant that disrupts dimerization at the α4-α5 KRAS dimer interface without changing other fundamental biochemical properties of KRAS, both in vitro and in vivo. Moreover, dimerization has a critical role in the oncogenic activity of mutant KRAS. Our studies provide mechanistic and biological insights into the role of KRAS dimerization and highlight a role for disruption of dimerization as a therapeutic strategy for KRAS mutant cancers.


Subject(s)
Adenocarcinoma of Lung , Enzyme Inhibitors/pharmacology , Lung Neoplasms , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mutation, Missense , Protein Multimerization/drug effects , Proto-Oncogene Proteins p21(ras)/metabolism , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/enzymology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Amino Acid Substitution , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Loss of Heterozygosity , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Knockout , Protein Multimerization/genetics , Proto-Oncogene Proteins p21(ras)/genetics
6.
Bioorg Med Chem ; 25(3): 838-846, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28011204

ABSTRACT

TAK1 (transforming growth factor-ß-activated kinase 1) is an essential intracellular mediator of cytokine and growth factor signaling and a potential therapeutic target for the treatment of immune diseases and cancer. Herein we report development of a series of 2,4-disubstituted pyrimidine covalent TAK1 inhibitors that target Cys174, a residue immediately adjacent to the 'DFG-motif' of the kinase activation loop. Co-crystal structures of TAK1 with candidate compounds enabled iterative rounds of structure-based design and biological testing to arrive at optimized compounds. Lead compounds such as 2 and 10 showed greater than 10-fold biochemical selectivity for TAK1 over the closely related kinases MEK1 and ERK1 which possess an equivalently positioned cysteine residue. These compounds are smaller, more easily synthesized, and exhibit a different spectrum of kinase selectivity relative to previously reported macrocyclic natural product TAK1 inhibitors such as 5Z-7-oxozeanol.


Subject(s)
MAP Kinase Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Animals , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , MAP Kinase Kinase Kinases/metabolism , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship
7.
Br J Radiol ; 89(1063): 20160138, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27109738

ABSTRACT

OBJECTIVE: Rheumatoid arthritis (RA) and psoriatic arthritis (PsA) commonly affect the small joints of the wrist and hand. We evaluated the performance of a new, high-resolution extremity positron emission tomography (PET)/CT scanner for characterizing and quantifying pathologies associated with the two arthritides in the wrist and hand joints. METHODS: Patients with RA or PsA underwent fluorine-18 fludeoxyglucose ((18)F-FDG) PET/CT wrist and hand imaging, respectively, on the high-resolution scanner. Calibrated CT images and co-registered PET images were reconstructed. PET/CT was derived for the radiocarpal and pisiform-triquetral compartments, joints with erosive changes, sites of synovitis or tenosynovitis and the nail bed and were correlated with clinical and MRI findings. RESULTS: Significantly elevated (18)F-FDG uptake was measured for the radiocarpal and pisiform-triquetral compartments and at sites of bone erosion, synovitis, pannus and oedema, compared with unaffected joints (p < 0.05) in patients with RA, consistent with their clinical findings. In patients with PsA, significantly elevated (18)F-FDG uptake was measured for joints with synovitis compared with unaffected joints (p < 0.05), with patterns of (18)F-FDG uptake along the tendons, at the enthesis and in the nail bed, consistent with tenosynovitis, enthesitis and nail dystrophy, respectively. CONCLUSION: High-resolution (18)F-FDG PET/CT imaging of the wrist and hand is feasible in an RA or PsA patient cohort and is capable of providing quantifiable measures of disease activity (synovitis, enthesitis, oedema and bone destruction). ADVANCES IN KNOWLEDGE: High-resolution PET/CT imaging shows promise as a tool for understanding the pathogenesis of the arthritic process and for non-invasive, objective assessment of RA or PsA severity and therapy selection.


Subject(s)
Arthritis, Psoriatic/diagnostic imaging , Arthritis, Rheumatoid/diagnostic imaging , Fluorodeoxyglucose F18 , Hand Joints/diagnostic imaging , Positron Emission Tomography Computed Tomography/methods , Radiopharmaceuticals , Disease Progression , Female , Humans , Male , Middle Aged , Pilot Projects , Prospective Studies , Reproducibility of Results , Wrist Joint/diagnostic imaging
8.
Mol Cancer Res ; 13(9): 1325-35, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26037647

ABSTRACT

UNLABELLED: KRAS mutations are the most common genetic abnormalities in cancer, but the distribution of specific mutations across cancers and the differential responses of patients with specific KRAS mutations in therapeutic clinical trials suggest that different KRAS mutations have unique biochemical behaviors. To further explain these high-level clinical differences and to explore potential therapeutic strategies for specific KRAS isoforms, we characterized the most common KRAS mutants biochemically for substrate binding kinetics, intrinsic and GTPase-activating protein (GAP)-stimulated GTPase activities, and interactions with the RAS effector, RAF kinase. Of note, KRAS G13D shows rapid nucleotide exchange kinetics compared with other mutants analyzed. This property can be explained by changes in the electrostatic charge distribution of the active site induced by the G13D mutation as shown by X-ray crystallography. High-resolution X-ray structures are also provided for the GDP-bound forms of KRAS G12V, G12R, and Q61L and reveal additional insight. Overall, the structural data and measurements, obtained herein, indicate that measurable biochemical properties provide clues for identifying KRAS-driven tumors that preferentially signal through RAF. IMPLICATIONS: Biochemical profiling and subclassification of KRAS-driven cancers will enable the rational selection of therapies targeting specific KRAS isoforms or specific RAS effectors.


Subject(s)
Mutation , Neoplasms/genetics , Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Crystallography, X-Ray , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/metabolism , Guanosine Diphosphate/chemistry , Humans , Proto-Oncogene Proteins p21(ras)/metabolism , raf Kinases/metabolism
9.
Nat Commun ; 5: 4387, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-25034454

ABSTRACT

Injury to lung epithelial cells has a role in multiple lung diseases. We previously identified mitsugumin 53 (MG53) as a component of the cell membrane repair machinery in striated muscle cells. Here we show that MG53 also has a physiological role in the lung and may be used as a treatment in animal models of acute lung injury. Mice lacking MG53 show increased susceptibility to ischaemia-reperfusion and overventilation-induced injury to the lung when compared with wild-type mice. Extracellular application of recombinant human MG53 (rhMG53) protein protects cultured lung epithelial cells against anoxia/reoxygenation-induced injuries. Intravenous delivery or inhalation of rhMG53 reduces symptoms in rodent models of acute lung injury and emphysema. Repetitive administration of rhMG53 improves pulmonary structure associated with chronic lung injury in mice. Our data indicate a physiological function for MG53 in the lung and suggest that targeting membrane repair may be an effective means for treatment or prevention of lung diseases.


Subject(s)
Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Cell Membrane/drug effects , Molecular Targeted Therapy/methods , Acute Lung Injury/genetics , Animals , Carrier Proteins/administration & dosage , Carrier Proteins/genetics , Cell Hypoxia/drug effects , Cell Membrane/metabolism , Cells, Cultured , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Humans , Lung/pathology , Male , Membrane Proteins , Mice, Knockout , Rats , Rats, Sprague-Dawley , Rats, Wistar , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Respiration, Artificial/adverse effects , Tripartite Motif Proteins
10.
Proc Natl Acad Sci U S A ; 111(24): 8895-900, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24889603

ABSTRACT

Directly targeting oncogenic V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (K-Ras) with small-molecule inhibitors has historically been considered prohibitively challenging. Recent reports of compounds that bind directly to the K-Ras G12C mutant suggest avenues to overcome key obstacles that stand in the way of developing such compounds. We aim to target the guanine nucleotide (GN)-binding pocket because the natural contents of this pocket dictate the signaling state of K-Ras. Here, we characterize the irreversible inhibitor SML-8-73-1 (SML), which targets the GN-binding pocket of K-Ras G12C. We report a high-resolution X-ray crystal structure of G12C K-Ras bound to SML, revealing that the compound binds in a manner similar to GDP, forming a covalent linkage with Cys-12. The resulting conformation renders K-Ras in the open, inactive conformation, which is not predicted to associate productively with or activate downstream effectors. Conservation analysis of the Ras family GN-binding pocket reveals variability in the side chains surrounding the active site and adjacent regions, especially in the switch I region. This variability may enable building specificity into new iterations of Ras and other GTPase inhibitors. High-resolution in situ chemical proteomic profiling of SML confirms that SML effectively discriminates between K-Ras G12C and other cellular GTP-binding proteins. A biochemical assay provides additional evidence that SML is able to compete with millimolar concentrations of GTP and GDP for the GN-binding site.


Subject(s)
Acetamides/chemistry , Genes, ras , Guanosine Diphosphate/analogs & derivatives , ras Proteins/antagonists & inhibitors , ras Proteins/chemistry , Binding Sites , Biotin/chemistry , Catalytic Domain , Conserved Sequence , Crystallography, X-Ray , GTP Phosphohydrolases/chemistry , GTP-Binding Proteins/chemistry , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , Humans , Ligands , Models, Molecular , Mutation , Phosphatidylinositol 3-Kinases/chemistry , Protein Binding , Protein Conformation , Proteomics , Signal Transduction
11.
J Pharmacol Exp Ther ; 349(1): 75-84, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24492651

ABSTRACT

Nasal congestion is one of the most troublesome symptoms of many upper airways diseases. We characterized the effect of selective α2c-adrenergic agonists in animal models of nasal congestion. In porcine mucosa tissue, compound A and compound B contracted nasal veins with only modest effects on arteries. In in vivo experiments, we examined the nasal decongestant dose-response characteristics, pharmacokinetic/pharmacodynamic relationship, duration of action, potential development of tolerance, and topical efficacy of α2c-adrenergic agonists. Acoustic rhinometry was used to determine nasal cavity dimensions following intranasal compound 48/80 (1%, 75 µl). In feline experiments, compound 48/80 decreased nasal cavity volume and minimum cross-sectional areas by 77% and 40%, respectively. Oral administration of compound A (0.1-3.0 mg/kg), compound B (0.3-5.0 mg/kg), and d-pseudoephedrine (0.3 and 1.0 mg/kg) produced dose-dependent decongestion. Unlike d-pseudoephedrine, compounds A and B did not alter systolic blood pressure. The plasma exposure of compound A to produce a robust decongestion (EC(80)) was 500 nM, which related well to the duration of action of approximately 4.0 hours. No tolerance to the decongestant effect of compound A (1.0 mg/kg p.o.) was observed. To study the topical efficacies of compounds A and B, the drugs were given topically 30 minutes after compound 48/80 (a therapeutic paradigm) where both agents reversed nasal congestion. Finally, nasal-decongestive activity was confirmed in the dog. We demonstrate that α2c-adrenergic agonists behave as nasal decongestants without cardiovascular actions in animal models of upper airway congestion.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Nasal Decongestants/pharmacology , Receptors, Adrenergic, alpha-2/metabolism , Rhinitis, Vasomotor/drug therapy , Administration, Intranasal , Administration, Oral , Adrenergic alpha-2 Receptor Agonists/administration & dosage , Adrenergic alpha-2 Receptor Agonists/pharmacokinetics , Adrenergic alpha-2 Receptor Agonists/therapeutic use , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Animals , Cats , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Nasal Decongestants/administration & dosage , Nasal Decongestants/pharmacokinetics , Nasal Decongestants/therapeutic use , Nasal Mucosa/blood supply , Nasal Mucosa/drug effects , Nasal Mucosa/metabolism , Rhinitis, Vasomotor/metabolism , Swine , Vasoconstriction/drug effects
12.
Pharmacol Biochem Behav ; 118: 36-45, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24211858

