Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neuroscience ; 178: 196-207, 2011 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-21277945

RESUMO

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.


Assuntos
Osso e Ossos/inervação , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Amielínicas/metabolismo , Receptor trkA/biossíntese , Células Receptoras Sensoriais/metabolismo , Animais , Medula Óssea/inervação , Osso e Ossos/citologia , Peptídeo Relacionado com Gene de Calcitonina/biossíntese , Cartilagem/inervação , Fêmur/citologia , Fêmur/inervação , Camundongos , Camundongos Endogâmicos C3H , Proteínas de Neurofilamentos/biossíntese , Periósteo/inervação , Células Receptoras Sensoriais/citologia
2.
Neuroscience ; 171(2): 588-98, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20851743

RESUMO

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.


Assuntos
Neoplasias Ósseas/fisiopatologia , Fibras Nervosas/patologia , Fator de Crescimento Neural/antagonistas & inibidores , Neuroma/prevenção & controle , Dor/prevenção & controle , Animais , Anticorpos/farmacologia , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Camundongos , Transplante de Neoplasias , Fibras Nervosas/efeitos dos fármacos , Fator de Crescimento Neural/imunologia , Neuroma/patologia , Dor/patologia , Dor/fisiopatologia
3.
Neuroscience ; 162(4): 1244-54, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19486928

RESUMO

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.


Assuntos
Capsaicina/farmacologia , Fraturas do Fêmur/fisiopatologia , Fibras Nervosas/fisiologia , Dor/fisiopatologia , Células Receptoras Sensoriais/fisiologia , Animais , Animais Recém-Nascidos , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Fraturas do Fêmur/complicações , Masculino , Fibras Nervosas/efeitos dos fármacos , Proteínas de Neurofilamentos/metabolismo , Dor/etiologia , Periósteo/metabolismo , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/efeitos dos fármacos
4.
Neuroscience ; 137(4): 1417-26, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16388907

RESUMO

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.


Assuntos
Neurônios Aferentes/fisiologia , Pâncreas/inervação , Sistema Nervoso Simpático/fisiologia , Animais , Peptídeo Relacionado com Gene de Calcitonina/análise , Duodeno/anatomia & histologia , Duodeno/inervação , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/fisiologia , Fibras Nervosas/fisiologia , Fibras Nervosas/ultraestrutura , Pâncreas/anatomia & histologia
5.
Neuroscience ; 126(4): 1043-52, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15207337

RESUMO

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.


Assuntos
Neoplasias Ósseas/metabolismo , Endotelina-1/fisiologia , Dor/metabolismo , Sarcoma/metabolismo , Análise de Variância , Animais , Atrasentana , Comportamento Animal , Neoplasias Ósseas/complicações , Neoplasias Ósseas/tratamento farmacológico , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Modelos Animais de Doenças , Dinorfinas/metabolismo , Antagonistas dos Receptores de Endotelina , Endotelina-1/sangue , Gânglios Espinais/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica/métodos , Masculino , Camundongos , Camundongos Endogâmicos , Dor/tratamento farmacológico , Dor/etiologia , Medição da Dor/efeitos dos fármacos , Pirrolidinas/uso terapêutico , Receptores de Endotelina/metabolismo , Sarcoma/complicações , Sarcoma/tratamento farmacológico , Nervo Isquiático/metabolismo , Fatores de Tempo
7.
J Struct Biol ; 130(2-3): 174-83, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10940224

RESUMO

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.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Doença de Alzheimer/etiologia , Substituição de Aminoácidos , Peptídeos beta-Amiloides/metabolismo , Sequência de Bases , Encéfalo/metabolismo , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Peptídeos/síntese química , Peptídeos/química , Placa Amiloide/química , Placa Amiloide/metabolismo , Conformação Proteica , Temperatura , Termodinâmica
8.
Biochemistry ; 39(21): 6288-95, 2000 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-10828941

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Amiloidose/patologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Substituição de Aminoácidos , Cromatografia Líquida de Alta Pressão , Humanos , Cinética , Modelos Químicos , Placa Amiloide/patologia , Conformação Proteica , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
9.
Brain Res ; 815(1): 1-10, 1999 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-9974116

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Comportamento Animal/efeitos dos fármacos , Hipocampo/fisiopatologia , Fragmentos de Peptídeos/farmacologia , Animais , Química Encefálica/fisiologia , Condicionamento Psicológico/efeitos dos fármacos , Masculino , Microinjeções , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
10.
J Neurosci ; 14(6): 3958-68, 1994 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8207499

RESUMO

Neuropeptide Y (NPY) has been suggested to exert antinociceptive actions by inhibiting the release of neurotransmitters from trigeminal and dorsal root ganglia (DRG) neurons, but the site of direct NPY action in vivo and the NPY receptor subtype mediating these effects are unknown. 125I-peptide YY (PYY) was used to localize and characterize NPY receptor binding sites in trigeminal ganglia, DRG, and spinal cord of the rat, rabbit, and monkey. In the rat, rabbit, and monkey, 5-20% of trigeminal ganglia and DRG neurons express NPY binding sites. Unilateral cuff-induced neuropathy or transection of the rat sciatic nerve did not significantly alter the density or number of DRG neurons expressing NPY receptors. A unimodal size distribution for L4 and L5 DRG neurons expressing NPY binding sites in the rat was determined, with a mean cross-sectional area of 947 microns 2. In the spinal cord the highest concentration of NPY receptors is found in laminae I, II, V, X, and Onuf's nucleus. Pharmacological experiments using selective Y1 and Y2 receptor antagonists suggest that Y2 is the prominent NPY receptor subtype expressed in trigeminal ganglia neurons, DRG neurons, and spinal cord. Previous studies have demonstrated that a population of large-diameter, presumably myelinated primary afferents express NPY after peripheral nerve injury. NPY released from these injured large-diameter DRG neurons may act in a paracrine fashion to block the transmission of nociceptive information from the small- and medium-diameter DRG neurons that constitutively express NPY receptors. NPY receptors are therefore uniquely positioned to inhibit primary afferent nociceptors directly, especially after peripheral nerve injury.


Assuntos
Inibição Neural/fisiologia , Neurônios Aferentes/metabolismo , Nociceptores/fisiologia , Traumatismos dos Nervos Periféricos , Receptores de Neuropeptídeo Y/metabolismo , Animais , Autorradiografia , Sítios de Ligação , Denervação , Gânglios Espinais/metabolismo , Macaca nemestrina , Masculino , Peptídeo YY , Peptídeos/metabolismo , Coelhos , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Medula Espinal/metabolismo , Gânglio Trigeminal/metabolismo
11.
J Neurochem ; 61(3): 1171-4, 1993 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8360682

RESUMO

A major pathological feature of Alzheimer's disease (AD) is the presence of a high density of amyloid plaques in the brain tissue of patients. The plaques are predominantly composed of human beta-amyloid peptide beta A4, a 40-mer whose neurotoxicity is related to its aggregation. Certain metals have been proposed as risk factors for AD, but the mechanism by which the metals may exert their effects is unclear. Radioiodinated human beta A4 has been used to assess the effects of various metals on the aggregation of the peptide in dilute solution (10(-10) M). In physiological buffers, 10(-3) M calcium, cobalt, copper, manganese, magnesium, sodium, or potassium had no effect on the rate of beta A4 aggregation. In sharp contrast, aluminum, iron, and zinc under the same conditions strongly promoted aggregation (rate enhancement of 100-1,000-fold). The aggregation of beta A4 induced by aluminum and iron is distinguishable from that induced by zinc in terms of rate, extent, pH and temperature dependence. These results suggest that high concentrations of certain metals may play a role in the pathogenesis of AD by promoting aggregation of beta A4.


Assuntos
Alumínio/farmacologia , Peptídeos beta-Amiloides/fisiologia , Ferro/farmacologia , Zinco/farmacologia , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Humanos , Concentração de Íons de Hidrogênio , Íons , Concentração Osmolar
12.
Neuroscience ; 53(3): 889-98, 1993 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7683778

RESUMO

[125I]Peptide YY was used to localize and characterize peptide YY and neuropeptide Y receptor binding sites in the heart. In the rat and rabbit heart, nearly every artery and arteriole that could be histologically identified also expressed saturable binding sites for [125I]peptide YY. In the arteries, these [125I]peptide YY binding sites were primarily associated with the smooth muscle layer. Pharmacological experiments demonstrated that peptide YY and neuropeptide Y were equipotent in competing for [125I]peptide YY binding in the heart. In another competition series, [Leu31,Pro34]-neuropeptide Y (a Y1 receptor-specific agonist when used with [125I]peptide YY) was significantly more potent than neuropeptide Y (a Y2 receptor-specific agonist when used with [125I]peptide YY) in competing for [125I]peptide YY binding from coronary arteries, suggesting that the receptor binding sites on cardiac arteries and arterioles are of the Y1 subtype. These results demonstrate that smooth muscle cells of the atrial and ventricular arteries and arterioles in rat and rabbit heart express Y1 receptors and suggest a possible direct effect of neuropeptide Y on coronary blood vessels to induce vasoconstriction.


Assuntos
Hormônios Gastrointestinais/metabolismo , Miocárdio/metabolismo , Peptídeos/metabolismo , Receptores de Neuropeptídeo Y/metabolismo , Animais , Autorradiografia , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/metabolismo , Coração/efeitos dos fármacos , Processamento de Imagem Assistida por Computador , Radioisótopos do Iodo , Masculino , Peptídeo YY , Coelhos , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptores de Neuropeptídeo Y/efeitos dos fármacos , Coloração e Rotulagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA