Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Pain ; 156(1): 157-165, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25599311

ABSTRACT

The number of patients suffering from postoperative pain due to orthopedic surgery and bone fracture is projected to dramatically increase because the human life span, weight, and involvement in high-activity sports continue to rise worldwide. Joint replacement or bone fracture frequently results in skeletal pain that needs to be adequately controlled for the patient to fully participate in needed physical rehabilitation. Currently, the 2 major therapies used to control skeletal pain are nonsteroidal anti-inflammatory drugs and opiates, both of which have significant unwanted side effects. To assess the efficacy of novel therapies, mouse models of orthopedic and fracture pain were developed and evaluated here. These models, orthopedic surgery pain and bone fracture pain, resulted in skeletal pain-related behaviors that lasted 3 weeks and 8 to 10 weeks, respectively. These skeletal pain behaviors included spontaneous and palpation-induced nocifensive behaviors, dynamic weight bearing, limb use, and voluntary mechanical loading of the injured hind limb. Administration of anti-nerve growth factor before orthopedic surgery or after bone fracture attenuated skeletal pain behaviors by 40% to 70% depending on the end point being assessed. These data suggest that nerve growth factor is involved in driving pain due to orthopedic surgery or bone fracture. These animal models may be useful in developing an understanding of the mechanisms that drive postoperative orthopedic and bone fracture pain and the development of novel therapies to treat these skeletal pains.


Subject(s)
Femur/injuries , Fractures, Bone/drug therapy , Nerve Growth Factor/antagonists & inhibitors , Orthopedic Procedures/adverse effects , Pain, Postoperative/drug therapy , Pain/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Femur/diagnostic imaging , Fractures, Bone/diagnostic imaging , Male , Mice , Mice, Inbred C3H , Pain/diagnostic imaging , Pain, Postoperative/diagnostic imaging , Radiography , Time Factors
2.
Cancer Res ; 74(23): 7014-23, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25287160

ABSTRACT

Studies in animals and humans show that blockade of nerve growth factor (NGF) attenuates both malignant and nonmalignant skeletal pain. While reduction of pain is important, a largely unanswered question is what other benefits NGF blockade might confer in patients with bone cancer. Using a mouse graft model of bone sarcoma, we demonstrate that early treatment with an NGF antibody reduced tumor-induced bone destruction, delayed time to bone fracture, and increased the use of the tumor-bearing limb. Consistent with animal studies in osteoarthritis and head and neck cancer, early blockade of NGF reduced weight loss in mice with bone sarcoma. In terms of the extent and time course of pain relief, NGF blockade also reduced pain 40% to 70%, depending on the metric assessed. Importantly, this analgesic effect was maintained even in animals with late-stage disease. Our results suggest that NGF blockade immediately upon detection of tumor metastasis to bone may help preserve the integrity and use, delay the time to tumor-induced bone fracture, and maintain body weight.


Subject(s)
Antibodies/pharmacology , Bone Neoplasms/drug therapy , Nerve Growth Factor/antagonists & inhibitors , Nerve Growth Factors/antagonists & inhibitors , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Extremities/pathology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Male , Mice , Mice, Inbred C3H , Neoplasm Metastasis , Nerve Growth Factor/metabolism , Nerve Growth Factors/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Pain/drug therapy , Pain/metabolism , Random Allocation , Sarcoma/drug therapy , Sarcoma/metabolism , Sarcoma/pathology
3.
J Investig Med ; 60(8): 1131-40, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23138008

ABSTRACT

The blood-brain barrier (BBB) is a physical and metabolic barrier that separates the central nervous system from the peripheral circulation. Central nervous system drug delivery across the BBB is challenging, primarily because of the physical restriction of paracellular diffusion between the endothelial cells that comprise the microvessels of the BBB and the activity of efflux transporters that quickly expel back into the capillary lumen a wide variety of xenobiotics. Therapeutic manipulation of protein trafficking is emerging as a novel means of modulating protein function, and in this minireview, the targeting of the trafficking of 2 key BBB proteins, P-glycoprotein and occludin, is presented as a novel, reversible means of optimizing central nervous system drug delivery.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blood-Brain Barrier/metabolism , Central Nervous System Agents/metabolism , Drug Delivery Systems/methods , Occludin/metabolism , Animals , Blood-Brain Barrier/drug effects , Central Nervous System Agents/administration & dosage , Humans , Protein Binding/physiology , Protein Transport/physiology
4.
J Neurochem ; 122(5): 962-75, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22716933

ABSTRACT

P-glycoprotein (ABCB1/MDR1, EC 3.6.3.44), the major efflux transporter at the blood-brain barrier (BBB), is a formidable obstacle to CNS pharmacotherapy. Understanding the mechanism(s) for increased P-glycoprotein activity at the BBB during peripheral inflammatory pain is critical in the development of novel strategies to overcome the significant decreases in CNS analgesic drug delivery. In this study, we employed the λ-carrageenan pain model (using female Sprague-Dawley rats), combined with confocal microscopy and subcellular fractionation of cerebral microvessels, to determine if increased P-glycoprotein function, following the onset of peripheral inflammatory pain, is associated with a change in P-glycoprotein trafficking which leads to pain-induced effects on analgesic drug delivery. Injection of λ-carrageenan into the rat hind paw induced a localized, inflammatory pain (hyperalgesia) and simultaneously, at the BBB, a rapid change in colocalization of P-glycoprotein with caveolin-1, a key scaffolding/trafficking protein. Subcellular fractionation of isolated cerebral microvessels revealed that the bulk of P-glycoprotein constitutively traffics to membrane domains containing high molecular weight, disulfide-bonded P-glycoprotein-containing structures that cofractionate with membrane domains enriched with monomeric and high molecular weight, disulfide-bonded, caveolin-1-containing structures. Peripheral inflammatory pain promoted a dynamic redistribution between membrane domains of P-glycoprotein and caveolin-1. Disassembly of high molecular weight P-glycoprotein-containing structures within microvascular endothelial luminal membrane domains was accompanied by an increase in ATPase activity, suggesting a potential for functionally active P-glycoprotein. These results are the first observation that peripheral inflammatory pain leads to specific structural changes in P-glycoprotein responsible for controlling analgesic drug delivery to the CNS.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blood-Brain Barrier/physiopathology , Hyperalgesia/etiology , Hyperalgesia/pathology , Neurogenic Inflammation/complications , Adenosine Triphosphatases/metabolism , Animals , Blood-Brain Barrier/drug effects , Carrageenan/toxicity , Caveolin 1/metabolism , Disease Models, Animal , Female , Hyperalgesia/drug therapy , Microvessels/drug effects , Microvessels/metabolism , Microvessels/pathology , Molecular Weight , Neurogenic Inflammation/chemically induced , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley
5.
Am J Physiol Heart Circ Physiol ; 302(3): H582-93, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22081706

ABSTRACT

Our laboratory has shown that λ-carrageenan-induced peripheral inflammatory pain (CIP) can alter tight junction (TJ) protein expression and/or assembly leading to changes in blood-brain barrier xenobiotic permeability. However, the role of reactive oxygen species (ROS) and subsequent oxidative stress during CIP is unknown. ROS (i.e., superoxide) are known to cause cellular damage in response to pain/inflammation. Therefore, we examined oxidative stress-associated effects at the blood-brain barrier (BBB) in CIP rats. During CIP, increased staining of nitrosylated proteins was detected in hind paw tissue and enhanced presence of protein adducts containing 3-nitrotyrosine occurred at two molecular weights (i.e., 85 and 44 kDa) in brain microvessels. Tempol, a pharmacological ROS scavenger, attenuated formation of 3-nitrotyrosine-containing proteins in both the hind paw and in brain microvessels when administered 10 min before footpad injection of λ-carrageenan. Similarly, CIP increased 4-hydroxynoneal staining in brain microvessels and this effect was reduced by tempol. Brain permeability to [(14)C]sucrose and [(3)H]codeine was increased, and oligomeric assemblies of occludin, a critical TJ protein, were altered after 3 h CIP. Tempol attenuated both [(14)C]sucrose and [(3)H]codeine brain uptake as well as protected occludin oligomers from disruption in CIP animals, suggesting that ROS production/oxidative stress is involved in modulating BBB functional integrity during pain/inflammation. Interestingly, tempol administration reduced codeine analgesia in CIP animals, indicating that oxidative stress during pain/inflammation may affect opioid delivery to the brain and subsequent efficacy. Taken together, our data show for the first time that ROS pharmacological scavenging is a viable approach for maintaining BBB integrity and controlling central nervous system drug delivery during acute inflammatory pain.