ABSTRACT

Akathisia is a subset of the larger antipsychotic side effect profile known as extrapyramidal syndrome (EPS). It is associated with antipsychotic treatment and is characterized as a feeling of inner restlessness that results in a compulsion to move. There are currently no primate models available to assess drug-induced akathisia; the present research was designed to address this shortcoming. We developed a novel rating scale based on both the Barnes Akathisia Rating Scale (BARS) and the Hillside Akathisia Scale (HAS) to measure the objective, observable incidence of antipsychotic-induced akathisia-like behavior in Cebus apella non-human primates (NHPs). To induce akathisia, we administered the atypical antipsychotic aripiprazole (1 mg/kg) or the selective phosphodiesterase 10A (PDE10A) inhibitor MP-10 (1-3 mg/kg). Treatment with both compounds produced significantly greater akathisia scores on the rating scale than vehicle treatment. Characteristic behaviors observed included vocalizations, stereotypies, teeth grinding, restless limb movements, and hyperlocomotion. Adenosine A2A receptor antagonists have previously been shown to be effective in blocking antipsychotic-induced EPS in primates. The selective A2A receptor antagonist, SCH 412348 (10-30 mg/kg), effectively reduced or reversed akathisia-like behavior induced by both aripiprazole and MP-10. This work represents the first NHP measurement scale of akathisia and demonstrates that NHPs are responsive to akathisia-inducing agents. As such, it provides a useful tool for the preclinical assessment of putative antipsychotics. In addition, these results provide further evidence of the utility of A2A receptor antagonists for the treatment of antipsychotic-induced movement disorders.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Akathisia, Drug-Induced/drug therapy , Akathisia, Drug-Induced/physiopathology , Akathisia, Drug-Induced/psychology , Animals , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/antagonists & inhibitors , Antipsychotic Agents/toxicity , Aripiprazole , Behavior, Animal/drug effects , Cebus , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Haloperidol/administration & dosage , Haloperidol/antagonists & inhibitors , Haloperidol/toxicity , Humans , Male , Motor Activity/drug effects , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/toxicity , Piperazines/administration & dosage , Piperazines/antagonists & inhibitors , Piperazines/toxicity , Pyrazoles/administration & dosage , Pyrazoles/antagonists & inhibitors , Pyrazoles/toxicity , Pyrimidines/pharmacology , Quinolines/administration & dosage , Quinolines/antagonists & inhibitors , Quinolines/toxicity , Quinolones/administration & dosage , Quinolones/antagonists & inhibitors , Quinolones/toxicity , Triazoles/pharmacology
13.
Foot Ankle Spec ; 6(6): 417-21, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23966263

ABSTRACT

BACKGROUND: The proportion of patients with clinical findings referable to peroneal pathology and magnetic resonance imaging (MRI)-diagnosed peroneal tendon pathology is unknown. Previous studies have correlated surgical findings with clinical data and preoperative MRI, but there are no published studies that reference clinical examination findings to imaging findings. PURPOSE: To determine the relationship between peroneal tendon pathology as diagnosed by MRI and clinical findings of peroneal tendon pathology. METHODS: Fifty-six patients who had both MRI evidence of peroneal tendon pathology and an associated clinical examination of the ankle were evaluated over a 3-year period at a tertiary care institution. Clinical examination criteria included lateral ankle tenderness, dislocation/snapping, and lateral ankle pain. A board-certified, fellowship-trained musculoskeletal radiologist confirmed the presence of MRI findings consistent with peroneal tendon pathology. RESULTS: Of the 56 patients with positive findings on MRI, 27 patients had an associated positive clinical exam and 29 patients had a negative clinical exam. The positive predictive value of MRI for peroneal tendon tears with positive clinical findings was 48% (95% confidence interval = 35% to 61%). CONCLUSION: Patients with MRI findings of peroneal tendon pathology should undergo careful clinical examination, as the positive predictive value of MRI for peroneal tendon pathology with actual clinical findings is low. This study demonstrates that peroneal tendon tears are often incidental findings on MRI.


Subject(s)
Ankle Injuries/diagnosis , Magnetic Resonance Imaging/methods , Physical Examination/methods , Tendon Injuries/diagnosis , Tendons/pathology , Adult , Ankle Injuries/surgery , Cohort Studies , Confidence Intervals , Female , Humans , Injury Severity Score , Joint Instability/diagnosis , Joint Instability/etiology , Male , Middle Aged , Predictive Value of Tests , Reproducibility of Results , Retrospective Studies , Sensitivity and Specificity , Tendinopathy/pathology , Tendinopathy/physiopathology , Tendon Injuries/complications , Tendon Injuries/pathology , Tendons/physiopathology
14.
Am J Rhinol Allergy ; 27(2): 84-90, 2013.
Article in English | MEDLINE | ID: mdl-23562194

ABSTRACT

BACKGROUND: Blood vessels of the nasal mucosa are richly innervated by sympathetic nerves and neural mechanism is of great interest in upper respiratory tract disorders. This study was designed to determine the role of α2-adrenoceptors and, more specifically, α2C-adrenoceptors, on neurogenic sympathetic vasoconstrictor responses in pig nasal mucosa, and to define the pharmacologic profile of a novel selective α2C-adrenoreceptor agonist. METHODS: Electrical field stimulation (EFS) was applied to nasal mucosa strips placed in an organ bath and attached to force displacement transducers for continuous recording of isometric tension. The affinity and functional activity of compound B for α2C-adrenoceptors were determined by binding analysis and the ability of compound B to stimulate [(35)S]GTPγS binding to the receptors. Compound B was also tested in a postjunctional α2C-adrenoreceptor bioassay. RESULTS: EFS-induced contractions were partly blocked by the α2-adrenoreceptor antagonist yohimbine (41.1%) and the α2C-adrenoreceptor antagonist JP-1302 had no effect. The α2-adrenoreceptor agonist clonidine, but not compound B, exerted a significant blockade (70.6%). Compound B had high affinity (K(i) = 18 nM), produced potent agonist (EC50 = 279 nM) and good efficacy (E(max) = 73%) responses at the α2C-adrenoceptors, and displayed good functional agonist potency in the human saphenous vein α2C-adrenoreceptor bioassay (pD2 = 6.2). CONCLUSION: (1) Neurogenic vasomotor contractility is largely regulated through an α-adrenergic mechanism; (2) pig nasal mucosa possesses post- and prejunctional α2-adrenoceptors; (3) the α2C-adrenoreceptor subtype does not seem to be involved; and (4) compound B is a novel, highly selective, and potent α2C-adrenoreceptor agonist.


Subject(s)
Nasal Mucosa/drug effects , Receptors, Adrenergic, alpha-2/physiology , Acridines/pharmacology , Adrenergic Agonists/pharmacology , Adrenergic Antagonists/pharmacology , Animals , Clonidine/pharmacology , Electric Stimulation , Excitation Contraction Coupling , Nasal Mucosa/innervation , Organ Culture Techniques , Piperazines/pharmacology , Saphenous Vein/drug effects , Swine , Sympathetic Nervous System , Vasoconstriction/drug effects , Yohimbine/pharmacology
15.
Clin Rev Allergy Immunol ; 45(2): 281-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23417597

