Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Br J Clin Pharmacol ; 85(6): 1103-1113, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30357885

RESUMO

Disorders of the skeleton are frequently accompanied by bone pain and a decline in the functional status of the patient. Bone pain occurs following a variety of injuries and diseases including bone fracture, osteoarthritis, low back pain, orthopedic surgery, fibrous dysplasia, rare bone diseases, sickle cell disease and bone cancer. In the past 2 decades, significant progress has been made in understanding the unique population of sensory and sympathetic nerves that innervate bone and the mechanisms that drive bone pain. Following physical injury of bone, mechanotranducers expressed by sensory nerve fibres that innervate bone are activated and sensitized so that even normally non-noxious loading or movement of bone is now being perceived as noxious. Injury of the bone also causes release of factors that; directly excite and sensitize sensory nerve fibres, upregulate proalgesic neurotransmitters, receptors and ion channels expressed by sensory neurons, induce ectopic sprouting of sensory and sympathetic nerve fibres resulting in a hyper-innervation of bone, and central sensitization in the brain that amplifies pain. Many of these mechanisms appear to be involved in driving both nonmalignant and malignant bone pain. Results from human clinical trials suggest that mechanism-based therapies that attenuate one type of bone pain are often effective in attenuating pain in other seemingly unrelated bone diseases. Understanding the specific mechanisms that drive bone pain in different diseases and developing mechanism-based therapies to control this pain has the potential to fundamentally change the quality of life and functional status of patients suffering from bone pain.


Assuntos
Osso e Ossos/inervação , Células Quimiorreceptoras/metabolismo , Longevidade , Mecanorreceptores/metabolismo , Dor Musculoesquelética/fisiopatologia , Limiar da Dor , Sistema Nervoso Simpático/fisiopatologia , Fatores Etários , Analgésicos/uso terapêutico , Animais , Sensibilização do Sistema Nervoso Central , Humanos , Dor Musculoesquelética/tratamento farmacológico , Dor Musculoesquelética/epidemiologia , Dor Musculoesquelética/psicologia , Percepção da Dor , Limiar da Dor/efeitos dos fármacos , Qualidade de Vida , Fatores de Risco
2.
Curr Osteoporos Rep ; 16(4): 325-332, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29948820

RESUMO

PURPOSE OF REVIEW: This paper describes recent advances in understanding the mechanisms that drive fracture pain and how these findings are helping develop new therapies to treat fracture pain. RECENT FINDINGS: Immediately following fracture, mechanosensitive nerve fibers that innervate bone are mechanically distorted. This results in these nerve fibers rapidly discharging and signaling the initial sharp fracture pain to the brain. Within minutes to hours, a host of neurotransmitters, cytokines, and nerve growth factor are released by cells at the fracture site. These factors stimulate, sensitize, and induce ectopic nerve sprouting of the sensory and sympathetic nerve fibers which drive the sharp pain upon movement and the dull aching pain at rest. If rapid and effective healing of the fracture occurs, these factors return to baseline and the pain subsides, but if not, these factors can drive chronic bone pain. New mechanism-based therapies have the potential to fundamentally change the way acute and chronic fracture pain is managed.


Assuntos
Dor Aguda/fisiopatologia , Osso e Ossos/inervação , Dor Crônica/fisiopatologia , Fraturas Ósseas/fisiopatologia , Neuralgia/fisiopatologia , Dor Nociceptiva/fisiopatologia , Dor Aguda/etiologia , Dor Aguda/terapia , Analgésicos Opioides/uso terapêutico , Animais , Sensibilização do Sistema Nervoso Central , Dor Crônica/etiologia , Dor Crônica/terapia , Modelos Animais de Doenças , Consolidação da Fratura , Fraturas Ósseas/complicações , Fraturas Ósseas/terapia , Humanos , Neuralgia/etiologia , Neuralgia/terapia , Dor Nociceptiva/etiologia , Dor Nociceptiva/terapia , Nociceptores , Manejo da Dor , Traumatismos dos Nervos Periféricos/etiologia , Traumatismos dos Nervos Periféricos/fisiopatologia , Células Receptoras Sensoriais
3.
Mol Pain ; 13: 1744806917745465, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29166838

