Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Neuroscience ; 170(2): 633-44, 2010 Oct 13.
Article in English | MEDLINE | ID: mdl-20633608

ABSTRACT

Neuroinflammation is associated with glial activation following a variety of brain injuries, including stroke. While activation of perilesional astrocytes and microglia following ischemic brain injury is well documented, the influence of age on these cellular responses after stroke is unclear. This study investigated the influence of advanced age on neuronal degeneration, neuroinflammation, and glial activation in female Sprague-Dawley rats after reversible embolic occlusion of the middle cerebral artery (MCAO). Results indicate that in comparison to young adult rats (3 months), aged rats (18 months) showed enhanced neuronal degeneration, altered microglial response, and a markedly increased expression of proinflammatory cytokines/chemokines following MCAO. In addition, the time-course for activation of signal transducers and activators of transcription 3 (STAT3), the signaling mechanism that regulates astrocyte reactivity, was truncated in the aged rats after MCAO. Moreover, the expression of suppressor of cytokine signaling 3 (SOCS3), which is associated with termination of astrogliosis, was enhanced as a function of age after MCAO. These findings are suggestive of an enhanced proinflammatory response and a truncated astroglial response as a function of advanced age following MCAO. These data provide further evidence of the prominent role played by age in the molecular and cellular responses to ischemic stroke and suggest that astrocytes may represent targets for future therapies aimed at improving stroke outcome.


Subject(s)
Brain Ischemia/immunology , Cytokines/metabolism , Nerve Degeneration/pathology , Signal Transduction/immunology , Stroke/immunology , Age Factors , Animals , Astrocytes/pathology , Brain/immunology , Brain/metabolism , Brain/pathology , Brain Ischemia/metabolism , Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Disease Models, Animal , Female , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/metabolism , Microglia/pathology , Protein Transport , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Stroke/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism
2.
Curr Pharm Des ; 14(16): 1594-600, 2008.
Article in English | MEDLINE | ID: mdl-18673200

ABSTRACT

From a complications standpoint, diabetes mellitus is a disease of the vasculature. Diabetics face a considerably higher risk of developing cardiovascular and cerebrovascular diseases. Both large and small blood vessels are susceptible to alterations from diabetes. Endothelial cell dysfunction associated with small vessel (known as microangiopathy) is a primary factor in the development and progression of diabetes-related disabilities, including blindness, kidney failure, and peripheral neuropathy. Recent clinical evidence show that people with diabetes have increased incidences of vascular dementia, ventricular hypertrophy, lacunar infarcts, hemorrhage, and may be a predisposing factor for Alzheimer's disease. However, the effects of diabetes mellitus on the cerebral microvascular are still largely unknown. This communication will review the relationship between diabetes mellitus and changes in cognition with a particular focus on how alterations in blood-brain barrier structure and function may play a long term role in worsened cognitive abilities.


Subject(s)
Blood-Brain Barrier , Cognition/physiology , Diabetes Mellitus/physiopathology , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Blood-Brain Barrier/ultrastructure , Brain/blood supply , Brain/physiopathology , Cognition Disorders/etiology , Cognition Disorders/metabolism , Cognition Disorders/pathology , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Glucose/metabolism , Humans , Microcirculation/metabolism , Microcirculation/pathology , Microcirculation/physiopathology
3.
Am J Physiol Heart Circ Physiol ; 290(2): H732-40, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16199477

ABSTRACT

Previous studies showed that peripheral inflammatory pain increased blood-brain barrier (BBB) permeability and altered tight junction protein expression and the delivery of opioid analgesics to the brain. What remains unknown is which pathways and mediators during peripheral inflammation affect BBB function and structure. The current study investigated effects of lambda-carrageenan-induced inflammatory pain (CIP) on BBB expression of ICAM-1. We also examined the systemic contribution of a number of proinflammatory cytokines and microglial activation in the brain to elucidate pathways involved in BBB disruption during CIP. We investigated ICAM-1 RNA and protein expression levels in isolated rat brain microvessels after CIP using RT-PCR and Western blot analyses, screened inflammatory cytokines during the time course of inflammation, assessed white blood cell counts, and probed for BBB and central nervous system stimulation and leukocyte transmigration using immunohistochemistry and flow cytometry. Results showed an early increase in ICAM-1 RNA and protein expression after CIP with no change in circulating levels of several proinflammatory cytokines. Changes in ICAM-1 protein expression were noted at 48 h. Immunohistochemistry showed that the induction of ICAM-1 was region specific with increased expression noted in the thalamus and frontal and parietal cortices, which directly correlated with increased expression of activated microglia. The findings of the present study were that CIP induces increased ICAM-1 mRNA and protein expression at the BBB and that systemic proinflammatory mediators play no apparent role in the early response (1-6 h); however, brain region-specific increases in microglial activation suggest a potential for a central-mediated response.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/physiopathology , Encephalitis/physiopathology , Intercellular Adhesion Molecule-1/metabolism , Microglia , Pain/physiopathology , Animals , Blood Cell Count , Blood Vessels/metabolism , Blotting, Western , Brain/blood supply , Brain/pathology , Carrageenan , Cytokines/blood , Encephalitis/chemically induced , Encephalitis/pathology , Female , Immunohistochemistry , Immunophenotyping , Inflammation Mediators/blood , Intercellular Adhesion Molecule-1/genetics , Macrophages/pathology , Microcirculation , Neutrophil Infiltration , Pain/chemically induced , Pain/pathology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
4.
Am J Physiol Heart Circ Physiol ; 283(4): H1531-7, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12234806

ABSTRACT

In this study, we examined the effect of lambda-carrageenan-induced inflammatory pain on the functional and structural properties of the rat blood-brain barrier (BBB) over a 72-h time period. Systemic inflammation was induced by an intraplantar injection of 3% lambda-carrageenan into the right hind paw of female Sprague-Dawley rats. In situ brain perfusion and Western blot analyses were performed at 1, 3, 6, 12, 24, 48, and 72 h. In situ brain perfusion showed lambda-carrageenan significantly increased brain uptake of [(14)C]sucrose at 1, 3, 6, and 48 h (139 +/- 9%, 166 +/- 19%, 138 +/- 13%, and 146 +/- 7% compared with control, respectively). Capillary depletion analysis insured the increased brain uptake was due to increased BBB permeability and not vascular trapping. Western blot analyses for zonula occludens-1 (ZO-1) and occludin were performed on isolated cerebral microvessels. ZO-1 expression was significantly increased at 1, 3, and 6 h and returned to control expression levels by 12 h. Total occludin expression was significantly reduced at 1, 3, 6, 12, and 48 h. This investigation demonstrated that lambda-carrageenan-induced inflammatory pain elicits a biphasic increase in BBB permeability with the first phase occurring from 1-6 h and the second phase occuring at 48 h. Furthermore, changes in BBB function are correlated with altered tight junctional protein expression of occludin and ZO-1. Changes in the structure of tight junctions may have important clinical ramifications concerning central nervous system homeostasis and therapeutic drug delivery.


Subject(s)
Blood-Brain Barrier/physiology , Pain/physiopathology , Tight Junctions/physiology , Animals , Carrageenan , Female , Guanylate Kinases , Immunoblotting , Inflammation/chemically induced , Inflammation/physiopathology , Membrane Proteins/analysis , Nucleoside-Phosphate Kinase/analysis , Occludin , Pain/chemically induced , Perfusion , Phosphoproteins/analysis , Precipitin Tests , Rats , Rats, Sprague-Dawley , Tight Junctions/chemistry , Zonula Occludens-1 Protein , Zonula Occludens-2 Protein
5.
J Pharmacol Exp Ther ; 299(3): 967-72, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11714884