ABSTRACT

Multicentric reticulohistiocytosis (MRH) is a rare systemic inflammatory granulomatous disease that primarily manifests clinically with severe erosive arthritis and widespread papulonodular skin lesions but can involve multiple other organ systems. Despite the fact that this condition can become aggressive, debilitating as well as deforming with significant detrimental consequences, the etiology of this disease remains poorly understood. Moreover, the fact that MRH is such an uncommon disease has created an obstacle in the path of adequate clinical trials that are needed for better understanding of this phenomenon and for the development of treatment options for this patient population. In this review, we will attempt to discuss the epidemiology, pathophysiology, clinical features, associated conditions, differential diagnoses, diagnostic workup, and available treatments of MRH with the hope of creating a better understanding of this very challenging yet elusive disease process.


Subject(s)
Arthritis/diagnosis , Arthritis/epidemiology , Histiocytosis, Non-Langerhans-Cell/diagnosis , Histiocytosis, Non-Langerhans-Cell/epidemiology , Skin/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Diagnosis, Differential , Giant Cells/pathology , Granuloma/pathology , Histiocytosis, Non-Langerhans-Cell/drug therapy , Humans , Prevalence
16.
J Bone Joint Surg Am ; 94(16): 1442-7, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22992814

ABSTRACT

BACKGROUND: Rotator cuff syndrome is often treated with subacromial injection of corticosteroid and local anesthetic. It has not been established if the common injection routes of the bursa are equally accurate. METHODS: We conducted a prospective clinical trial involving seventy-five shoulders in seventy-five patients who were randomly assigned to receive a subacromial injection through an anterior, lateral, or posterior route with respect to the acromion. An experienced physician performed the injections, which contained radiopaque contrast medium, corticosteroid, and local anesthetic. After the injection, a musculoskeletal radiologist, blinded to the injection route, interpreted all of the radiographs. RESULTS: The rate of accuracy varied with the route of injection, with a rate of 56% for the posterior route, 84% for the anterior route, and 92% for the lateral route (p = 0.006; chi-square test). The accuracy of injection through the posterior route was significantly lower than that through either the anterior or the lateral route (p < 0.05 for both comparisons; Poisson regression). In addition, the accuracy of injection was significantly lower in females than in males (p < 0.006; chi-square test). Among males, no differences between the routes were noted (with accuracy rates of 89% for the posterior route, 92% for the anterior route, and 93% for the lateral route). Among females, however, the accuracy of injection was lower for the posterior route than for either the anterior or the lateral route (with accuracy rates of 38% for the posterior route, 77% for the anterior route, and 91% for the lateral route) (p < 0.05). CONCLUSIONS: The anterior and lateral routes of subacromial bursal injection were more accurate than the posterior route. The accuracy of subacromial bursal injection was significantly different between males and females, mainly because of a lower accuracy of bursal injection with use of the posterior route in females. The present study suggests that the posterior route is the least accurate method for injection of the subacromial bursa in females.


Subject(s)
Injections, Intra-Articular/methods , Pain Management/methods , Shoulder Impingement Syndrome/diagnostic imaging , Shoulder Impingement Syndrome/drug therapy , Adult , Aged , Anti-Inflammatory Agents/administration & dosage , Bursa, Synovial/diagnostic imaging , Contrast Media/administration & dosage , Drug Combinations , Female , Glucocorticoids/administration & dosage , Humans , Iopamidol/administration & dosage , Lidocaine/administration & dosage , Male , Middle Aged , Prospective Studies , Radiography , Reproducibility of Results , Sex Factors , Treatment Outcome , Triamcinolone/administration & dosage , Young Adult
17.
Acad Radiol ; 19(6): 746-51, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22578228