RESUMO

Sequestration of nerve growth factor (NGF) significantly attenuates skeletal pain in both animals and humans. However, relatively little is known about the specific cell types that express NGF or its cognate receptors tropomyosin receptor kinase A (TrkA) and p75 in the intact bone and articular cartilage. In the present study, antibodies raised against NGF, TrkA, and p75 (also known as CD271) were used to explore the expression of these antigens in the non-decalcified young mouse femur. In general, all three antigens displayed a remarkably restricted expression in bone and cartilage with less than 2% of all DAPI+ cells in the femur displaying expression of any one of the three antigens. Robust NGF immunoreactivity was found in mostly CD-31- blood vessel-associated cells, a small subset of CD-31+ endothelial cells, an unidentified group of cells located at the subchondral bone/articular cartilage interface, and a few isolated, single cells in the bone marrow. In contrast, p75 and TrkA were almost exclusively expressed by nerve fibers located nearby NGF+ blood vessels. The only non-neuronal expression of either p75 or TrkA in the femur was the expression of p75 by a subset of cells located in the deep and middle zone of the articular cartilage. Understanding the factors that tightly regulate the basal level of expression in normal bone and how the expression of NGF, TrkA, and p75 change in injury, disease, and aging may provide insights into novel therapies that can reduce skeletal pain and improve skeletal health.


Assuntos
Cartilagem Articular/metabolismo , Fêmur/metabolismo , Fator de Crescimento Neural/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Receptor trkA/metabolismo , Animais , Calcificação Fisiológica , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Imuno-Histoquímica , Camundongos Endogâmicos C57BL , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
4.
J Neurosci ; 35(16): 6307-17, 2015 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-25904784

RESUMO

The mechanisms that lead to the maintenance of chronic pain states are poorly understood, but their elucidation could lead to new insights into how pain becomes chronic and how it can potentially be reversed. We investigated the role of spinal dorsal horn neurons and descending circuitry in plasticity mediating a transition to pathological pain plasticity suggesting the presence of a chronic pain state using hyperalgesic priming. We found that when dorsal horn neurokinin 1 receptor-positive neurons or descending serotonergic neurons were ablated before hyperalgesic priming, IL-6- and carrageenan-induced mechanical hypersensitivity was impaired, and subsequent prostaglandin E2 (PGE2) response was blunted. However, when these neurons were lesioned after the induction of priming, they had no effect on the PGE2 response, reflecting differential mechanisms driving plasticity in a primed state. In stark contrast, animals with a spinally applied dopaminergic lesion showed intact IL-6- and carrageenan-induced mechanical hypersensitivity, but the subsequent PGE2 injection failed to cause mechanical hypersensitivity. Moreover, ablating spinally projecting dopaminergic neurons after the resolution of the IL-6- or carrageenan-induced response also reversed the maintenance of priming as assessed through mechanical hypersensitivity and the mouse grimace scale. Pharmacological antagonism of spinal dopamine D1/D5 receptors reversed priming, whereas D1/D5 agonists induced mechanical hypersensitivity exclusively in primed mice. Strikingly, engagement of D1/D5 coupled with anisomycin in primed animals reversed a chronic pain state, consistent with reconsolidation-like effects in the spinal dorsal horn. These findings demonstrate a novel role for descending dopaminergic neurons in the maintenance of pathological pain plasticity.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Células do Corno Posterior/fisiologia , Receptores de Dopamina D1/fisiologia , Receptores de Dopamina D5/fisiologia , Receptores da Neurocinina-1/fisiologia , Animais , Benzazepinas/farmacologia , Carragenina/farmacologia , Dinoprostona/metabolismo , Dinoprostona/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Hiperalgesia/induzido quimicamente , Interleucina-6/farmacologia , Masculino , Camundongos , Células do Corno Posterior/efeitos dos fármacos , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D5/agonistas , Receptores de Dopamina D5/antagonistas & inibidores , Neurônios Serotoninérgicos/fisiologia , Sulpirida/farmacologia
5.
Mol Pain ; 122016.
Artigo em Inglês | MEDLINE | ID: mdl-27837171

RESUMO

Age-related bone fractures are usually painful and have highly negative effects on a geriatric patient's functional status, quality of life, and survival. Currently, there are few analgesic therapies that fully control bone fracture pain in the elderly without significant unwanted side effects. However, another way of controlling age-related fracture pain would be to preemptively administer an osteo-anabolic agent to geriatric patients with high risk of fracture, so as to build new cortical bone and prevent the fracture from occurring. A major question, however, is whether an osteo-anabolic agent can stimulate the proliferation of osteogenic cells and build significant amounts of new cortical bone in light of the decreased number and responsiveness of osteogenic cells in aging bone. To explore this question, geriatric and young mice, 20 and 4 months old, respectively, received either vehicle or a monoclonal antibody that sequesters sclerostin (anti-sclerostin) for 28 days. From days 21 to 28, animals also received sustained administration of the thymidine analog, bromodeoxyuridine (BrdU), which labels the DNA of dividing cells. Animals were then euthanized at day 28 and the femurs were examined for cortical bone formation, bone mineral density, and newly borne BrdU+ cells in the periosteum which is a tissue that is pivotally involved in the formation of new cortical bone. In both the geriatric and young mice, anti-sclerostin induced a significant increase in the thickness of the cortical bone, bone mineral density, and the proliferation of newly borne BrdU+ cells in the periosteum. These results suggest that even in geriatric animals, anti-sclerostin therapy can build new cortical bone and increase the proliferation of osteogenic cells and thus reduce the likelihood of painful age-related bone fractures.


Assuntos
Envelhecimento , Anticorpos/uso terapêutico , Proliferação de Células/fisiologia , Osso Cortical/patologia , Fraturas Ósseas/complicações , Dor , Periósteo/patologia , Proteínas Adaptadoras de Transdução de Sinal , Análise de Variância , Animais , Densidade Óssea , Bromodesoxiuridina/metabolismo , Glicoproteínas/imunologia , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Osteogênese/efeitos dos fármacos , Dor/etiologia , Dor/patologia , Dor/prevenção & controle
6.
Eur J Neurosci ; 39(3): 508-19, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24494689

RESUMO

Disorders of the skeleton are one of the most common causes of chronic pain and long-term physical disability in the world. Chronic skeletal pain is caused by a remarkably diverse group of conditions including trauma-induced fracture, osteoarthritis, osteoporosis, low back pain, orthopedic procedures, celiac disease, sickle cell disease and bone cancer. While these disorders are diverse, what they share in common is that when chronic skeletal pain occurs in these disorders, there are currently few therapies that can fully control the pain without significant unwanted side effects. In this review we focus on recent advances in our knowledge concerning the unique population of primary afferent sensory nerve fibers that innervate the skeleton, the nociceptive and neuropathic mechanisms that are involved in driving skeletal pain, and the neurochemical and structural changes that can occur in sensory and sympathetic nerve fibers and the CNS in chronic skeletal pain. We also discuss therapies targeting nerve growth factor or sclerostin for treating skeletal pain. These therapies have provided unique insight into the factors that drive skeletal pain and the structural decline that occurs in the aging skeleton. We conclude by discussing how these advances have changed our understanding and potentially the therapeutic options for treating and/or preventing chronic pain in the injured, diseased and aged skeleton.


Assuntos
Osso e Ossos/inervação , Dor Musculoesquelética/fisiopatologia , Neuralgia/fisiopatologia , Dor Nociceptiva/fisiopatologia , Anabolizantes/uso terapêutico , Analgésicos/uso terapêutico , Animais , Osso e Ossos/fisiopatologia , Humanos , Dor Musculoesquelética/tratamento farmacológico , Neuralgia/tratamento farmacológico , Dor Nociceptiva/tratamento farmacológico
7.
Arthritis Rheum ; 64(7): 2223-32, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22246649

RESUMO

OBJECTIVE: Many forms of arthritis are accompanied by significant chronic joint pain. This study was undertaken to investigate whether there is significant sprouting of sensory and sympathetic nerve fibers in the painful arthritic knee joint and whether nerve growth factor (NGF) drives this pathologic reorganization. METHODS: A painful arthritic knee joint was produced by injection of Freund's complete adjuvant (CFA) into the knee joint of young adult mice. CFA-injected mice were then treated systemically with vehicle or anti-NGF antibody. Pain behaviors were assessed, and at 28 days following the initial CFA injection, the knee joints were processed for immunohistochemistry analysis using antibodies against calcitonin gene-related peptide (CGRP; sensory nerve fibers), neurofilament 200 kd (NF200; sensory nerve fibers), growth-associated protein 43 (GAP-43; sprouted nerve fibers), tyrosine hydroxylase (TH; sympathetic nerve fibers), CD31 (endothelial cells), or CD68 (monocyte/macrophages). RESULTS: In CFA-injected mice, there was a significant increase in the density of CD68+ macrophages, CD31+ blood vessels, and CGRP+, NF200+, GAP-43+, and TH+ nerve fibers in the synovium, as well as a significant increase in joint pain-related behaviors. None of these findings were observed in sham-injected mice. Administration of anti-NGF reduced these pain-related behaviors and the ectopic sprouting of nerve fibers, but had no significant effect on the increase in density of CD31+ blood vessels or CD68+ macrophages. CONCLUSION: These findings demonstrate that ectopic sprouting of sensory and sympathetic nerve fibers occurs in the painful arthritic joint and may be involved in the generation and maintenance of arthritic pain.