Subject(s)
Blood-Brain Barrier , Capillary Permeability/drug effects , Cyclic N-Oxides/pharmacology , Membrane Proteins/metabolism , Neuralgia , Xenobiotics/pharmacokinetics , Acute Disease , Aldehydes/pharmacokinetics , Analgesics, Opioid/pharmacokinetics , Animals , Antioxidants/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Capillary Permeability/immunology , Carbon Radioisotopes , Codeine/pharmacokinetics , Cysteine Proteinase Inhibitors/pharmacokinetics , Hyperalgesia/drug therapy , Hyperalgesia/immunology , Hyperalgesia/metabolism , Male , Membrane Proteins/immunology , Neuralgia/drug therapy , Neuralgia/immunology , Neuralgia/metabolism , Neuritis/drug therapy , Neuritis/immunology , Neuritis/metabolism , Occludin , Oxidative Stress/immunology , Rats , Rats, Sprague-Dawley , Spin Labels , Sucrose/pharmacokinetics , Tight Junctions/drug effects , Tight Junctions/immunology , Tight Junctions/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
6.
J Cereb Blood Flow Metab ; 30(9): 1625-36, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20234382

ABSTRACT

The blood-brain barrier (BBB) has a critical role in central nervous system homeostasis. Intercellular tight junction (TJ) protein complexes of the brain microvasculature limit paracellular diffusion of substances from the blood into the brain. Hypoxia and reoxygenation (HR) is a central component to numerous disease states and pathologic conditions. We have previously shown that HR can influence the permeability of the BBB as well as the critical TJ protein occludin. During HR, free radicals are produced, which may lead to oxidative stress. Using the free radical scavenger tempol (200 mg/kg, intraperitoneal), we show that oxidative stress produced during HR (6% O(2) for 1 h, followed by room air for 20 min) mediates an increase in BBB permeability in vivo using in situ brain perfusion. We also show that these changes are associated with alterations in the structure and localization of occludin. Our data indicate that oxidative stress is associated with movement of occludin away from the TJ. Furthermore, subcellular fractionation of cerebral microvessels reveals alterations in occludin oligomeric assemblies in TJ associated with plasma membrane lipid rafts. Our data suggest that pharmacological inhibition of disease states with an HR component may help preserve BBB functional integrity.


Subject(s)
Blood-Brain Barrier/physiology , Hypoxia, Brain/metabolism , Membrane Proteins/metabolism , Oxidative Stress/physiology , Animals , Blotting, Western , Capillaries/metabolism , Capillaries/physiology , Centrifugation, Density Gradient , Cerebrovascular Circulation/physiology , Cyclic N-Oxides/pharmacology , Electrophoresis, Polyacrylamide Gel , Female , Fluorescent Antibody Technique , Free Radical Scavengers/pharmacology , HSP70 Heat-Shock Proteins/biosynthesis , Hypoxia, Brain/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Indicators and Reagents , Microscopy, Confocal , Occludin , Permeability , Rats , Rats, Sprague-Dawley , Spin Labels , Translocation, Genetic
7.
J Neurochem ; 110(1): 58-71, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19457074