ABSTRACT

RATIONALE AND OBJECTIVES: At the authors' institutions, faculty members and trainees work at multiple sites scattered miles apart, making it difficult to physically attend weekly teaching conferences. As a possible solution, a weekly online musculoskeletal teaching conference was undertaken. This quickly grew to include multiple other sites around North America. The authors share their experiences to assist other radiologists in organizing similar educational conferences. MATERIALS AND METHODS: The conferences are run using the Citrix GoToMeeting online meeting system. It runs on multiple platforms, including Mac, PC, iPhone, iPad, and Android. Attendees use a wide variety of microphones, sound cards, powered speakers, and webcams. Most users have fast institutional Internet connections, though several attend via slower connections, such as 3G. RESULTS: The conference has run successfully for 2 years, with participants logging in from 24 different sites in 18 states, two Canadian provinces, and three countries. About 48 sessions are held each year, with 10 to 15 sites joining the conferences each week and about 10 to 15 cases seen each week. Most attendees are from university medical centers, though several private practice radiologists attend regularly. Screen-sharing quality is superb, with no discernible difference between local and remote slide quality. Audio quality is usually quite good, particularly for those using computer audio. Audio feedback is an occasional problem, but this issue is now more easily addressed. No single time is equally convenient for participants scattered among four to six time zones. However, some sites find the conferences sufficiently valuable to rearrange their afternoon procedure schedules to reduce conflicts with the conferences. The social aspect of visiting weekly with friends and colleagues from afar is highly valued, as are seeing the wide range of pathology from other institutions and the ability to confer with colleagues on difficult cases. The conferences have also spawned several collaborative educational projects, such as an online journal club, a published book of conference cases, and an online musculoskeletal hardware atlas. CONCLUSIONS: The weekly online musculoskeletal conference described in this report has matured over 2 years from a peculiar experiment to a very popular conference. Cases not seen locally provide enrichment, and attendees gain educational opportunities not otherwise available. Other radiology groups should be able to create and maintain similar conferences.


Subject(s)
Congresses as Topic/organization & administration , Faculty/organization & administration , Internet/organization & administration , Radiology/education , Radiology/organization & administration , Online Systems , Organizational Objectives , United States
18.
Radiol Case Rep ; 7(3): 751, 2012.
Article in English | MEDLINE | ID: mdl-27326309

ABSTRACT

We present a case of a superficial acral fibromyxoma (SAFM) of the distal aspect of the thumb with radiographic evidence of extrinsic pressure erosion of the underlying cortex. This 47-year-old woman presented with a slow-growing mass over the distal aspect of the right thumb that proved to be SAFM on surgical pathology. This is a relatively rare mesenchymal neoplasm of the periungual and subungual regions of fingers and toes.

19.
Eur J Pharmacol ; 661(1-3): 63-71, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21545797

ABSTRACT

The endogenous opioid-like peptide, nociceptin, produces anxiolytic-like effects that are mediated via the nociceptin (NOP) receptor. Similarly, synthetic, non-peptide NOP agonists produce robust anxiolytic-like effects although these effects are limited by marked side effects. In the present studies, the effects of a novel NOP receptor agonist, SCH 655842, were examined in rodent models sensitive to anxiolytic drugs and tests measuring potential adverse affects. Oral administration of SCH 655842 produced robust, anxiolytic-like effects in three species, i.e., rat, guinea pig, and mouse. Specifically, SCH 655842 was effective in rat conditioned lick suppression (3-10 mg/kg) and fear-potentiated startle (3-10 mg/kg) tests, a guinea pig pup vocalization test (1-3 mg/kg), as well as in mouse Geller-Seifter (30 mg/kg) and marble burying (30 mg/kg) tests. The anxiolytic-like effect of SCH 655842 in the conditioned lick suppression test was attenuated by the NOP antagonist, J-113397. In mice, SCH 655842 reduced locomotor activity and body temperature at doses similar to the anxiolytic-like dose and these effects were absent in NOP receptor knockout mice. In rats, SCH 655842 did not produce adverse behavioral effects up to doses of 70-100 mg/kg. Pharmacokinetic studies in the rat confirmed dose-related increases in plasma and brain levels of SCH 655842 across a wide oral dose range. Taken together, SCH 655842 may represent a NOP receptor agonist with improved tolerability compared to other members of this class although further studies are necessary to establish whether this extends to higher species.