ABSTRACT

Neuropeptide pharmaceuticals have potential for the treatment of neurological disorders, but the blood-brain barrier (BBB) limits entry of peptides to the brain. Several strategies to improve brain delivery are currently under investigation, including glycosylation. In this study we investigated the effect of O-linked glycosylation on Ser(6) of a linear opioid peptide amide Tyr-D-Thr-Gly-Phe-Leu-Ser-NH(2) on metabolic stability, BBB transport, and analgesia. Peptide stability was studied in brain and serum from both rat and mouse by high-performance liquid chromatography. BBB transport properties were investigated by rat in situ perfusion. Tail-flick analgesia studies were performed on male ICR mice, injected i.v. with 100 microg of peptide ligand. Glycosylation of Ser(6) of the peptide led to a significant increase in enzymatic stability in both serum and brain. Glycosylation significantly increased the BBB permeability of the peptide from a value of 1.0 +/- 0.2 microl x min(-1) x g(-1) to 2.2 +/- 0.2 microl x min(-1) x g(-1) (p < 0.05), without significantly altering the initial volume of distribution. Analgesia studies showed that the glycosylated peptide gave a significantly improved analgesia after i.v. administration compared with nonglycosylated peptide. The improved analgesia profile shown by the glycosylated peptide is due in part to an improvement in bioavailability to the central nervous system. The bioavailability is increased by improving stability and transport into the brain.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Enkephalin, Leucine/pharmacokinetics , Opioid Peptides/pharmacokinetics , Analgesics, Opioid/chemistry , Analgesics, Opioid/therapeutic use , Animals , Blood-Brain Barrier , Drug Stability , Enkephalin, Leucine/analogs & derivatives , Enkephalin, Leucine/chemistry , Enkephalin, Leucine/therapeutic use , Glycosylation , Male , Mice , Mice, Inbred ICR , Opioid Peptides/chemistry , Opioid Peptides/therapeutic use , Pain/drug therapy , Perfusion
6.
Trends Neurosci ; 24(12): 719-25, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11718877

ABSTRACT

Disruption of the tight junctions (TJs) of the blood-brain barrier (BBB) is a hallmark of many CNS pathologies, including stroke, HIV encephalitis, Alzheimer's disease, multiple sclerosis and bacterial meningitis. Furthermore, systemic-derived inflammation has recently been shown to cause BBB tight junctional disruption and increased paracellular permeability. The BBB is capable of rapid modulation in response to physiological stimuli at the cytoskeletal level, which enables it to protect the brain parenchyma and maintain a homeostatic environment. By allowing the "loosening" of TJs and an increase in paracellular permeability, the BBB is able to "bend without breaking"; thereby, maintaining structural integrity.


Subject(s)
Blood-Brain Barrier/physiology , Brain Diseases/physiopathology , Tight Junctions/physiology , Animals , Brain/blood supply , Brain/physiology , Humans
7.
Pharmacol Biochem Behav ; 69(3-4): 451-9, 2001.
Article in English | MEDLINE | ID: mdl-11509204

ABSTRACT

The present study assessed the ability of N-methyl-D-aspartate (NMDA) receptor antagonist, dizocilpine (MK-801), to modulate neonatal cocaine-induced neurobehavioral changes in the rat. Sprague-Dawley rats were randomly assigned on postnatal day 0 (PND 0) to one of four treatment groups. Treatments began on PND 4 and continued until PND 10. Treatments consisted of an oral bolus of either cocaine HCl (40 mg/kg), (+)MK-801 (0.4 mg/kg), (+)MK-801 (0.4 mg/kg) followed 30 min later with cocaine HCl (40 mg/kg) or 0.9% saline. On PND 21, 30, 40 and 60, males and females were examined for stress response using the cold-water swim test. Cocaine-treated male and female rats exhibited significantly diminished tolerance to cold-water stress compared to control and MK-801/cocaine-treated groups. In addition, neonatal exposure to cocaine was associated with increased severity of motor symptoms (tail twitches, wet dog shaking and convulsions) following the administration of NMDA (35 mg/kg). Treatment groups were also tested for pain sensitivity using the tail flick (TF) and hot plate (HP) methods. The results indicated that neonatal cocaine exposure altered pain sensitivity in both tests. NMDA receptor binding studies showed a significant increase in receptor densities in the hippocampus and hypothalamus of the cocaine-treated group compared to control. MK-801 administered to rat pups before cocaine treatment blocked the increase in receptor density. The results indicated that neonatal cocaine exposure was associated with altered responses to NMDA, stress tolerance and pain sensitivity. Moreover, the pretreatment with NMDA receptor antagonist, MK-801, abolished or attenuated these cocaine-induced neurobehavioral changes.