ABSTRACT

Hypoxic (low oxygen) and reperfusion (post-hypoxic reoxygenation) phases of stroke promote an increase in microvascular permeability at tight junctions (TJs) of the blood-brain barrier (BBB) that may lead to cerebral edema. To investigate the effect of hypoxia (Hx) and reoxygenation on oligomeric assemblies of the transmembrane TJ protein occludin, rats were subjected to either normoxia (Nx, 21% O(2), 60 min), Hx (6% O(2), 60 min), or hypoxia/reoxygenation (H/R, 6% O(2), 60 min followed by 21% O(2), 10 min). After treatment, cerebral microvessels were isolated, fractionated by detergent-free density gradient centrifugation, and occludin oligomeric assemblies associated with plasma membrane lipid rafts were solubilized by perfluoro-octanoic acid (PFO) exclusively as high molecular weight protein complexes. Analysis by non-reducing and reducing sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis/western blot of PFO-solubilized occludin revealed that occludin oligomeric assemblies co-localizing with 'TJ-associated' raft domains contained a high molecular weight 'structural core' that was resistant to disassembly by either SDS or a hydrophilic reducing agent ex vivo, and by Hx and H/R conditions in vivo. However, exposure of PFO-solubilized occludin oligomeric assemblies to SDS ex vivo revealed the non-covalent association of a significant amount of dimeric and monomeric occludin isoforms to the disulfide-bonded inner core, and dispersal of these non-covalently attached occludin subunits to lipid rafts of higher density in vivo was differentially promoted by Hx and H/R. Our data suggest a model of isoform interaction within occludin oligomeric assemblies at the BBB that enables occludin to simultaneously perform a structural role in inhibiting paracellular diffusion, and a signaling role involving interactions of dimeric and monomeric occludin isoforms with a variety of regulatory molecules within different plasma membrane lipid raft domains.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Edema/metabolism , Hypoxia, Brain/metabolism , Membrane Proteins/metabolism , Reperfusion Injury/metabolism , Tight Junctions/metabolism , Animals , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Blotting, Western , Brain Edema/pathology , Brain Edema/physiopathology , Cerebral Arteries/chemistry , Cerebral Arteries/metabolism , Cerebral Arteries/ultrastructure , Diffusion , Electrophoresis, Polyacrylamide Gel , Female , Hypoxia, Brain/pathology , Hypoxia, Brain/physiopathology , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Membrane Microdomains/ultrastructure , Membrane Proteins/analysis , Membrane Proteins/chemistry , Models, Molecular , Occludin , Protein Multimerization/physiology , Protein Subunits/chemistry , Protein Subunits/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Stress, Physiological/physiology , Subcellular Fractions/metabolism , Tight Junctions/chemistry , Tight Junctions/pathology
8.
J Neurochem ; 106(6): 2395-409, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18647175

ABSTRACT

Tight junctions (TJs) at the blood-brain barrier (BBB) dynamically alter paracellular diffusion of blood-borne substances from the peripheral circulation to the CNS in response to external stressors, such as pain, inflammation, and hypoxia. In this study, we investigated the effect of lambda-carrageenan-induced peripheral inflammatory pain (i.e., hyperalgesia) on the oligomeric assembly of the key TJ transmembrane protein, occludin. Oligomerization of integral membrane proteins is a critical step in TJ complex assembly that enables the generation of tightly packed, large multiprotein complexes capable of physically obliterating the interendothelial space to inhibit paracellular diffusion. Intact microvessels isolated from rat brains were fractionated by detergent-free density gradient centrifugation, and gradient fractions were analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis/ Western blot. Injection of lambda-carrageenan into the rat hind paw produced after 3 h a marked change in the relative amounts of oligomeric, dimeric, and monomeric occludin isoforms associated with different plasma membrane lipid raft domains and intracellular compartments in endothelial cells at the BBB. Our findings suggest that increased BBB permeability (i.e., leak) associated with lambda-carrageenan-induced peripheral inflammatory pain is promoted by the disruption of disulfide-bonded occludin oligomeric assemblies, which renders them incapable of forming an impermeant physical barrier to paracellular transport.


Subject(s)
Blood-Brain Barrier/metabolism , Hyperalgesia/physiopathology , Inflammation/physiopathology , Membrane Proteins/metabolism , Tight Junctions/metabolism , Animals , Biological Transport, Active/drug effects , Blood-Brain Barrier/physiopathology , Carrageenan/pharmacology , Cell Compartmentation/drug effects , Diffusion/drug effects , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Female , Hyperalgesia/chemically induced , Hyperalgesia/complications , Inflammation/chemically induced , Inflammation/complications , Macromolecular Substances/metabolism , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Membrane Proteins/biosynthesis , Microcirculation/metabolism , Microcirculation/ultrastructure , Occludin , Protein Binding/drug effects , Protein Subunits/metabolism , Rats , Rats, Sprague-Dawley , Tight Junctions/ultrastructure
9.
J Neurochem ; 103(6): 2540-55, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17931362