Subject(s)
Anti-Anxiety Agents/adverse effects , Anti-Anxiety Agents/pharmacology , Azabicyclo Compounds/adverse effects , Azabicyclo Compounds/pharmacology , Receptors, Opioid/agonists , Animals , Anti-Anxiety Agents/blood , Anti-Anxiety Agents/pharmacokinetics , Azabicyclo Compounds/blood , Azabicyclo Compounds/pharmacokinetics , Behavior, Animal/drug effects , Behavior, Animal/physiology , Body Temperature/drug effects , Brain/drug effects , Brain/metabolism , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Dose-Response Relationship, Drug , Fear/drug effects , Fear/physiology , Female , Gene Knockout Techniques , Guinea Pigs , Male , Mice , Motor Activity/drug effects , Rats , Receptors, Opioid/deficiency , Receptors, Opioid/genetics , Rotarod Performance Test , Species Specificity , Vocalization, Animal/drug effects , Nociceptin Receptor
20.
J Pharmacol Exp Ther ; 337(1): 256-66, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21233198

ABSTRACT

We define the pharmacological and pharmacokinetic profiles of a novel α(2C)-adrenoceptor agonist, compound A [N-[3,4-dihydro-4-(1H-imidazol-4-ylmethyl)-2H-1,4-benzoxazin-6-yl]-N-ethyl-N'-methylurea]. This compound has high affinity (K(i)) for the human α(2C)-adrenoceptor (K(i) = 12 nM), and 190- to 260-fold selectivity over the α(2A)- and α(2B)-adrenoceptor subtypes. In cell-based functional assays, compound A produced good agonist (EC(50) = 166 nM) and efficacy (E(max) = 64%) responses at the α(2C)-adrenoceptor, much lower potency and efficacy at the α(2A)-adrenoceptor (EC(50) = 1525 nM; E(max) = 8%) and α(2B)-adrenoceptor (EC(50) = 5814 nM; E(max) = 21%) subtypes, and low or no affinity and functional activity at the α(1A)-, α(1B)-, and α(1D)-adrenoceptor subtypes. In the human saphenous vein postjunctional α(2C)-adrenoceptor bioassay, compound A functions as a potent agonist (pD(2) = 6.3). In a real-time contraction bioassay of pig nasal mucosa, compound A preferentially constricted the veins (EC(50) = 108 nM), and the magnitude of arteriolar contraction reached only 50% of the maximum venular responses. Compound A exhibited no effect on locomotor activity, sedation, and body temperature in mice (up to 100 mg/kg) and did not cause hypertension and mydriasis (30 mg/kg) in conscious rats. Compound A is orally bioavailable (24%) with good plasma exposure. This compound is a substrate for the efflux P-glycoprotein transporter, resulting in very low central nervous system (CNS) penetration. In summary, compound A is a highly selective, orally active, and non-CNS-penetrating α(2C)-adrenoceptor agonist with desirable in vitro and in vivo pharmacological properties suitable for the treatment of nasal congestion.


Subject(s)
Adrenergic Agonists/chemistry , Adrenergic Agonists/pharmacology , Methylurea Compounds/chemistry , Methylurea Compounds/pharmacology , Morpholines/chemistry , Morpholines/pharmacology , Motor Activity/drug effects , Nasal Mucosa/drug effects , Receptors, Adrenergic, alpha-2/metabolism , Saphenous Vein/drug effects , Adrenergic Agonists/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Male , Methylurea Compounds/metabolism , Mice , Mice, Inbred C57BL , Morpholines/metabolism , Motor Activity/physiology , Nasal Mucosa/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/agonists , Recombinant Proteins/metabolism , Saphenous Vein/metabolism , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...