Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Neuroscience ; 178: 196-207, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21277945

ABSTRACT

Although skeletal pain is a leading cause of chronic pain and disability, relatively little is known about the specific populations of nerve fibers that innervate the skeleton. Recent studies have reported that therapies blocking nerve growth factor (NGF) or its cognate receptor, tropomyosin receptor kinase A (TrkA) are efficacious in attenuating skeletal pain. A potential factor to consider when assessing the analgesic efficacy of targeting NGF-TrkA signaling in a pain state is the fraction of NGF-responsive TrkA+ nociceptors that innervate the tissue from which the pain is arising, as this innervation and the analgesic efficacy of targeting NGF-TrkA signaling may vary considerably from tissue to tissue. To explore this in the skeleton, tissue slices and whole mount preparations of the normal, adult mouse femur were analyzed using immunohistochemistry and confocal microscopy. Analysis of these preparations revealed that 80% of the unmyelinated/thinly myelinated sensory nerve fibers that express calcitonin gene-related peptide (CGRP) and innervate the periosteum, mineralized bone and bone marrow also express TrkA. Similarly, the majority of myelinated sensory nerve fibers that express neurofilament 200 kDa (NF200) which innervate the periosteum, mineralized bone and bone marrow also co-express TrkA. In the normal femur, the relative density of CGRP+, NF200+ and TrkA+ sensory nerve fibers per unit volume is: periosteum>bone marrow>mineralized bone>cartilage with the respective relative densities being 100:2:0.1:0. The observation that the majority of sensory nerve fibers innervating the skeleton express TrkA+, may in part explain why therapies that block NGF/TrkA pathway are highly efficacious in attenuating skeletal pain.


Subject(s)
Bone and Bones/innervation , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Unmyelinated/metabolism , Receptor, trkA/biosynthesis , Sensory Receptor Cells/metabolism , Animals , Bone Marrow/innervation , Bone and Bones/cytology , Calcitonin Gene-Related Peptide/biosynthesis , Cartilage/innervation , Femur/cytology , Femur/innervation , Mice , Mice, Inbred C3H , Neurofilament Proteins/biosynthesis , Periosteum/innervation , Sensory Receptor Cells/cytology
2.
Neuroscience ; 171(2): 588-98, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20851743

ABSTRACT

For many patients, pain is the first sign of cancer and, while pain can be present at any time, the frequency and intensity of pain tend to increase with advancing stages of the disease. Thus, between 75 and 90% of patients with metastatic or advanced-stage cancer will experience significant cancer-induced pain. One major unanswered question is why cancer pain increases and frequently becomes more difficult to fully control with disease progression. To gain insight into this question we used a mouse model of bone cancer pain to demonstrate that as tumor growth progresses within bone, tropomyosin receptor kinase A (TrkA)-expressing sensory and sympathetic nerve fibers undergo profuse sprouting and form neuroma-like structures. To address what is driving the pathological nerve reorganization we administered an antibody to nerve growth factor (anti-NGF). Early sustained administration of anti-NGF, whose cognate receptor is TrkA, blocks the pathological sprouting of sensory and sympathetic nerve fibers, the formation of neuroma-like structures, and inhibits the development of cancer pain. These results suggest that cancer cells and their associated stromal cells release nerve growth factor (NGF), which induces a pathological remodeling of sensory and sympathetic nerve fibers. This pathological remodeling of the peripheral nervous system then participates in driving cancer pain. Similar to therapies that target the cancer itself, the data presented here suggest that, the earlier therapies blocking this pathological nerve remodeling are initiated, the more effective the control of cancer pain.


Subject(s)
Bone Neoplasms/physiopathology , Nerve Fibers/pathology , Nerve Growth Factor/antagonists & inhibitors , Neuroma/prevention & control , Pain/prevention & control , Animals , Antibodies/pharmacology , Bone Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Female , Mice , Neoplasm Transplantation , Nerve Fibers/drug effects , Nerve Growth Factor/immunology , Neuroma/pathology , Pain/pathology , Pain/physiopathology
3.
Neuroscience ; 162(4): 1244-54, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19486928