Subject(s)
Cocaine/toxicity , Dopamine Uptake Inhibitors/toxicity , Receptors, N-Methyl-D-Aspartate/metabolism , Adrenocorticotropic Hormone/blood , Animals , Animals, Newborn , Corticosterone/blood , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Male , N-Methylaspartate/pharmacology , Pain Measurement/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Stress, Physiological/blood
8.
J Pharmacol Exp Ther ; 298(2): 848-56, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11454951

ABSTRACT

Poly(ethylene glycol), or PEG, conjugation to proteins and peptides is a growing technology used to enhance efficacy of therapeutics. This investigation assesses pharmacodynamic and pharmacokinetic characteristics of PEG-conjugated [D-Pen2,D-Pen5]-enkephalin (DPDPE), a met-enkephalin analog, in rodent (in vivo, in situ) and bovine (in vitro) systems. PEG-DPDPE showed increased analgesia (i.v.) compared with nonconjugated form (p < 0.01), despite a 172-fold lower binding affinity for the delta-opioid receptor. [125I]PEG-DPDPE had a 36-fold greater hydrophilicity (p < 0.01) and 12% increase in the unbound plasma protein fraction (p < 0.01), compared with [(125)I]DPDPE. [125I]PEG-DPDPE had a 2.5-fold increase in elimination half-life (p < 0.01), 2.7-fold decrease in volume of distribution (p < 0.01), and a 7-fold decrease in plasma clearance rate (p < 0.01) to [125I]DPDPE. Time course distribution showed significant concentration differences (p < 0.01) in plasma, whole blood, liver, gallbladder, gastrointestinal (GI) content, GI tract, kidneys, spleen, urine, and brain (brain, p < 0.05), between the conjugated and nonconjugated forms. Increased brain uptake of [(125)I]PEG-DPDPE corresponded to analgesia data. [125I]PEG-DPDPE in brain was shown to be 58.9% intact, with 41.1% existing as [125I]DPDPE (metabolite), whereas [125I]DPDPE was 25.7% intact in the brain (at 30 min). In vitro P-glycoprotein affinity was shown for [125I]DPDPE (p < 0.01) but not shown for [125I]PEG-DPDPE. In vitro saturable uptake, with 100 microM DPDPE, was shown for [125I]PEG-DPDPE (p < 0.05). In this study, PEG-conjugated DPDPE seems to act as a prodrug, enhancing peripheral pharmacokinetics, while undergoing hydrolysis in the brain and allowing nonconjugated DPDPE to act at the receptor.


Subject(s)
Analgesics, Opioid/pharmacology , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Polyethylene Glycols/chemistry , Analgesics, Opioid/chemistry , Animals , Binding, Competitive/drug effects , Brain/metabolism , Capillary Permeability/drug effects , Cattle , Chemical Phenomena , Chemistry, Physical , Enkephalin, D-Penicillamine (2,5)-/chemistry , Female , In Vitro Techniques , Injections, Intraventricular , Iodine/chemistry , Mice , Mice, Inbred ICR , Pain Measurement/drug effects , Protein Binding , Rats , Rats, Sprague-Dawley , Tissue Distribution
9.
Am J Physiol Heart Circ Physiol ; 280(3): H1241-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179069

ABSTRACT

Effects of inflammatory pain states on functional and molecular properties of the rat blood-brain barrier (BBB) were investigated. Inflammation was produced by subcutaneous injection of formalin, lambda-carrageenan, or complete Freund's adjuvant (CFA) into the right hind paw. In situ perfusion and Western blot analyses were performed to assess BBB integrity after inflammatory insult. In situ brain perfusion determined that peripheral inflammation significantly increased the uptake of sucrose into the cerebral hemispheres. Capillary depletion and cerebral blood flow analyses indicated the perturbations were due to increased paracellular permeability rather than vascular volume changes. Western blot analyses showed altered tight junctional protein expression during peripheral inflammation. Occludin significantly decreased in the lambda-carrageenan- and CFA-treated groups. Zonula occluden-1 expression was significantly increased in all pain models. Claudin-1 protein expression was present at the BBB and remained unchanged during inflammation. Actin expression was significantly increased in the lambda-carrageenan- and CFA-treated groups. We have shown that inflammatory-mediated pain alters both the functional and molecular properties of the BBB. Inflammatory-induced changes may significantly alter delivery of therapeutic agents to the brain, thus affecting dosing regimens during chronic pain.