ABSTRACT

Tight junctions (TJs) are major components of the blood-brain barrier (BBB) that physically obstruct the interendothelial space and restrict paracellular diffusion of blood-borne substances from the peripheral circulation to the CNS. TJs are dynamic structures whose intricate arrangement of oligomeric transmembrane and accessory proteins rapidly alters in response to external stressors to produce changes in BBB permeability. In this study, we investigate the constitutive trafficking of the TJ transmembrane proteins occludin and claudin-5 that are essential for forming the TJ seal between microvascular endothelial cells that inhibits paracellular diffusion. Using a novel, detergent-free OptiPrep density-gradient method to fractionate rat cerebral microvessels, we identify a plasma membrane lipid raft domain that contains oligomeric occludin and claudin-5. Our data suggest that oligomerization of occludin involves disulfide bond formation within transmembrane regions, and that assembly of the TJ oligomeric protein complex is facilitated by an oligomeric caveolin scaffold. This is the first time that distribution of oligomeric TJ transmembrane proteins within plasma membrane lipid rafts at the BBB has been examined in vivo. The findings reported in this study are critical to understand the mechanism of assembly of the TJ multiprotein complex that is essential for maintaining BBB integrity.


Subject(s)
Blood-Brain Barrier/embryology , Blood-Brain Barrier/physiology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Tight Junctions/metabolism , Tight Junctions/physiology , Animals , Blotting, Western , Capillaries/metabolism , Cell Membrane/metabolism , Claudin-5 , Disulfides/chemistry , Electrophoresis, Polyacrylamide Gel , Female , Hydrogen-Ion Concentration , Indicators and Reagents , Membrane Microdomains/metabolism , Membrane Proteins/metabolism , Microscopy, Confocal , Occludin , Phosphoproteins/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Subcellular Fractions/metabolism , Zonula Occludens-1 Protein
10.
Arthritis Rheum ; 54(11): 3452-64, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17075840

ABSTRACT

OBJECTIVE: Scientific evidence is lacking for the antiarthritic efficacy of turmeric dietary supplements that are being promoted for arthritis treatment. Therefore, we undertook studies to determine the antiarthritic efficacy and mechanism of action of a well-characterized turmeric extract using an animal model of rheumatoid arthritis (RA). METHODS: The composition of commercial turmeric dietary supplements was determined by high-performance liquid chromatography. A curcuminoid-containing turmeric extract similar in composition to these supplements was isolated and administered intraperitoneally to female Lewis rats prior to or after the onset of streptococcal cell wall-induced arthritis. Efficacy in preventing joint swelling and destruction was determined clinically, histologically, and by measurement of bone mineral density. Mechanism of action was elucidated by analysis of turmeric's effect on articular transcription factor activation, microarray analysis of articular gene expression, and verification of the physiologic effects of alterations in gene expression. RESULTS: A turmeric fraction depleted of essential oils profoundly inhibited joint inflammation and periarticular joint destruction in a dose-dependent manner. In vivo treatment prevented local activation of NF-kappaB and the subsequent expression of NF-kappaB-regulated genes mediating joint inflammation and destruction, including chemokines, cyclooxygenase 2, and RANKL. Consistent with these findings, inflammatory cell influx, joint levels of prostaglandin E(2), and periarticular osteoclast formation were inhibited by turmeric extract treatment. CONCLUSION: These translational studies demonstrate in vivo efficacy and identify a mechanism of action for a well-characterized turmeric extract that supports further clinical evaluation of turmeric dietary supplements in the treatment of RA.


Subject(s)
Antioxidants/pharmacology , Arthritis, Experimental/drug therapy , Plant Extracts/pharmacology , Animals , Arthritis, Experimental/pathology , Bacterial Outer Membrane Proteins/pharmacology , Curcuma , Dietary Supplements , Dinoprostone/metabolism , Disease Models, Animal , Female , Gene Expression/drug effects , Granuloma/drug therapy , Granuloma/pathology , Joints/metabolism , Joints/pathology , Liver/pathology , Monocytes/drug effects , Monocytes/pathology , NF-kappa B/metabolism , Neutrophils/drug effects , Neutrophils/pathology , Osteoclasts/drug effects , Osteoclasts/pathology , Rats , Rats, Inbred Lew , Rhizome/chemistry , Spleen/pathology , Streptococcus
SELECTION OF CITATIONS
SEARCH DETAIL
...