Assuntos
Artrite Experimental/fisiopatologia , Fibras Nervosas/fisiologia , Plasticidade Neuronal/fisiologia , Dor/fisiopatologia , Células Receptoras Sensoriais/fisiologia , Sistema Nervoso Simpático/fisiopatologia , Fibras Adrenérgicas/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Artrite Experimental/metabolismo , Comportamento Animal/fisiologia , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Modelos Animais de Doenças , Proteína GAP-43/metabolismo , Articulação do Joelho/metabolismo , Articulação do Joelho/fisiopatologia , Masculino , Camundongos , Fibras Nervosas/metabolismo , Proteínas de Neurofilamentos/metabolismo , Dor/metabolismo , Medição da Dor , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Células Receptoras Sensoriais/metabolismo , Sistema Nervoso Simpático/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
8.
Nat Rev Cancer ; 2(3): 201-9, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11990856

RESUMO

Pain is the most disruptive influence on the quality of life of cancer patients. Although significant advances are being made in cancer treatment and diagnosis, the basic neurobiology of cancer pain is poorly understood. New insights into these mechanisms are now arising from animal models, and have the potential to fundamentally change the way that cancer pain is controlled.


Assuntos
Vias Aferentes/fisiopatologia , Neoplasias/fisiopatologia , Dor , Humanos , Neurônios Aferentes/fisiologia , Nociceptores/fisiologia
9.
J Pharmacol Exp Ther ; 342(2): 416-28, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22570364

RESUMO

The transient receptor potential vanilloid-1 (TRPV1) channel is involved in the development and maintenance of pain and participates in the regulation of temperature. The channel is activated by diverse agents, including capsaicin, noxious heat (≥ 43°C), acidic pH (< 6), and endogenous lipids including N-arachidonoyl dopamine (NADA). Antagonists that block all modes of TRPV1 activation elicit hyperthermia. To identify efficacious TRPV1 antagonists that do not affect temperature antagonists representing multiple TRPV1 pharmacophores were evaluated at recombinant rat and human TRPV1 channels with Ca(2+) flux assays, and two classes of antagonists were identified based on their differential ability to inhibit acid activation. Although both classes of antagonists completely blocked capsaicin- and NADA-induced activation of TRPV1, select compounds only partially inhibited activation of the channel by protons. Electrophysiology and calcitonin gene-related peptide release studies confirmed the differential pharmacology of these antagonists at native TRPV1 channels in the rat. Comparison of the in vitro pharmacological properties of these TRPV1 antagonists with their in vivo effects on core body temperature confirms and expands earlier observations that acid-sparing TRPV1 antagonists do not significantly increase core body temperature. Although both classes of compounds elicit equivalent analgesia in a rat model of knee joint pain, the acid-sparing antagonist tested is not effective in a mouse model of bone cancer pain.


Assuntos
Temperatura Corporal/efeitos dos fármacos , Canais de Cátion TRPV/antagonistas & inibidores , Analgésicos/farmacologia , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Cálcio/metabolismo , Capsaicina/farmacologia , Linhagem Celular Transformada , Febre/tratamento farmacológico , Febre/fisiopatologia , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Dor/tratamento farmacológico , Dor/metabolismo , Dor/fisiopatologia , Prótons , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Canais de Cátion TRPV/metabolismo
10.
Neuron ; 55(3): 377-91, 2007 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-17678852

RESUMO

Our understanding of the neural correlates of pain perception in humans has increased significantly since the advent of neuroimaging. Relating neural activity changes to the varied pain experiences has led to an increased awareness of how factors (e.g., cognition, emotion, context, injury) can separately influence pain perception. Tying this body of knowledge in humans to work in animal models of pain provides an opportunity to determine common features that reliably contribute to pain perception and its modulation. One key system that underpins the ability to change pain intensity is the brainstem's descending modulatory network with its pro- and antinociceptive components. We discuss not only the latest data describing the cerebral signature of pain and its modulation in humans, but also suggest that the brainstem plays a pivotal role in gating the degree of nociceptive transmission so that the resultant pain experienced is appropriate for the particular situation of the individual.


Assuntos
Encéfalo/fisiopatologia , Dor/fisiopatologia , Dor/psicologia , Afeto , Animais , Tronco Encefálico/fisiopatologia , Sistema Nervoso Central/fisiopatologia , Cognição , Vias Eferentes/fisiopatologia , Emoções , Humanos , Nociceptores
11.
J Neurosci ; 30(44): 14649-56, 2010 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-21048122

RESUMO

Pain frequently accompanies cancer. What remains unclear is why this pain frequently becomes more severe and difficult to control with disease progression. Here we test the hypothesis that with disease progression, sensory nerve fibers that innervate the tumor-bearing tissue undergo a pathological sprouting and reorganization, which in other nonmalignant pathologies has been shown to generate and maintain chronic pain. Injection of canine prostate cancer cells into mouse bone induces a remarkable sprouting of calcitonin gene-related peptide (CGRP(+)) and neurofilament 200 kDa (NF200(+)) sensory nerve fibers. Nearly all sensory nerve fibers that undergo sprouting also coexpress tropomyosin receptor kinase A (TrkA(+)). This ectopic sprouting occurs in sensory nerve fibers that are in close proximity to colonies of prostate cancer cells, tumor-associated stromal cells and newly formed woven bone, which together form sclerotic lesions that closely mirror the osteoblastic bone lesions induced by metastatic prostate tumors in humans. Preventive treatment with an antibody that sequesters nerve growth factor (NGF), administered when the pain and bone remodeling were first observed, blocks this ectopic sprouting and attenuates cancer pain. Interestingly, reverse transcription PCR analysis indicated that the prostate cancer cells themselves do not express detectable levels of mRNA coding for NGF. This suggests that the tumor-associated stromal cells express and release NGF, which drives the pathological reorganization of nearby TrkA(+) sensory nerve fibers. Therapies that prevent this reorganization of sensory nerve fibers may provide insight into the evolving mechanisms that drive cancer pain and lead to more effective control of this chronic pain state.


Assuntos
Neoplasias Ósseas/patologia , Neoplasias Ósseas/secundário , Osso e Ossos/inervação , Nociceptores/patologia , Dor/patologia , Neoplasias da Próstata/patologia , Animais , Osso e Ossos/patologia , Osso e Ossos/fisiopatologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Cães , Masculino , Camundongos , Camundongos Nus , Nociceptores/metabolismo , Dor/tratamento farmacológico , Dor/etiologia , Neoplasias da Próstata/fisiopatologia , Células Receptoras Sensoriais/patologia
12.
Anesthesiology ; 115(1): 189-204, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21602663

RESUMO

Nerve growth factor (NGF) was originally discovered as a neurotrophic factor essential for the survival of sensory and sympathetic neurons during development. However, in the adult NGF has been found to play an important role in nociceptor sensitization after tissue injury. The authors outline mechanisms by which NGF activation of its cognate receptor, tropomyosin-related kinase A receptor, regulates a host of ion channels, receptors, and signaling molecules to enhance acute and chronic pain. The authors also document that peripherally restricted antagonism of NGF-tropomyosin-related kinase A receptor signaling is effective for controlling human pain while appearing to maintain normal nociceptor function. Understanding whether there are any unexpected adverse events and how humans may change their behavior and use of the injured/degenerating tissue after significant pain relief without sedation will be required to fully appreciate the patient populations that may benefit from these therapies targeting NGF.


Assuntos
Analgésicos/farmacologia , Analgésicos/uso terapêutico , Fatores de Crescimento Neural/antagonistas & inibidores , Dor/tratamento farmacológico , Receptor trkA/antagonistas & inibidores , Adulto , Animais , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Modelos Animais de Doenças , Humanos , Fatores de Crescimento Neural/fisiologia , Neuroma/patologia , Nociceptores/efeitos dos fármacos , Nociceptores/fisiologia , Receptor trkA/fisiologia , Receptores de Fator de Crescimento Neural/metabolismo , Transdução de Sinais/efeitos dos fármacos
13.
Mol Pain ; 6: 87, 2010 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-21138586

RESUMO

Pain often accompanies cancer and most current therapies for treating cancer pain have significant unwanted side effects. Targeting nerve growth factor (NGF) or its cognate receptor tropomyosin receptor kinase A (TrkA) has become an attractive target for attenuating chronic pain. In the present report, we use a mouse model of bone cancer pain and examine whether oral administration of a selective small molecule Trk inhibitor (ARRY-470, which blocks TrkA, TrkB and TrkC kinase activity at low nm concentrations) has a significant effect on cancer-induced pain behaviors, tumor-induced remodeling of sensory nerve fibers, tumor growth and tumor-induced bone remodeling. Early/sustained (initiated day 6 post cancer cell injection), but not late/acute (initiated day 18 post cancer cell injection) administration of ARRY-470 markedly attenuated bone cancer pain and significantly blocked the ectopic sprouting of sensory nerve fibers and the formation of neuroma-like structures in the tumor bearing bone, but did not have a significant effect on tumor growth or bone remodeling. These data suggest that, like therapies that target the cancer itself, the earlier that the blockade of TrkA occurs, the more effective the control of cancer pain and the tumor-induced remodeling of sensory nerve fibers. Developing targeted therapies that relieve cancer pain without the side effects of current analgesics has the potential to significantly improve the quality of life and functional status of cancer patients.


Assuntos
Neuroma/tratamento farmacológico , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/administração & dosagem , Pirazóis/farmacologia , Pirimidinas/administração & dosagem , Pirimidinas/farmacologia , Receptor trkA/antagonistas & inibidores , Sarcoma/tratamento farmacológico , Animais , Remodelação Óssea/efeitos dos fármacos , Modelos Animais de Doenças , Camundongos , Neuroma/prevenção & controle , Inibidores de Proteínas Quinases/uso terapêutico , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico , Sarcoma/patologia , Células Receptoras Sensoriais/efeitos dos fármacos , Fatores de Tempo , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos
14.
Brain Res Rev ; 60(1): 187-201, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19166876

RESUMO

Non-malignant musculoskeletal pain is the most common clinical symptom that causes patients to seek medical attention and is a major cause of disability in the world. Musculoskeletal pain can arise from a variety of common conditions including osteoarthritis, rheumatoid arthritis, osteoporosis, surgery, low back pain and bone fracture. A major problem in designing new therapies to treat musculoskeletal pain is that the underlying mechanisms driving musculoskeletal pain are not well understood. This lack of knowledge is largely due to the scarcity of animal models that closely mirror the human condition which would allow the development of a mechanistic understanding and novel therapies to treat this pain. To begin to develop a mechanism-based understanding of the factors involved in generating musculoskeletal pain, in this review we present recent advances in preclinical models of osteoarthritis, post-surgical pain and bone fracture pain. The models discussed appear to offer an attractive platform for understanding the factors that drive this pain and the preclinical screening of novel therapies to treat musculoskeletal pain. Developing both an understanding of the mechanisms that drive persistent musculoskeletal pain and novel mechanism-based therapies to treat these unique pain states would address a major unmet clinical need and have significant clinical, economic and societal benefits.


Assuntos
Artralgia/fisiopatologia , Doenças Musculoesqueléticas/fisiopatologia , Animais , Artralgia/tratamento farmacológico , Artralgia/etiologia , Osso e Ossos/inervação , Osso e Ossos/fisiopatologia , Modelos Animais de Doenças , Fraturas Ósseas/fisiopatologia , Humanos , Hiperalgesia/etiologia , Hiperalgesia/fisiopatologia , Inflamação/fisiopatologia , Camundongos , Osteoartrite/fisiopatologia , Dor Pós-Operatória/fisiopatologia , Ratos , Toracotomia/efeitos adversos
15.
Vet Rec ; 184(1): 23, 2019 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-30368458

RESUMO

Nerve growth factor (NGF) is essential for the survival of sensory and sympathetic neurons during development. However, in the adult, NGF and its interaction with tropomyosin receptor kinase A receptor (TrkA) has been found to play a critical role in nociception and nervous system plasticity in pain conditions. Thus, various monoclonal antibody (mAb) therapies targeting this pathway have been investigated in the development of new pharmacotherapies for chronic pain. Although none of the mAbs against NGF are yet approved for use in humans, they look very promising for the effective control of pain. Recently, species-specific anti-NGF mAbs for the management of osteoarthritis (OA)-associated pain in dogs and cats has been developed, and early clinical trials have been conducted. Anti-NGF therapy looks to be both very effective and very promising as a novel therapy against chronic pain in dogs and cats. This review outlines the mechanism of action of NGF, the role of NGF in osteoarthritis, research in rodent OA models and the current status of the development of anti-NGF mAbs in humans. Furthermore, we describe and discuss the recent development of species-specific anti-NGF mAbs for the treatment of OA-associated pain in veterinary medicine.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Doenças do Gato/terapia , Doenças do Cão/terapia , Fatores de Crescimento Neural/antagonistas & inibidores , Osteoartrite/veterinária , Manejo da Dor/veterinária , Animais , Gatos , Cães , Osteoartrite/complicações , Osteoartrite/terapia , Dor/etiologia , Dor/veterinária , Ensaios Clínicos Controlados Aleatórios como Assunto
16.
Mol Pain ; 4: 10, 2008 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-18353190