ABSTRACT

Although skeletal pain can have a marked impact on a patient's functional status and quality of life, relatively little is known about the specific populations of peripheral nerve fibers that drive non-malignant bone pain. In the present report, neonatal male Sprague-Dawley rats were treated with capsaicin or vehicle and femoral fracture was produced when the animals were young adults (15-16 weeks old). Capsaicin treatment, but not vehicle, resulted in a significant (>70%) depletion in the density of calcitonin-gene related peptide positive (CGRP(+)) sensory nerve fibers, but not 200 kDa neurofilament H positive (NF200(+)) sensory nerve fibers in the periosteum. The periosteum is a thin, cellular and fibrous tissue that tightly adheres to the outer surface of all but the articulated surface of bone and appears to play a pivotal role in driving fracture pain. In animals treated with capsaicin, but not vehicle, there was a 50% reduction in the severity, but no change in the time course, of fracture-induced skeletal pain-related behaviors as measured by spontaneous flinching, guarding and weight bearing. These results suggest that both capsaicin-sensitive (primarily CGRP(+) C-fibers) and capsaicin-insensitive (primarily NF200(+) A-delta fibers) sensory nerve fibers participate in driving skeletal fracture pain. Skeletal pain can be a significant impediment to functional recovery following trauma-induced fracture, osteoporosis-induced fracture and orthopedic surgery procedures such as knee and hip replacement. Understanding the specific populations of sensory nerve fibers that need to be targeted to inhibit the generation and maintenance of skeletal pain may allow the development of more specific mechanism-based therapies that can effectively attenuate acute and chronic skeletal pain.


Subject(s)
Capsaicin/pharmacology , Femoral Fractures/physiopathology , Nerve Fibers/physiology , Pain/physiopathology , Sensory Receptor Cells/physiology , Animals , Animals, Newborn , Calcitonin Gene-Related Peptide/metabolism , Femoral Fractures/complications , Male , Nerve Fibers/drug effects , Neurofilament Proteins/metabolism , Pain/etiology , Periosteum/metabolism , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects
4.
Neuroscience ; 137(4): 1417-26, 2006.
Article in English | MEDLINE | ID: mdl-16388907

ABSTRACT

Pain from pancreatitis or pancreatic cancer can be both chronic and severe although little is known about the mechanisms that generate and maintain this pain. To define the peripheral sensory and sympathetic fibers involved in transmitting and modulating pancreatic pain, immunohistochemistry and confocal microscopy were used to examine the sensory and sympathetic innervation of the head, body and tail of the normal mouse pancreas. Myelinated sensory fibers were labeled with an antibody raised against 200 kD neurofilament H (clone RT97), thinly myelinated and unmyelinated peptidergic sensory fibers were labeled with antibodies raised against calcitonin gene-related peptide (CGRP) and post-ganglionic sympathetic fibers were labeled with an antibody raised against tyrosine hydroxylase (TH). RT97, CGRP, and TH immunoreactive fibers were present in parenchyma of the head, body and tail of the pancreas with the relative density of both RT97 and CGRP expressing fibers being head>body>tail, whereas for TH, a relatively even distribution was observed. In all three regions of the pancreas, RT97 fibers were associated mainly with large blood vessels, the CGRP fibers were associated with the large- and medium-sized blood vessels and the TH were associated with the large- and medium-sized blood vessels as well as capillaries. In addition to this extensive set of sensory and sympathetic nerve fibers that terminate in the pancreas, there were large bundles of en passant nerve fibers in the dorsal region of the pancreas that expressed RT97 or CGRP and were associated with the superior mesenteric plexus. These data suggest the pancreas receives a significant sensory and sympathetic innervation. Understanding the factors and disease states that sensitize and/or directly excite the nerve fibers that terminate in the pancreas as well as those that are en passant may aid in the development of therapies that more effectively modulate the pain that frequently accompanies diseases of the pancreas, such as pancreatitis and pancreatic cancer.


Subject(s)
Neurons, Afferent/physiology , Pancreas/innervation , Sympathetic Nervous System/physiology , Animals , Calcitonin Gene-Related Peptide/analysis , Duodenum/anatomy & histology , Duodenum/innervation , Female , Mice , Mice, Inbred C57BL , Myelin Sheath/physiology , Nerve Fibers/physiology , Nerve Fibers/ultrastructure , Pancreas/anatomy & histology
5.
Neuroscience ; 126(4): 1043-52, 2004.
Article in English | MEDLINE | ID: mdl-15207337

ABSTRACT

Tumors including sarcomas and breast, prostate, and lung carcinomas frequently grow in or metastasize to the skeleton where they can induce significant bone remodeling and cancer pain. To define products that are released from tumors that are involved in the generation and maintenance of bone cancer pain, we focus here on endothelin-1 (ET-1) and endothelin receptors as several tumors including human prostate and breast have been shown to express high levels of ETs and the application of ETs to peripheral nerves can induce pain. Here we show that in a murine osteolytic 2472 sarcoma model of bone cancer pain, the 2472 sarcoma cells express high levels of ET-1, but express low or undetectable levels of endothelin A (ETAR) or B (ETBR) receptors whereas a subpopulation of sensory neurons express the ETAR and non-myelinating Schwann cells express the ETBR. Acute (10 mg/kg, i.p.) or chronic (10 mg/kg/day, p.o.) administration of the ETAR selective antagonist ABT-627 significantly attenuated ongoing and movement-evoked bone cancer pain and chronic administration of ABT-627 reduced several neurochemical indices of peripheral and central sensitization without influencing tumor growth or bone destruction. In contrast, acute treatment (30 mg/kg, i.p.) with the ETBR selective antagonist, A-192621 increased several measures of ongoing and movement evoked pain. As tumor expression and release of ET-1 has been shown to be regulated by the local environment, location specific expression and release of ET-1 by tumor cells may provide insight into the mechanisms that underlie the heterogeneity of bone cancer pain that is frequently observed in humans with multiple skeletal metastases.


Subject(s)
Bone Neoplasms/metabolism , Endothelin-1/physiology , Pain/metabolism , Sarcoma/metabolism , Analysis of Variance , Animals , Atrasentan , Behavior, Animal , Bone Neoplasms/complications , Bone Neoplasms/drug therapy , Calcitonin Gene-Related Peptide/metabolism , Disease Models, Animal , Dynorphins/metabolism , Endothelin Receptor Antagonists , Endothelin-1/blood , Ganglia, Spinal/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry/methods , Male , Mice , Mice, Inbred Strains , Pain/drug therapy , Pain/etiology , Pain Measurement/drug effects , Pyrrolidines/therapeutic use , Receptors, Endothelin/metabolism , Sarcoma/complications , Sarcoma/drug therapy , Sciatic Nerve/metabolism , Time Factors
7.
J Neurosci ; 21(3): 999-1006, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11157085

ABSTRACT

The endothelins (ETs) are peptides that have a diverse array of functions mediated by two receptor subtypes, the endothelin A receptor (ET(A)R) and the endothelin B receptor (ET(B)R). Pharmacological studies have suggested that in peripheral tissues, ET(A)R expression may play a role in signaling acute or neuropathic pain, whereas ET(B)R expression may be involved in the transmission of chronic inflammatory pain. To begin to define the mechanisms by which ET can drive nociceptive signaling, autoradiography and immunohistochemistry were used to examine the distribution of ET(A)R and ET(B)R in dorsal root ganglia (DRG) and peripheral nerve of the rat, rabbit, and monkey. In DRG and peripheral nerve, ET(A)R-immunoreactivity was present in a subset of small-sized peptidergic and nonpeptidergic sensory neurons and their axons and to a lesser extent in a subset of medium-sized sensory neurons. However, ET(B)R-immunoreactivity was not seen in DRG neurons or axons but rather in DRG satellite cells and nonmyelinating ensheathing Schwann cells. Thus, when ETs are released in peripheral tissues, they could act directly on ET(A)R-expressing sensory neurons and on ET(B)R-expressing DRG satellite cells or nonmyelinating Schwann cells. These data indicate that ETs can have direct, nociceptive effects on the peripheral sensory nervous system and that peripheral glia may be directly involved in signaling nociceptive events in peripheral tissues.


Subject(s)
Neuroglia/metabolism , Pain/metabolism , Peripheral Nerves/metabolism , Receptors, Endothelin/biosynthesis , Animals , Autoradiography , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Ligation , Macaca mulatta , Male , Neuroglia/cytology , Pain/etiology , Pain Measurement , Peripheral Nerves/cytology , Peripheral Nerves/surgery , Rabbits , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A , Receptor, Endothelin B , Schwann Cells/cytology , Schwann Cells/metabolism , Sciatic Nerve/cytology , Sciatic Nerve/metabolism , Sciatic Nerve/surgery
8.
J Struct Biol ; 130(2-3): 174-83, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10940224

ABSTRACT

Brain amyloid composed of the approximately 40-amino-acid human beta-amyloid peptide A beta is integral to Alzheimer's disease pathology. To probe the importance of a conformational transition in Abeta during amyloid growth, we synthesized and examined the solution conformation and amyloid deposition activity of A beta congeners designed to have similar solution structures but to vary substantially in their barriers to conformational transition. Although all these peptides adopt similar solution conformations, a covalently restricted Abeta congener designed to have a very high barrier to conformational rearrangement was inactive, while a peptide designed to have a reduced barrier to conformational transition displayed an enhanced deposition rate relative to wild-type A beta. The hyperactive peptide, which is linked to a heritable A beta amyloidosis characterized by massive amyloid deposition at an early age, displayed a reduced activation barrier to deposition consistent with a larger difference in activation entropy than in activation enthalpy relative to wild-type A beta. These results suggest that in Alzheimer's disease, as in the prion diseases, a conformational transition in the depositing peptide is essential for the conversion of soluble monomer to insoluble amyloid, and alterations in the activation barrier to this transition affect amyloidogenicity and directly contribute to human disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Alzheimer Disease/etiology , Amino Acid Substitution , Amyloid beta-Peptides/metabolism , Base Sequence , Brain/metabolism , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemical synthesis , Peptides/chemistry , Plaque, Amyloid/chemistry , Plaque, Amyloid/metabolism , Protein Conformation , Temperature , Thermodynamics
9.
Biochemistry ; 39(21): 6288-95, 2000 May 30.
Article in English | MEDLINE | ID: mdl-10828941

ABSTRACT

Amyloid plaques composed of the peptide Abeta are an integral part of Alzheimer's disease (AD) pathogenesis. We have modeled the process of amyloid plaque growth by monitoring the deposition of soluble Abeta onto amyloid in AD brain tissue or synthetic amyloid fibrils and show that it is mediated by two distinct kinetic processes. In the first phase, "dock", Abeta addition to the amyloid template is fully reversible (dissociation t(1/2) approximately 10 min), while in the second phase, "lock", the deposited peptide becomes irreversibly associated (dissociation t(1/2) >> 1000 min) with the template in a time-dependent manner. The most recently deposited peptide dissociates first while Abeta previously deposited becomes irreversibly "locked" onto the template. Thus, the transition from monomer to neurotoxic amyloid is mediated by interaction with the template, a mechanism that has also been proposed for the prion diseases. Interestingly, two Abeta peptides bearing primary sequence alterations implicated in heritable Abeta amyloidoses displayed faster lock-phase kinetics than wild-type Abeta. Inhibiting the initial weak docking interaction between depositing Abeta and the template is a viable therapeutic target to prevent the critical conformational transition in the conversion of Abeta((solution)) to Abeta((amyloid)) and thus prevent stable amyloid accumulation. While thermodynamics suggest that inhibiting amyloid assembly would be difficult, the present study illustrates that the protein misfolding diseases are kinetically vulnerable to intervention.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloidosis/pathology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Amino Acid Substitution , Chromatography, High Pressure Liquid , Humans , Kinetics , Models, Chemical , Plaque, Amyloid/pathology , Protein Conformation , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
10.
Science ; 286(5444): 1558-61, 1999 Nov 19.
Article in English | MEDLINE | ID: mdl-10567262

ABSTRACT

Substance P receptor (SPR)-expressing spinal neurons were ablated with the selective cytotoxin substance P-saporin. Loss of these neurons resulted in a reduction of thermal hyperalgesia and mechanical allodynia associated with persistent neuropathic and inflammatory pain states. This loss appeared to be permanent. Responses to mildly painful stimuli and morphine analgesia were unaffected by this treatment. These results identify a target for treating persistent pain and suggest that the small population of SPR-expressing neurons in the dorsal horn of the spinal cord plays a pivotal role in the generation and maintenance of chronic neuropathic and inflammatory pain.


Subject(s)
Immunotoxins , N-Glycosyl Hydrolases , Pain/drug therapy , Pain/physiopathology , Plant Proteins/pharmacology , Posterior Horn Cells/physiology , Receptors, Neurokinin-1/metabolism , Substance P/pharmacology , Animals , Dose-Response Relationship, Drug , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Inflammation/physiopathology , Ligation , Neuralgia/drug therapy , Neuralgia/physiopathology , Plant Proteins/administration & dosage , Posterior Horn Cells/drug effects , Rats , Ribosome Inactivating Proteins, Type 1 , Saporins , Spinal Nerves , Substance P/administration & dosage , Time Factors
11.
Biochemistry ; 38(32): 10424-31, 1999 Aug 10.
Article in English | MEDLINE | ID: mdl-10441137

ABSTRACT

Senile plaques composed of the peptide Abeta contribute to the pathogenesis of Alzheimer's disease (AD), and mechanisms underlying their formation and growth may be exploitable as therapeutic targets. To examine the process of amyloid plaque growth in human brain, we have utilized size exclusion chromatography (SEC), translational diffusion measured by NMR, and in vitro models of Abeta amyloid growth to identify the oligomerization state of Abeta that is competent to add onto an existing amyloid deposit. SEC of radiolabeled and unlabeled Abeta over a concentration range of 10(-)(10)-10(-)(4) M demonstrated that the freshly dissolved peptide eluted as a single low molecular weight species, consistent with monomer or dimer. This low molecular weight Abeta species isolated by SEC was competent to deposit onto preexisting amyloid in preparations of AD cortex, with first-order kinetic dependence on soluble Abeta concentration, establishing that solution-phase oligomerization is not rate limiting. Translational diffusion measurements of the low molecular weight Abeta fraction demonstrate that the form of the peptide active in plaque deposition is a monomer. In deliberately aged (>6 weeks) Abeta solutions, a high molecular weight (>100 000 M(r)) species was detectable in the SEC column void. In contrast to the active monomer, assembled Abeta isolated from the column showed little or no focal association with AD tissue. These studies establish that, at least in vitro, Abeta exists as a monomer at physiological concentrations and that deposition of monomers, rather than of oligomeric Abeta assemblies, mediates the growth of existing amyloid in human brain preparations.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Brain/pathology , Plaque, Amyloid/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/isolation & purification , Amyloid beta-Peptides/physiology , Cell Division , Chromatography, Gel , Dimerization , Humans , Kinetics , Molecular Sequence Data , Molecular Weight , Peptide Fragments/isolation & purification , Plaque, Amyloid/pathology , Protein Biosynthesis , Solutions
12.
Biopolymers ; 49(6): 505-14, 1999 May.
Article in English | MEDLINE | ID: mdl-10193196

ABSTRACT

The formation and growth of insoluble amyloid deposits composed primarily of the human beta-amyloid peptide (A beta) in brain is an essentially invariant feature of Alzheimer's disease (AD) and is widely believed to contribute to the progressive neurodegeneration of the disorder. To probe the specificity of amyloid formation and growth, we synthesized and examined the self-assembly of D- and L-stereoisomers of A beta in vitro. While both enantiomers formed insoluble aggregates at similar rates with amyloid-like fibrillar morphology, deposition of soluble A beta peptide onto preexisting A beta aggregates was stereospecific. Although the L-peptide deposited readily onto immobilized L-A beta aggregates with first-order kinetic dependence on soluble peptide concentration, essentially no association between the D-peptide and L-template was observed. Similarly, the D-peptide deposited with first-order kinetics onto a D-A beta aggregate template but did not deposit onto a similar template composed of aggregates of the L-enantiomer. Furthermore, although the L-A beta isomer deposited onto authentic AD amyloid in preparations of unfixed AD brain, no focal association between the D-peptide and brain amyloid was detected. These results establish that deposition of soluble A beta onto preexisting amyloid template is stereospecific, likely involving direct docking interactions between peptide backbone and/or side chains rather than simple hydrophobic association.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/ultrastructure , Biopolymers/chemistry , Biopolymers/metabolism , Humans , In Vitro Techniques , Kinetics , Microscopy, Electron , Stereoisomerism
13.
Brain Res ; 815(1): 1-10, 1999 Jan 02.
Article in English | MEDLINE | ID: mdl-9974116

