Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Pain ; 157(6): 1239-1247, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27186713

ABSTRACT

Recent studies have suggested that in humans and animals with significant skeletal pain, changes in the mechanical hypersensitivity of the skin can be detected. However, whether measuring changes in skin hypersensitivity can be a reliable surrogate for measuring skeletal pain itself remains unclear. To explore this question, we generated skeletal pain by injecting and confining GFP-transfected NCTC 2472 osteosarcoma cells unilaterally to the femur of C3H male mice. Beginning at day 7 post-tumor injection, animals were administered vehicle, an antibody to the P2X3 receptor (anti-P2X3) or anti-NGF antibody. Pain and analgesic efficacy were then measured on days 21, 28, and 35 post-tumor injection using a battery of skeletal pain-related behaviors and von Frey assessment of mechanical hypersensitivity on the plantar surface of the hind paw. Animals with bone cancer pain treated with anti-P2X3 showed a reduction in skin hypersensitivity but no attenuation of skeletal pain behaviors, whereas animals with bone cancer pain treated with anti-NGF showed a reduction in both skin hypersensitivity and skeletal pain behaviors. These results suggest that although bone cancer can induce significant skeletal pain-related behaviors and hypersensitivity of the skin, relief of hypersensitivity of the skin is not always accompanied by attenuation of skeletal pain. Understanding the relationship between skeletal and skin pain may provide insight into how pain is processed and integrated and help define the preclinical measures of skeletal pain that are predictive end points for clinical trials.


Subject(s)
Behavior, Animal/physiology , Bone Neoplasms/physiopathology , Cancer Pain/physiopathology , Osteosarcoma/physiopathology , Skin/physiopathology , Animals , Bone Neoplasms/psychology , Cancer Pain/psychology , Cell Line, Tumor , Disease Models, Animal , Hyperalgesia/physiopathology , Hyperalgesia/psychology , Male , Mice , Mice, Inbred C3H , Neoplasm Transplantation , Osteosarcoma/psychology
2.
Pain ; 156(1): 157-165, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25599311

ABSTRACT

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


Subject(s)
Femur/injuries , Fractures, Bone/drug therapy , Nerve Growth Factor/antagonists & inhibitors , Orthopedic Procedures/adverse effects , Pain, Postoperative/drug therapy , Pain/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Femur/diagnostic imaging , Fractures, Bone/diagnostic imaging , Male , Mice , Mice, Inbred C3H , Pain/diagnostic imaging , Pain, Postoperative/diagnostic imaging , Radiography , Time Factors
3.
Cancer Res ; 74(23): 7014-23, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25287160

ABSTRACT

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


Subject(s)
Antibodies/pharmacology , Bone Neoplasms/drug therapy , Nerve Growth Factor/antagonists & inhibitors , Nerve Growth Factors/antagonists & inhibitors , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Extremities/pathology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Male , Mice , Mice, Inbred C3H , Neoplasm Metastasis , Nerve Growth Factor/metabolism , Nerve Growth Factors/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Pain/drug therapy , Pain/metabolism , Random Allocation , Sarcoma/drug therapy , Sarcoma/metabolism , Sarcoma/pathology
4.
Pain ; 155(11): 2323-36, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25196264

ABSTRACT

Skeletal injury is a leading cause of chronic pain and long-term disability worldwide. While most acute skeletal pain can be effectively managed with nonsteroidal anti-inflammatory drugs and opiates, chronic skeletal pain is more difficult to control using these same therapy regimens. One possibility as to why chronic skeletal pain is more difficult to manage over time is that there may be nerve sprouting in nonhealed areas of the skeleton that normally receive little (mineralized bone) to no (articular cartilage) innervation. If such ectopic sprouting did occur, it could result in normally nonnoxious loading of the skeleton being perceived as noxious and/or the generation of a neuropathic pain state. To explore this possibility, a mouse model of skeletal pain was generated by inducing a closed fracture of the femur. Examined animals had comminuted fractures and did not fully heal even at 90+days post fracture. In all mice with nonhealed fractures, exuberant sensory and sympathetic nerve sprouting, an increase in the density of nerve fibers, and the formation of neuroma-like structures near the fracture site were observed. Additionally, all of these animals exhibited significant pain behaviors upon palpation of the nonhealed fracture site. In contrast, sprouting of sensory and sympathetic nerve fibers or significant palpation-induced pain behaviors was never observed in naïve animals. Understanding what drives this ectopic nerve sprouting and the role it plays in skeletal pain may allow a better understanding and treatment of this currently difficult-to-control pain state.


Subject(s)
Adrenergic Fibers/pathology , Fractures, Bone/complications , Musculoskeletal Pain/etiology , Musculoskeletal Pain/pathology , Adrenergic Fibers/physiology , Animals , Calcification, Physiologic/physiology , Calcitonin Gene-Related Peptide/metabolism , Chronic Pain , Disease Models, Animal , Fractures, Bone/pathology , GAP-43 Protein/metabolism , Imaging, Three-Dimensional , Male , Mice , Mice, Inbred C57BL , Neurofilament Proteins/metabolism , Neuroma/etiology , Neuroma/pathology , Pain Measurement , Palpation/adverse effects , Statistics, Nonparametric , X-Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...