RESUMO

Although a variety of industrial chemicals, as well as several chemotherapeutic agents used to treat cancer or HIV, preferentially induce a peripheral sensory neuropathy what remains unclear is why these agents induce a sensory vs. a motor or mixed neuropathy. Previous studies have shown that the endothelial cells that vascularize the dorsal root ganglion (DRG), which houses the primary afferent sensory neurons, are unique in that they have large fenestrations and are permeable to a variety of low and high molecular weight agents. In the present report we used whole-mount preparations, immunohistochemistry, and confocal laser scanning microscopy to show that the cell body-rich area of the L4 mouse DRG has a 7 fold higher density of CD31+ capillaries than cell fiber rich area of the DRG or the distal or proximal aspect of the sciatic nerve. This dense vascularization, coupled with the high permeability of these capillaries, may synergistically contribute, and in part explain, why many potentially neurotoxic agents preferentially accumulate and injure cells within the DRG. Currently, cancer survivors and HIV patients constitute the largest and most rapidly expanding groups that have chemically induced peripheral sensory neuropathy. Understanding the unique aspects of the vascularization of the DRG and closing the endothelial fenestrations of the rich vascular bed of capillaries that vascularize the DRG before intravenous administration of anti-neoplastic or anti-HIV therapies, may offer a mechanism based approach to attenuate these chemically induced peripheral neuropathies in these patients.


Assuntos
Gânglios Espinais/irrigação sanguínea , Neovascularização Patológica/metabolismo , Nervos Periféricos/irrigação sanguínea , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Animais , Modelos Animais de Doenças , Gânglios Espinais/ultraestrutura , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C3H , Nervos Periféricos/ultraestrutura , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia
17.
Anesthesiology ; 108(3): 473-83, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18292685

RESUMO

BACKGROUND: Because of the relative lack of understanding of the mechanisms that drive skeletal pain, the purpose of this study was to adapt a previously validated closed femur fracture model to quantitatively evaluate skeletal pain in female and male rats. METHODS: Three-month-old female and male Sprague-Dawley rats were anesthetized, and a stainless steel pin was inserted into the intramedullary space of the left femur. Three weeks later, the rats were reanesthetized, and left femoral diaphyses were fractured using a standardized impactor device. At 1-21 days after fracture, skeletal pain was measured by quantitatively assessing spontaneous guarding, spontaneous flinching, and weight bearing of the fractured hind limb. RESULTS: Females and males showed highly robust pain behaviors that were maximal at day 1 after fracture and returned gradually to normal nonfractured levels at days 14-21 after fracture. The magnitude of fracture pain was not significantly different at most time points between female and male rats. In both females and males, the pain-related behaviors were attenuated by subcutaneous morphine in a dose-dependent manner. CONCLUSIONS: This model may help in developing a mechanism-based understanding of the factors that generate and maintain fracture pain in both females and males and in translating these findings into new therapies for treating fracture pain.


Assuntos
Doenças Ósseas/fisiopatologia , Modelos Animais de Doenças , Fraturas do Fêmur/fisiopatologia , Medição da Dor/métodos , Dor/fisiopatologia , Animais , Doenças Ósseas/tratamento farmacológico , Feminino , Fraturas do Fêmur/tratamento farmacológico , Masculino , Morfina/uso terapêutico , Dor/tratamento farmacológico , Ratos , Ratos Sprague-Dawley
18.
Neuroscience ; 387: 178-190, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29432884

RESUMO

Although bone is continually being remodeled and ultimately declines with aging, little is known whether similar changes occur in the sensory and sympathetic nerve fibers that innervate bone. Here, immunohistochemistry and confocal microscopy were used to examine changes in the sensory and sympathetic nerve fibers that innervate the young (10 days post-partum), adult (3 months) and aging (24 months) C57Bl/6 mouse femur. In all three ages examined, the periosteum was the most densely innervated bone compartment. With aging, the total number of sensory and sympathetic nerve fibers clearly declines as the cambium layer of the periosteum dramatically thins. Yet even in the aging femur, there remains a dense sensory and sympathetic innervation of the periosteum. In cortical bone, sensory and sympathetic nerve fibers are largely confined to vascularized Haversian canals and while there is no significant decline in the density of sensory fibers, there was a 75% reduction in sympathetic nerve fibers in the aging vs. adult cortical bone. In contrast, in the bone marrow the overall density/unit area of both sensory and sympathetic nerve fibers appeared to remain largely unchanged across the lifespan. The preferential preservation of sensory nerve fibers suggests that even as bone itself undergoes a marked decline with age, the nociceptors that detect injury and signal skeletal pain remain relatively intact.