Subject(s)
Blood-Brain Barrier/physiology , Membrane Proteins/biosynthesis , Neurogenic Inflammation/physiopathology , Tight Junctions/metabolism , Actins/analysis , Actins/biosynthesis , Animals , Blotting, Western , Capillaries/physiology , Carbon Radioisotopes , Carrageenan , Cerebrovascular Circulation/physiology , Claudin-1 , Disinfectants , Female , Formaldehyde , Freund's Adjuvant , Membrane Proteins/analysis , Neurogenic Inflammation/chemically induced , Occludin , Rats , Rats, Sprague-Dawley , Sucrose/pharmacokinetics , Tight Junctions/chemistry
10.
Brain Res ; 890(1): 38-48, 2001 Jan 26.
Article in English | MEDLINE | ID: mdl-11164767

ABSTRACT

Pathological states (i.e. stroke, cardiac arrest) can lead to reduced blood flow to the brain potentially altering blood-brain barrier (BBB) permeability and regulatory transport functions. BBB disruption leads to increased cerebrovascular permeability, an important factor in the development of ischemic brain injury and edema formation. In this study, reduced flow was investigated to determine the effects on cerebral blood flow (CBF), pressure, basal BBB permeability, and transport of insulin and K+ across the BBB. Anesthetized adult female Sprague-Dawley rats were measured at normal flow (3.1 ml min(-1)), half flow (1.5 ml min(-1)), and quarter flow (0.75 ml min(-1)), using bilateral in situ brain perfusion for 20 min followed by capillary depletion analysis. Reduction in perfusion flow rates demonstrated a modest reduction in CBF (1.27-1.56 ml min(-1) g(-1)), a decrease in pressure, and no significant effect on basal BBB permeability indicating that autoregulation remained functional. In contrast, there was a concomittant decrease in BBB transport of both insulin and K+ with reduced flow. At half and quarter flow, insulin transport was significantly reduced (R(Br)%=17.2 and R(Br)%=16.2, respectively) from control (R(Br)%=30.4). Additionally, a significant reduction in [86Rb+] was observed at quarter flow (R(Br)%=2.5) as compared to control (R(Br)%=4.8) suggesting an alteration in ion homeostasis as a result of low flow. This investigation suggests that although autoregulation maintains CBF, BBB transport mechanisms were significantly compromised in states of reduced flow. These flow alterations may have a significant impact on brain homeostasis in pathological states.


Subject(s)
Blood-Brain Barrier/physiology , Cerebrovascular Circulation/physiology , Hypoglycemic Agents/pharmacokinetics , Insulin/pharmacokinetics , Potassium/metabolism , Animals , Blood Gas Analysis , Capillaries/cytology , Capillaries/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Iodine Radioisotopes , Rats , Rats, Sprague-Dawley , Rubidium Radioisotopes , Stroke/metabolism , Sucrose/pharmacokinetics
11.
Peptides ; 22(12): 2329-43, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11786210

ABSTRACT

Peptides have the potential to be potent pharmaceutical agents for the treatment of many central nervous system derived maladies. Unfortunately peptides are generally water-soluble compounds that will not enter the central nervous system, via passive diffusion, due to the existence of the blood-brain barrier. Peptides can also undergo metabolic deactivation by peptidases, thus further reducing their therapeutic benefits. In targeting peptides to the central nervous system consideration must be focused both on increasing bioavailability and enhancing brain uptake. To date multiple strategies have been examined with this focus. However, each strategy comes with its own complications and considerations. In this review we assess the strengths and weaknesses of many of the methods currently being examined to enhance peptide entry into the central nervous system.


Subject(s)
Blood-Brain Barrier , Peptides/chemistry , Peptides/pharmacokinetics , Biological Availability
12.
J Pharmacol Exp Ther ; 295(3): 972-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11082431

ABSTRACT

Insulin crosses the blood-brain barrier (BBB) via receptor-mediated transcytosis and has been suggested to augment uptake of peripheral substances across the BBB. The delta-opioid receptor-selective peptide D-penicillamine(2,5) (DPDPE), a Met-enkephalin analog, produces analgesia via a central nervous system-derived effect. In vitro (K(cell), microl. min(-1). mg(-1)) and in situ (K(in), microl. min(-1). g(-1)) analyses of DPDPE transport (K(cell) = 0.56 +/- 0. 15; K(in) = 0.28 +/- 0.03) revealed significant (P <.01) increases in DPDPE uptake by the BBB with 10 microM insulin (K(cell) = 1.61 +/- 0.25; K(in) = 0.48 +/- 0.04). In vitro cellular uptake was significantly increased (P <.05) at 1 microM insulin, whereas no significant uptake was observed with CTAP (a somatostatin opioid peptide analog) or sucrose (a paracellular diffusionary marker). No significant change in uptake was seen with DPDPE, CTAP, or sucrose in the presence of holo-transferrin (0-100 microM), indicating that the effect of insulin on DPDPE was not a generalized effect of receptor endocytosis. Insulin did not affect P-glycoprotein efflux, a mechanism that has shown affinity for DPDPE. A similar uptake of DPDPE into the brain (64% increase) was seen with the in situ brain perfusion model. Analgesic assessment revealed a significant decline in DPDPE (i.v.)-induced analgesia with increasing concentrations of insulin (i.v., i.c.v., s.c.) in a dose-dependent manner. Thus, insulin significantly increases DPDPE uptake across the BBB by a specific mechanism. The analgesic effect seen with DPDPE and insulin coadministration was shown to decrease, indicating that insulin reduces the analgesic effect within the central nervous system rather than at the BBB.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Blood-Brain Barrier/drug effects , Insulin/pharmacology , Analgesics, Opioid/pharmacology , Animals , Biological Transport/drug effects , Brain/metabolism , Cattle , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics
13.
Brain Res ; 881(1): 37-46, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-11033091

ABSTRACT

The blood-brain barrier prevents the entry of many potentially therapeutic peptide drugs to the brain. Glycosylation has shown potential as a methodology for improving delivery to the CNS. Previous studies have shown improved bioavailability and improved centrally mediated analgesia of glycosylated opioids. In this study we investigate the effect of glycosylation on the cyclic opioid peptide [D-Cys(2,5),Ser(6),Gly(7)] enkephalin. The peptide was glycosylated on the Ser(6) via an O-linkage with various sugar moieties and alignments. The peptides were then investigated for receptor binding, physiochemical attributes, in situ brain uptake in female Sprague-Dawley rats and antinociception in male ICR mice. Glycosylation resulted in a slight decrease in affinity to the delta-opioid receptor, and mixed effect on binding to the mu-opioid receptor. There was a significant decrease in lipophilicity resulting from glycosylation and a slight reduction in binding to bovine serum albumin. In situ perfusion showed that brain uptake was improved by up to 98% for several of the glycosylated peptides, and the nociceptive profiles of the peptides, in general, followed the rank order of peptide entry to the brain with up to a 39-fold increase in A.U.C.


Subject(s)
Blood-Brain Barrier/drug effects , Brain/metabolism , Enkephalin, Methionine/pharmacokinetics , Pain Measurement/drug effects , Receptors, Opioid, delta/metabolism , Animals , Biological Availability , Blood-Brain Barrier/physiology , Cattle , Enkephalin, Methionine/analogs & derivatives , Female , Glycosylation , Male , Mice , Rats , Rats, Sprague-Dawley
15.
J Med Chem ; 43(13): 2586-90, 2000 Jun 29.
Article in English | MEDLINE | ID: mdl-10891118

ABSTRACT

Endogenous peptides (e.g. enkephalins) control many aspects of brain function, cognition, and perception. The use of these neuroactive peptides in diverse studies has led to an increased understanding of brain function. Unfortunately, the use of brain-derived peptides as pharmaceutical agents to alter brain chemistry in vivo has lagged because peptides do not readily penetrate the blood-brain barrier. Attachment of simple sugars to enkephalins increases their penetration of the blood-brain barrier and allows the resulting glycopeptide analogues to function effectively as drugs. The delta-selective glycosylated Leu-enkephalin amide 2, H(2)N-Tyr-D-Thr-Gly-Phe-Leu-Ser(beta-D-Glc)-CONH(2), produces analgesic effects similar to morphine, even when administered peripherally, yet possesses reduced dependence liability as indicated by naloxone-precipitated withdrawal studies. Similar glycopeptide-based pharmaceuticals hold forth the promise of pain relief with improved side-effect profiles over currently available opioid analgesics.


Subject(s)
Analgesics, Opioid/chemical synthesis , Enkephalin, Leucine/analogs & derivatives , Glycopeptides/chemical synthesis , Analgesics, Opioid/adverse effects , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Animals , Blood-Brain Barrier/drug effects , Enkephalin, Leucine/chemical synthesis , Enkephalin, Leucine/chemistry , Enkephalin, Leucine/metabolism , Enkephalin, Leucine/pharmacology , Female , Glycopeptides/adverse effects , Glycopeptides/chemistry , Glycopeptides/pharmacology , Injections, Intraventricular , Pain Measurement , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Substance Withdrawal Syndrome/etiology
16.
J Neurochem ; 75(1): 424-35, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10854288

ABSTRACT

[D-Pen(2),D-Pen(5)]-Enkephalin (DPDPE) is an enzymatically stable delta-opioid receptor-selective peptide, which was modified by the trimethylation of the Phe(4) residue to give beta-methyl-2', 6'-dimethylphenylalanine (TMP), resulting in four conformations : (2R,3S)-beta-Phe-DPDPE, (2R,3R)-beta-Phe-DPDPE, (2R, 3S)-beta-Phe-DPDPE, and (2S,3R)-beta-Phe-DPDPE. Synthesis was by solid-phase techniques using enantiomerically pure amino acids to give the four optically pure diastereoisomer peptides. The potency and selectivity (delta- versus mu-opioid receptor) were evaluated by radioreceptor binding in rat brain, with a mu/delta ratio decrease for all TMP conformations, compared with the parent compound (DPDPE). Octanol/buffer distribution analysis showed enhanced lipophilicity of all TMP forms, with a sixfold enhancement associated with (2S,3S)-TMP. In situ vascular perfusion in anesthetized rats showed a 1.6-fold (p < 0.01) increase in the ratio of brain uptake for (2S,3S)-TMP and a 1.5-fold (p < 0.01) decrease in uptake for (2R,3R)-TMP. Saturability of (2S,3S)-TMP was shown (p < 0.01) against 100 microM unlabeled DPDPE, showing a shared nondiffusionary transport system. P-glycoprotein affinity was shown in situ for the parent and (2S,3S)-TMP (p < 0.01). Protein binding capacity of the TMP compounds in rat plasma and in situ mammalian bovine serum albumin-Ringer showed (2R,3S)-TMP and (2S,3R)-TMP with the lowest degree of protein binding (p < 0.01), and (2S,3S)-TMP and (2R,3R)-TMP with comparable affinities to DPDPE. Analgesia, via intravenous administration, showed significantly reduced (p < 0.01) end effect and time course for (2R,3R)-TMP, (2R,3S)-TMP, and (2S, 3R)-TMP as compared with DPDPE. These results demonstrate that topographical modification in a conformationally restricted peptide can significantly modulate potency and receptor selectivity, binding capacity, enzymatic stability, lipophilicity, P-glycoprotein affinity, and blood-brain barrier permeability, resulting in a change of bioavailability, and thereby provides insight for future peptide drug design.


Subject(s)
Alanine/analogs & derivatives , Enkephalin, D-Penicillamine (2,5)-/analogs & derivatives , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Alanine/chemistry , Alanine/metabolism , Analgesia , Animals , Blood-Brain Barrier , Brain/blood supply , Brain/metabolism , Capillaries/metabolism , Cattle , Endothelium, Vascular/metabolism , Enkephalin, D-Penicillamine (2,5)-/chemistry , Female , Methylation , Molecular Conformation , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship
17.
Org Lett ; 1(2): 187-8, 1999 Jul 29.
Article in English | MEDLINE | ID: mdl-10822559

ABSTRACT

[formula: see text] A tight-binding, hydrophobic inhibitor of carbonic anhydrase II has been masked with a water-solubilizing, photolabile group derived from o-nitrophenylglycine. This caged inhibitor represents our first effort at the site-specific delivery of prodrugs that can be activated by light. Via this approach, we have begun to address the problems of water insolubility and systemic side effects on administration of tight-binding inhibitors of carbonic anhydrase.


Subject(s)
Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrases/chemistry , Carbonic Anhydrase Inhibitors/radiation effects , Fluorometry , Kinetics , Photolysis
18.
J Wildl Dis ; 27(2): 258-64, 1991 Apr.
Article in English | MEDLINE | ID: mdl-1906114

ABSTRACT

Protection against Brucella abortus induced abortion and infection provided by strain 19 (S19) vaccination was evaluated in American bison (Bison bison). Forty-eight pregnant bison were manually inoculated (MI) with S19 vaccine, 44 were ballistically inoculated (BI) with an absorbable hollow pellet containing lyophilized S19, and 46 were manually injected with buffered saline as non-vaccinated controls (NVC). All bison were Brucella spp. seronegative prior to the experiment, in the second trimester of pregnancy, and were randomly assigned to experimental groups. Approximately 60 days post-vaccination, abortions were observed in the vaccinated bison. Brucella abortus strain 19 was recovered from a bison that had recently aborted, her fetus, and from 11 of 12 other aborted fetuses. Fifty-eight percent (53 of 92) of vaccinated bison aborted, and no abortions were observed in the NVC bison. One cow aborted during her second post-vaccinal pregnancy and S19 was identified from the dam and fetus indicating that chronic S19 infections can occur in bison. Positive antibody titers were present 10 mo post-vaccination in 73% (66 of 91) of the bison. Thirteen mo post-vaccination, 30 MI vaccinates, 27 BI vaccinates, and 30 NVC bison were challenged during the second trimester of pregnancy with 1 x 10(7) CFU of B. abortus strain 2308 via bilateral conjunctival inoculation. Protection against abortion was 67% (P less than or equal to 0.0001) for vaccinated bison compared to 4% in NVC. Protection against B. abortus infection was determined to be 39% (P greater than or equal to 0.001) for vaccinates and 0% (zero of 30) for NCV. Persistent antibody titers, vaccine induced abortions, and chronic S19 infections indicate that the S19 vaccine doses used in this study are not suitable for pregnant bison.


Subject(s)
Bison , Brucella Vaccine , Brucella abortus/immunology , Brucellosis/veterinary , Pregnancy Complications, Infectious/veterinary , Abortion, Veterinary/prevention & control , Animals , Brucella Vaccine/administration & dosage , Brucellosis/prevention & control , Female , Injections, Intramuscular , Injections, Subcutaneous/veterinary , Pregnancy , Pregnancy Complications, Infectious/prevention & control , Random Allocation , Vaccination/veterinary
19.
J Bone Joint Surg Br ; 72(5): 866-73, 1990 Sep.
Article in English | MEDLINE | ID: mdl-2211774

ABSTRACT

To study the anatomy of subarticular bone and cartilage, fresh specimens of cartilage on bone from the human shoulder, hip and knee were treated with bleach or papain, or were fixed and decalcified. All were compared using scanning electron microscopy. Papain digestion selectively removed cartilage to the tidemark. The tidemark contour was highly variable; irregularities were indirectly related to degenerative lesions and were most prominent in peripheral non-weight-bearing areas of joints with central fibrillation. Decalcification exposed the interface between the bone and calcified cartilage. Collagen fibrils in articular cartilage did not interdigitate with those of bone. The subchondral bone was appositional, avascular, smooth and very thin in most areas of human joints. Perforations through subchondral bone or calcified cartilage were rare. Bleach maceration destroyed important details.


Subject(s)
Cartilage, Articular/anatomy & histology , Adolescent , Adult , Aged , Collagen/ultrastructure , Femur/anatomy & histology , Humans , Humerus/anatomy & histology , Microscopy, Electron, Scanning , Middle Aged , Osteocytes/ultrastructure , Reference Values , Tibia/anatomy & histology
20.
J Am Vet Med Assoc ; 189(5): 547-9, 1986 Sep 01.
Article in English | MEDLINE | ID: mdl-3759625

ABSTRACT

Fifty-six heifers were weaned from dams that were card-test positive for brucellosis. Forty-four dams were positive by rivanol and complement-fixation tests and Brucella abortus field strain was isolated from 14. Numbers of expected pregnancies following natural breeding and numbers of viable calves produced were not reduced in the heifers. Persistent B abortus infection was documented in 2 of 37 parturient heifers from reactor dams. The frequency of infection was 1 of 10 in strain 19-vaccinated heifers, and 1 of 27 in nonvaccinated heifers. The 2 persistently infected heifers had atypical serologic reaction patterns before normal parturitions.


Subject(s)
Brucellosis, Bovine/immunology , Agglutination Tests/veterinary , Animals , Brucellosis, Bovine/transmission , Cattle , Complement Fixation Tests/veterinary , Female
SELECTION OF CITATIONS
SEARCH DETAIL
...