ABSTRACT

Beta amyloid protein (A beta) is the major extracellular component of Alzheimer's disease (AD) plaques. In the current study, A beta (1-42) was aggregated in vitro using a method which produces A beta aggregates similar to those found in the AD brain. Twelve male Sprague-Dawley rats were trained in two-lever operant chambers under an alternating lever cyclic-ratio (ALCR) schedule. When performance was stable on the ALCR schedule, six subjects were injected (bilaterally into the CA3 area of the dorsal hippocampus) with 5.0 microliters aggregated A beta in suspension, and the remaining six subjects were injected with 5.0 microliters sterile water. Behavioral testing resumed 5 days after surgery and continued for 90 days post-injection. Aggregated A beta injection did not affect the number of lever switching errors made in a daily session but did affect the number of incorrect lever response perseverations. After approximately 30 days post-injection, aggregated A beta injection detrimentally affected ability to track the changing parameters of the schedule, and decreased the efficiency by which subjects obtained reinforcers. From approximately day 50 post-injection onward, A beta-injected subjects demonstrated significantly higher numbers of incorrect lever response perseverations than did sterile water-injected subjects. These effects appeared to be central rather than peripheral, as A beta injection did not decrease running response rates under the ALCR schedule. The delayed onset of behavioral effects seen in this and other behavioral studies may be a result of a cascade of potentially harmful responses induced through glial activation following aggregated A beta injection.


Subject(s)
Amyloid beta-Peptides/pharmacology , Behavior, Animal/drug effects , Hippocampus/physiopathology , Peptide Fragments/pharmacology , Animals , Brain Chemistry/physiology , Conditioning, Psychological/drug effects , Male , Microinjections , Rats , Rats, Sprague-Dawley , Time Factors
14.
J Neurosci ; 18(6): 2161-73, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9482801

ABSTRACT

To determine the stability of beta-amyloid peptide (Abeta) and the glial and neuronal changes induced by Abeta in the CNS in vivo, we made single injections of fibrillar Abeta (fAbeta), soluble Abeta (sAbeta), or vehicle into the rat striatum. Injected fAbeta is stable in vivo for at least 30 d after injection, whereas sAbeta is primarily cleared within 1 d. After injection of fAbeta, microglia phagocytize fAbeta aggregates, whereas nearby astrocytes form a virtual wall between fAbeta-containing microglia and the surrounding neuropil. Similar glial changes are not observed after sAbeta injection. Microglia and astrocytes near the injected fAbeta show a significant increase in inducible nitric oxide synthase (iNOS) expression compared with that seen with sAbeta or vehicle injection. Injection of fAbeta but not sAbeta or vehicle induces a significant loss of parvalbumin- and neuronal nitric oxide synthase-immunoreactive neurons, whereas the number of calbindin-immunoreactive neurons remains unchanged. These data demonstrate that fAbeta is remarkably stable in the CNS in vivo and suggest that fAbeta neurotoxicity is mediated in large part by factors released from activated microglia and astrocytes, as opposed to direct interaction between Abeta fibrils and neurons.


Subject(s)
Amyloid beta-Peptides/pharmacology , Brain/drug effects , Brain/enzymology , Microglia/drug effects , Nitric Oxide Synthase/metabolism , Phagocytosis/drug effects , Amyloid beta-Peptides/chemistry , Animals , Astrocytes/drug effects , Astrocytes/enzymology , Brain/cytology , Cell Count/drug effects , Drug Stability , Enzyme Induction/drug effects , Injections , Male , Microglia/enzymology , Microglia/physiology , Neurons/drug effects , Neurons/enzymology , Nitric Oxide Synthase Type II , Rats , Rats, Sprague-Dawley , Solubility
15.
Science ; 278(5336): 275-9, 1997 Oct 10.
Article in English | MEDLINE | ID: mdl-9323204

ABSTRACT

Substance P is released in the spinal cord in response to painful stimuli, but its role in nociceptive signaling remains unclear. When a conjugate of substance P and the ribosome-inactivating protein saporin was infused into the spinal cord, it was internalized and cytotoxic to lamina I spinal cord neurons that express the substance P receptor. This treatment left responses to mild noxious stimuli unchanged, but markedly attenuated responses to highly noxious stimuli and mechanical and thermal hyperalgesia. Thus, lamina I spinal cord neurons that express the substance P receptor play a pivotal role in the transmission of highly noxious stimuli and the maintenance of hyperalgesia.


Subject(s)
Hyperalgesia/therapy , Immunotoxins , N-Glycosyl Hydrolases , Neurons/metabolism , Pain Management , Receptors, Neurokinin-1/metabolism , Spinal Cord/cytology , Substance P/metabolism , Animals , Capsaicin , Cell Membrane/metabolism , Cells, Cultured , Fluorescent Antibody Technique , Hyperalgesia/physiopathology , Injections, Spinal , Neurons/cytology , Pain/physiopathology , Pain Measurement , Plant Proteins/metabolism , Plant Proteins/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/biosynthesis , Ribosome Inactivating Proteins, Type 1 , Saporins , Signal Transduction , Spinal Cord/metabolism , Substance P/pharmacology
16.
J Neurosci ; 17(15): 5921-7, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9221788

ABSTRACT

Dorsal root ganglia (DRG) neurons synthesize and transport substance P (SP) to the spinal cord where it is released in response to intense noxious somatosensory stimuli. We have shown previously that SP release in vivo causes a rapid and reversible internalization of SP receptors (SPRs) in dorsal horn neurons, which may provide a pharmacologically specific image of neurons activated by SP. Here, we report that noxious heat (43 degrees, 48 degrees, and 55 degrees C) and cold (10 degrees, 0 degrees, -10 degrees, and -20 degrees C) stimuli, but not innocuous warm (38 degrees C) and cold (20 degrees C) stimuli, applied to the hindpaw of anesthetized rats induce SPR internalization in spinal cord neurons that is graded with respect to the intensity of the thermal stimulus. Thus, with increasing stimulus intensities, both the total number of SPR+ lamina I neurons showing SPR internalization and the number of internalized SPR+ endosomes within each SPR immunoreactive neuron showed a significant increase. These data suggest that thermal stimuli induce a graded release of SP from primary afferent terminals and that agonist dependent receptor endocytosis provides evidence of a spatially and pharmacologically unique "neurochemical signature" after specific somatosensory stimuli.


Subject(s)
Neurons, Afferent/physiology , Spinal Cord/metabolism , Substance P/metabolism , Animals , Male , Microscopy, Confocal , Pain Measurement , Rats , Rats, Sprague-Dawley , Spinal Cord/ultrastructure
17.
Exp Neurol ; 145(1): 180-95, 1997 May.
Article in English | MEDLINE | ID: mdl-9184120

ABSTRACT

Previous studies have demonstrated that neonatal cultures of astrocytes express functional endothelin (ET) receptors. To determine if similar ET receptors are expressed by adult glia we used 125I-ET-1 to examine the expression of ET receptors both in vivo in the normal and transected optic nerves of the rabbit and rat and in vitro in cultures of astrocytes, microglia, or oligodendrocytes. Additionally, we examined the expression of ET receptors in the human optic nerve. Moderate levels of ET(B) receptors were identified in the rabbit and rat forebrain, whereas in the normal rabbit, rat, and human optic nerves a low density of ET(B) receptors was observed, mainly in association with glial fibrillary acidic protein + (GFAP+) astrocytes. After unilateral optic nerve transection, or damage to the retina, the density of glial ET(B) receptors in the optic nerve is significantly increased in all species examined. Thus, at 7 days posttransection there is a significant increase in ET(B) receptors, and by 90 days posttransection the density of ET(B) receptors in the rabbit or rat optic nerve was among the highest of any area in the central nervous system (CNS). Primary cultures of astrocytes or microglia, but not oligodendrocytes, express 125I-ET-1 binding sites. These data demonstrate that in the normal CNS, astrocytes express low but detectable levels of ET(B) receptors, and, after CNS injury, both astrocytes and microglia express high levels of ET(B) receptors. ET(B) receptors provide a therapeutic target for regulating glial proliferation and the release of neurotrophic factors from glia that occur in response to neuronal injury.


Subject(s)
Neuroglia/chemistry , Optic Nerve/surgery , Receptors, Endothelin/biosynthesis , Aged , Aged, 80 and over , Animals , Astrocytes/chemistry , Astrocytes/cytology , Astrocytes/metabolism , Autoradiography , Axons/chemistry , Cells, Cultured , Denervation , Female , Humans , Immunohistochemistry , Male , Microglia/chemistry , Microglia/cytology , Microglia/metabolism , Middle Aged , Neuroglia/cytology , Neuroglia/metabolism , Oligodendroglia/chemistry , Oligodendroglia/metabolism , Optic Nerve/chemistry , Optic Nerve/cytology , Rabbits , Rats , Rats, Sprague-Dawley , Receptor, Endothelin B , Receptors, Endothelin/analysis , Retinal Ganglion Cells/chemistry , Retinal Ganglion Cells/ultrastructure
18.
Nat Biotechnol ; 15(3): 258-63, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9062926

ABSTRACT

The formation, growth, and maturation of brain amyloid "senile" plaques are essential pathological processes in Alzheimer's disease (AD) and key targets for therapeutic intervention. The process of in vitro deposition of A beta at physiological concentrations onto plaques in AD brain preparations has been well characterized, but is cumbersome for drug discovery. We describe here a high-through put screen for inhibitors of A beta deposition onto a synthetic template (synthaloid) of fibrillar A beta immobilized in a polymer matrix. Synthaloid is indistinguishable from plaques in AD brain (the natural template) in deposition kinetics, pH profile, and structure-activity relationships for both A beta analogs and inhibitors. Synthaloid, in contrast to current A beta aggregation screens, accurately predicted inhibitor potency for A beta deposition onto AD cortex preparations, validating its use in searching for agents that can slow the progression of AD and exposing a previously inaccessible target for drug discovery.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid/pharmacology , Brain/metabolism , Drug Design , Amyloid/pharmacokinetics , Amyloid beta-Peptides/metabolism , Humans , Hydrogen-Ion Concentration , Membranes, Artificial , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology
19.
Neuroreport ; 7(15-17): 2607-11, 1996 Nov 04.
Article in English | MEDLINE | ID: mdl-8981432

ABSTRACT

Alzheimer's disease is characterized by extracellular amyloid deposits in the brain at both vascular sites (cerebrovascular amyloid, CVA) and within the neuropil (plaques). In the present study we demonstrated that brain amyloid of aged non-human primates is efficiently detected by [125I]A beta in vitro, and assessed the detection of that amyloid in vivo by intravascular infusion of [125I]A beta. Aged squirrel monkeys (Saimiri sciureus) were anesthetized and infused intra-arterially with [125I]A beta, and sacrificed 2 h later. Analysis of the anterior frontal and temporal cortices by autoradiography demonstrated that [125I]A beta was deposited on CVA and that essentially every amyloid deposit which could be detected with thioflavin S or anti-A beta antibodies was also labeled by [125I]A beta. These experiments suggest that intravascular infusion of radiolabeled A beta can be used to detect and image amyloid deposits in the human AD brain.


Subject(s)
Aging/metabolism , Amyloid beta-Protein Precursor/metabolism , Frontal Lobe/metabolism , Animals , Autoradiography , Female , Humans , Male , Saimiri
20.
Gastroenterology ; 111(5): 1272-80, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8898641

ABSTRACT

BACKGROUND & AIMS: Nerves have been suggested to mediate the effects of bacterial toxins in intestinal diseases. However, the mechanisms involved are unknown. This study examined endogenous substance P (SP) activation of the substance P receptor (SPR) on enteric neurons in the rat ileum after exposure to intraluminal Clostridium difficile toxin A. METHODS: After intraluminal injection of toxin A in ileal loops, tissue was examined for pathological changes by histology and for SPR activation by immunocytochemical analysis of SP-induced SPR endocytosis. RESULTS: After toxin A administration, > 70% of enteric neurons showed SPR endocytosis and became swollen with thickened dendrites. In contrast, SPRs in control rats were largely confined to the plasma membrane. Rats denervated of primary afferent fibers with neonatal capsaicin injection and animals pretreated with a nonpeptide SPR antagonist showed few endosomal SPRs, and the pathological inflammatory effects of toxin A were ablated. CONCLUSIONS: Intraluminal toxin A causes the release of SP from primary afferent neurons: this endogenous SP then acts on enteric neurons in the submucosal and myenteric plexuses. SP is the primary mediator of an axon reflex mediating neurogenic inflammation in the intestine. SPR blockade may prove to be a novel therapy used to prevent intestinal inflammation.


Subject(s)
Bacterial Toxins , Clostridioides difficile/pathogenicity , Enterotoxins/toxicity , Ileum/drug effects , Neurons/drug effects , Receptors, Neurokinin-1/drug effects , Substance P/physiology , Animals , Capsaicin/pharmacology , Female , Ileum/chemistry , Ileum/innervation , Male , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/analysis , Receptors, Neurokinin-1/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...