Assuntos
Fibras Adrenérgicas/fisiologia , Vias Aferentes/anatomia & histologia , Envelhecimento/fisiologia , Fêmur/inervação , Vias Aferentes/citologia , Animais , Imuno-Histoquímica , Masculino , Camundongos , Microscopia Confocal
19.
Pain ; 159(11): 2285-2295, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29994990

RESUMO

Anti-nerve growth factor (anti-NGF) therapy has shown significant promise in attenuating several types of skeletal pain. However, whether anti-NGF therapy changes the level of physical activity in individuals with or without skeletal pain is largely unknown. Here, automated day/night activity boxes monitored the effects of anti-NGF treatment on physical activity in normal young (3 months old) and aging (18-23 months old) mice and mice with bone fracture pain. Although aging mice were clearly less active and showed loss of bone mass compared with young mice, anti-NGF treatment had no effect on any measure of day/night activity in either the young or aging mice. By contrast, in mice with femoral fracture pain, anti-NGF treatment produced a clear increase (10%-27%) in horizontal activity, vertical rearing, and velocity of travel compared with the Fracture + Vehicle group. These results suggest, just as in humans, mice titrate their level of physical activity to their level of skeletal pain. The level of skeletal pain may in part be determined by the level of free NGF that seems to rise after injury but not normal aging of the skeleton. In terms of bone healing, animals that received anti-NGF showed an increase in the size of calcified callus but no increase in the number of displaced fractures or time to cortical union. As physical activity is the best nondrug treatment for many patients with skeletal pain, anti-NGF may be useful in reducing pain and promoting activity in these patients.


Assuntos
Envelhecimento , Anticorpos/uso terapêutico , Fator de Crescimento Neural/imunologia , Dor/tratamento farmacológico , Dor/etiologia , Condicionamento Físico Animal/fisiologia , Animais , Relação Dose-Resposta a Droga , Fraturas Ósseas/complicações , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Crescimento Neural/metabolismo , Fatores de Tempo , Cicatrização , Raios X
20.
J Bone Miner Res ; 22(11): 1732-42, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17638576

RESUMO

UNLABELLED: A closed femur fracture pain model was developed in the C57BL/6J mouse. One day after fracture, a monoclonal antibody raised against nerve growth factor (anti-NGF) was delivered intraperitoneally and resulted in a reduction in fracture pain-related behaviors of approximately 50%. Anti-NGF therapy did not interfere with bone healing as assessed by mechanical testing and histomorphometric analysis. INTRODUCTION: Current therapies to treat skeletal fracture pain are limited. This is because of the side effect profile of available analgesics and the scarcity of animal models that can be used to understand the mechanisms that drive this pain. Whereas previous studies have shown that mineralized bone, marrow, and periosteum are innervated by sensory and sympathetic fibers, it is not understood how skeletal pain is generated and maintained even in common conditions such as osteoarthritis, low back pain, or fracture. MATERIALS AND METHODS: In this study, we characterized the pain-related behaviors after a closed femur fracture in the C57BL/6J mouse. Additionally, we assessed the effect of a monoclonal antibody that binds to and sequesters nerve growth factor (anti-NGF) on pain-related behaviors and bone healing (mechanical properties and histomorphometric analysis) after fracture. RESULTS: Administration of anti-NGF therapy (10 mg/kg, days 1, 6, and 11 after fracture) resulted in a reduction of fracture pain-related behaviors of approximately 50%. Attenuation of fracture pain was evident as early as 24 h after the initial dosing and remained efficacious throughout the course of fracture pain. Anti-NGF therapy did not modify biomechanical properties of the femur or histomorphometric indices of bone healing. CONCLUSIONS: These findings suggest that therapies that target NGF or its cognate receptor(s) may be effective in attenuating nonmalignant fracture pain without interfering with bone healing.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Fraturas do Fêmur/tratamento farmacológico , Fator de Crescimento Neural/antagonistas & inibidores , Animais , Fenômenos Biomecânicos , Fraturas do Fêmur/diagnóstico por imagem , Fêmur/diagnóstico por imagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Crescimento Neural/imunologia , Dor/tratamento farmacológico , Radiografia , Cicatrização
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA