Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Postgrad Med ; 133(8): 879-894, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34252357

ABSTRACT

Osteoarthritis (OA) is a common difficult-to-treat condition where the goal, in the absence of disease-modifying treatments, is to alleviate symptoms such as pain and loss of function. Acetaminophen, nonsteroidal anti-inflammatory drugs (NSAIDs), and opioids are common pharmacologic treatments for OA. Antibodies directed against nerve growth factor (NGF-Abs) are a new class of agents under clinical investigation for the treatment of OA. This narrative review describes (and uses schematics to visualize) nociceptive signaling, chronification of pain, and the mechanisms of action (MOAs) of these different analgesics in the context of OA-related pain pathophysiology. Further, the varying levels of efficacy and safety of these agents observed in patients with OA is examined, based on an overview of published clinical data and/or treatment guidelines (when available), in the context of differences in their MOAs.


Subject(s)
Acetaminophen/therapeutic use , Analgesics, Non-Narcotic/therapeutic use , Analgesics, Opioid/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Osteoarthritis/drug therapy , Pain Management
2.
J Pain Res ; 13: 1223-1241, 2020.
Article in English | MEDLINE | ID: mdl-32547184

ABSTRACT

Nerve growth factor (NGF) is a neurotrophic protein essential for the growth, differentiation, and survival of sympathetic and sensory afferent neurons during development. A substantial body of evidence, based on both animal and human studies, demonstrates that NGF plays a pivotal role in modulation of nociception in adulthood. This has spurred development of a variety of novel analgesics that target the NGF signaling pathway. Here, we present a narrative review designed to summarize how NGF receptor activation and downstream signaling alters nociception through direct sensitization of nociceptors at the site of injury and changes in gene expression in the dorsal root ganglion that collectively increase nociceptive signaling from the periphery to the central nervous system. This review illustrates that NGF has a well-known and multifunctional role in nociceptive processing, although the precise signaling pathways downstream of NGF receptor activation that mediate nociception are complex and not completely understood. Additionally, much of the existing knowledge derives from studies performed in animal models and may not accurately represent the human condition. However, available data establish a role for NGF in the modulation of nociception through effects on the release of inflammatory mediators, nociceptive ion channel/receptor activity, nociceptive gene expression, and local neuronal sprouting. The role of NGF in nociception and the generation and/or maintenance of chronic pain has led to it becoming a novel and attractive target of pain therapeutics for the treatment of chronic pain conditions.

3.
Pain ; 160(10): 2210-2220, 2019 10.
Article in English | MEDLINE | ID: mdl-31145219

ABSTRACT

Chronic pain continues to be a significant global burden despite the availability of a variety of nonpharmacologic and pharmacologic treatment options. Thus, there is a need for new analgesics with novel mechanisms of action. In this regard, antibodies directed against nerve growth factor (NGF-Abs) are a new class of agents in development for the treatment of chronic pain conditions such as osteoarthritis and chronic low-back pain. This comprehensive narrative review summarizes evidence supporting pronociceptive functions for NGF that include contributing to peripheral and central sensitization through tropomyosin receptor kinase A activation and stimulation of local neuronal sprouting. The potential role of NGF in osteoarthritis and chronic low-back pain signaling is also examined to provide a mechanistic basis for the observed efficacy of NGF-Abs in clinical trials of these particular pain states. Finally, the safety profile of NGF-Abs in terms of common adverse events, joint safety, and nerve structure/function is discussed.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Chronic Pain/drug therapy , Low Back Pain/drug therapy , Nerve Growth Factor , Osteoarthritis/drug therapy , Animals , Antibodies, Monoclonal, Humanized/adverse effects , Chronic Pain/diagnosis , Chronic Pain/epidemiology , Clinical Trials as Topic/methods , Humans , Low Back Pain/diagnosis , Low Back Pain/epidemiology , Osteoarthritis/diagnosis , Osteoarthritis/epidemiology , Treatment Outcome
4.
Br J Clin Pharmacol ; 85(6): 1103-1113, 2019 06.
Article in English | MEDLINE | ID: mdl-30357885

ABSTRACT

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.


Subject(s)
Bone and Bones/innervation , Chemoreceptor Cells/metabolism , Longevity , Mechanoreceptors/metabolism , Musculoskeletal Pain/physiopathology , Pain Threshold , Sympathetic Nervous System/physiopathology , Age Factors , Analgesics/therapeutic use , Animals , Central Nervous System Sensitization , Humans , Musculoskeletal Pain/drug therapy , Musculoskeletal Pain/epidemiology , Musculoskeletal Pain/psychology , Pain Perception , Pain Threshold/drug effects , Quality of Life , Risk Factors
5.
Vet Rec ; 184(1): 23, 2019 Jan 05.
Article in English | MEDLINE | ID: mdl-30368458

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cat Diseases/therapy , Dog Diseases/therapy , Nerve Growth Factors/antagonists & inhibitors , Osteoarthritis/veterinary , Pain Management/veterinary , Animals , Cats , Dogs , Osteoarthritis/complications , Osteoarthritis/therapy , Pain/etiology , Pain/veterinary , Randomized Controlled Trials as Topic
6.
Pain ; 159(11): 2285-2295, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29994990

ABSTRACT

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.


Subject(s)
Aging , Antibodies/therapeutic use , Nerve Growth Factor/immunology , Pain/drug therapy , Pain/etiology , Physical Conditioning, Animal/physiology , Animals , Dose-Response Relationship, Drug , Fractures, Bone/complications , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factor/metabolism , Time Factors , Wound Healing , X-Rays
7.
Curr Osteoporos Rep ; 16(4): 325-332, 2018 08.
Article in English | MEDLINE | ID: mdl-29948820

ABSTRACT

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.


Subject(s)
Acute Pain/physiopathology , Bone and Bones/innervation , Chronic Pain/physiopathology , Fractures, Bone/physiopathology , Neuralgia/physiopathology , Nociceptive Pain/physiopathology , Acute Pain/etiology , Acute Pain/therapy , Analgesics, Opioid/therapeutic use , Animals , Central Nervous System Sensitization , Chronic Pain/etiology , Chronic Pain/therapy , Disease Models, Animal , Fracture Healing , Fractures, Bone/complications , Fractures, Bone/therapy , Humans , Neuralgia/etiology , Neuralgia/therapy , Nociceptive Pain/etiology , Nociceptive Pain/therapy , Nociceptors , Pain Management , Peripheral Nerve Injuries/etiology , Peripheral Nerve Injuries/physiopathology , Sensory Receptor Cells
8.
Neuroscience ; 387: 178-190, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29432884

ABSTRACT

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.


Subject(s)
Adrenergic Fibers/physiology , Afferent Pathways/anatomy & histology , Aging/physiology , Femur/innervation , Afferent Pathways/cytology , Animals , Immunohistochemistry , Male , Mice , Microscopy, Confocal
9.
Mol Pain ; 13: 1744806917745465, 2017.
Article in English | MEDLINE | ID: mdl-29166838

ABSTRACT

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.


Subject(s)
Cartilage, Articular/metabolism , Femur/metabolism , Nerve Growth Factor/metabolism , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Animals , Calcification, Physiologic , Calcitonin Gene-Related Peptide/metabolism , Immunohistochemistry , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Tyrosine 3-Monooxygenase/metabolism
10.
Pain ; 158(4): 605-617, 2017 04.
Article in English | MEDLINE | ID: mdl-28301858

ABSTRACT

Total knee arthroplasty (TKA) and total hip arthroplasty (THA) are 2 of the most common and successful surgical interventions to relieve osteoarthritis pain. Control of postoperative pain is critical for patients to fully participate in the required physical therapy which is the most influential factor in effective postoperative knee rehabilitation. Currently, opiates are a mainstay for managing postoperative orthopedic surgery pain including TKA or THA pain. Recently, issues including efficacy, dependence, overdose, and death from opiates have made clinicians and researchers more critical of use of opioids for treating nonmalignant skeletal pain. In the present report, a nonopiate therapy using a monoclonal antibody raised against nerve growth factor (anti-NGF) was assessed for its ability to increase the spontaneous activity of the operated knee joint in a mouse model of orthopedic surgery pain-induced by drilling and coring the trochlear groove of the mouse femur. Horizontal activity and velocity and vertical rearing were continually assessed over a 20 hours day/night period using automated activity boxes in an effort to reduce observer bias and capture night activity when the mice are most active. At days 1 and 3, after orthopedic surgery, there was a marked reduction in spontaneous activity and vertical rearing; anti-NGF significantly attenuated this decline. The present data suggest that anti-NGF improves limb use in a rodent model of joint/orthopedic surgery and as such anti-NGF may be useful in controlling pain after orthopedic surgeries such as TKA or THA.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Circadian Rhythm/drug effects , Immunologic Factors/therapeutic use , Nerve Growth Factor/immunology , Pain/drug therapy , Analysis of Variance , Animals , Antibodies, Monoclonal/pharmacology , Disease Models, Animal , Immunologic Factors/pharmacology , Locomotion/drug effects , Male , Mice , Orthopedic Procedures/adverse effects , Pain/etiology , Treatment Outcome
11.
Pain Rep ; 2(5): e614, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29392229

ABSTRACT

INTRODUCTION: Cancer-induced bone pain (CIBP) is the most common type of pain with cancer. In humans, this pain can be difficult to control and highly disabling. A major problem with CIBP in humans is that it increases on weight-bearing and/or movement of a tumor-bearing bone limiting the activity and functional status of the patient. Currently, there is less data concerning whether similar negative changes in activity occur in rodent models of CIBP. OBJECTIVES: To determine whether there are marked changes in activity in a rodent model of CIBP and compare this to changes in skin hypersensitivity. METHODS: Osteosarcoma cells were injected and confined to 1 femur of the adult male mouse. Every 7 days, spontaneous horizontal and vertical activities were assessed over a 20-hour day and night period using automated activity boxes. Mechanical hypersensitivity of the hind paw skin was assessed using von Frey testing. RESULTS: As the tumor cells grew within the femur, there was a significant decline in horizontal and vertical activity during the times of the day/night when the mice are normally most active. Mice also developed significant hypersensitivity in the skin of the hind paw in the tumor-bearing limb. CONCLUSION: Even when the tumor is confined to a single load-bearing bone, CIBP drives a significant loss of activity, which increases with disease progression. Understanding the mechanisms that drive this reduction in activity may allow the development of therapies that allow CIBP patients to better maintain their activity and functional status.

12.
J Pain ; 18(3): 233-246, 2017 03.
Article in English | MEDLINE | ID: mdl-27884691

ABSTRACT

Chronic cancer pain is a serious complication of malignancy or its treatment. Currently, no comprehensive, universally accepted cancer pain classification system exists. Clarity in classification of common cancer pain syndromes would improve clinical assessment and management. Moreover, an evidence-based taxonomy would enhance cancer pain research efforts by providing consistent diagnostic criteria, ensuring comparability across clinical trials. As part of a collaborative effort between the Analgesic, Anesthetic, and Addiction Clinical Trial Translations, Innovations, Opportunities, and Networks (ACTTION) and the American Pain Society (APS), the ACTTION-APS Pain Taxonomy initiative worked to develop the characteristics of an optimal diagnostic system. After the establishment of these characteristics, a working group consisting of clinicians and clinical and basic scientists with expertise in cancer and cancer-related pain was convened to generate core diagnostic criteria for an illustrative sample of 3 chronic pain syndromes associated with cancer (ie, bone pain and pancreatic cancer pain as models of pain related to a tumor) or its treatment (ie, chemotherapy-induced peripheral neuropathy). A systematic review and synthesis was conducted to provide evidence for the dimensions that comprise this cancer pain taxonomy. Future efforts will subject these diagnostic categories and criteria to systematic empirical evaluation of their feasibility, reliability, and validity and extension to other cancer-related pain syndromes. PERSPECTIVE: The ACTTION-APS chronic cancer pain taxonomy provides an evidence-based classification for 3 prevalent syndromes, namely malignant bone pain, pancreatic cancer pain, and chemotherapy-induced peripheral neuropathy. This taxonomy provides consistent diagnostic criteria, common features, comorbidities, consequences, and putative mechanisms for these potentially serious cancer pain conditions that can be extended and applied with other cancer-related pain syndromes.


Subject(s)
Cancer Pain/diagnosis , Pain Measurement/methods , Societies, Medical/standards , Chronic Pain/diagnosis , Evidence-Based Medicine , Humans , Pain Measurement/standards , Public-Private Sector Partnerships , United States
13.
Mol Pain ; 122016.
Article in English | MEDLINE | ID: mdl-27837171

ABSTRACT

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.


Subject(s)
Aging , Antibodies/therapeutic use , Cell Proliferation/physiology , Cortical Bone/pathology , Fractures, Bone/complications , Pain , Periosteum/pathology , Adaptor Proteins, Signal Transducing , Analysis of Variance , Animals , Bone Density , Bromodeoxyuridine/metabolism , Glycoproteins/immunology , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Osteogenesis/drug effects , Pain/etiology , Pain/pathology , Pain/prevention & control
14.
Pain ; 157(11): 2605-2616, 2016 11.
Article in English | MEDLINE | ID: mdl-27482630

ABSTRACT

Bone is one of the leading sites of metastasis for frequently diagnosed malignancies, including those arising in the breast, prostate and lung. Although these cancers develop unnoticed and are painless in their primary sites, bone metastases result in debilitating pain. Deeper investigation of this pain may reveal etiology and lead to early cancer detection. Cancer-induced bone pain (CIBP) is inadequately managed with current standard-of-care analgesics and dramatically diminishes patient quality of life. While CIBP etiology is multifaceted, elevated levels of glutamate, an excitatory neurotransmitter, in the bone-tumor microenvironment may drive maladaptive nociceptive signaling. Here, we establish a relationship between the reactive nitrogen species peroxynitrite, tumor-derived glutamate, and CIBP. In vitro and in a syngeneic in vivo model of breast CIBP, murine mammary adenocarcinoma cells significantly elevated glutamate via the cystine/glutamate antiporter system xc. The well-known system xc inhibitor sulfasalazine significantly reduced levels of glutamate and attenuated CIBP-associated flinching and guarding behaviors. Peroxynitrite, a highly reactive species produced in tumors, significantly increased system xc functional expression and tumor cell glutamate release. Scavenging peroxynitrite with the iron and mangano-based porphyrins, FeTMPyP and SRI10, significantly diminished tumor cell system xc functional expression, reduced femur glutamate levels and mitigated CIBP. In sum, we demonstrate how breast cancer bone metastases upregulate a cystine/glutamate co-transporter to elevate extracellular glutamate. Pharmacological manipulation of peroxynitrite or system xc attenuates CIBP, supporting a role for tumor-derived glutamate in CIBP and validating the targeting of system xc as a novel therapeutic strategy for the management of metastatic bone pain.


Subject(s)
Adenocarcinoma/complications , Bone Neoplasms/complications , Breast Neoplasms/metabolism , Cancer Pain/metabolism , Glutamic Acid/metabolism , Sulfasalazine/pharmacology , Adenocarcinoma/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antiporters/pharmacology , Bone Neoplasms/pathology , Breast Neoplasms/secondary , Calcium-Binding Proteins/metabolism , Cancer Pain/drug therapy , Cancer Pain/etiology , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Metalloporphyrins/pharmacology , Mice , Mice, Inbred BALB C , Peroxynitrous Acid/metabolism , Time Factors
15.
Article in English | MEDLINE | ID: mdl-27186076

ABSTRACT

INTRODUCTION: Cannabinoid compounds, both nonspecific as well as agonists selective for either cannabinoid receptor 1 (CB1) or cannabinoid receptor 2 (CB2), have been shown to modulate the tumor microenvironment by inducing apoptosis in tumor cells in several model systems. The mechanism of this modulation remains only partially delineated, and activity induced via the CB1 and CB2 receptors may be distinct despite significant sequence homology and structural similarity of ligands. METHODS: The CB2-selective agonist JWH-015 was used to investigate mechanisms downstream of CB2 activation in mouse and human breast cancer cell lines in vitro and in a murine mammary tumor model. RESULTS: JWH-015 treatment significantly reduced primary tumor burden and metastasis of luciferase-tagged murine mammary carcinoma 4T1 cells in immunocompetent mice in vivo. Furthermore, JWH-015 reduced the viability of murine 4T1 and human MCF7 mammary carcinoma cells in vitro by inducing apoptosis. JWH-015-mediated reduction of breast cancer cell viability was not dependent on Gαi signaling in vitro or modified by classical pharmacological blockade of CB1, GPR55, TRPV1, or TRPA1 receptors. JWH-015 effects were calcium dependent and induced changes in MAPK/ERK signaling. CONCLUSION: The results of this work characterize the actions of a CB2-selective agonist on breast cancer cells in a syngeneic murine model representing how a clinical presentation of cancer progression and metastasis may be significantly modulated by a G-protein-coupled receptor.

16.
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
17.
J Histochem Cytochem ; 64(3): 179-89, 2016 03.
Article in English | MEDLINE | ID: mdl-26701970

ABSTRACT

Sclerostin is a 24-kDa secreted glycoprotein that has been identified as a negative modulator of new bone formation and may play a major role in age-related decline in skeletal function. Although serum levels of sclerostin markedly increase with age, relatively little is known about whether cells in the skeleton change their expression of sclerostin with aging. Using immunohistochemistry and confocal microscopy, we explored sclerostin immunoreactivity (sclerostin-IR) in the femurs of 4-, 9-, and 24-month-old adult C3H/HeJ male mice. In the femur, the only two cell types that expressed detectable levels of sclerostin-IR were bone osteocytes and articular cartilage chondrocytes. At three different sites along the diaphysis of the femur, only a subset of osteocytes expressed sclerostin-IR and the percentage of osteocytes that expressed sclerostin-IR increased from approximately 36% to 48% in 4- vs. 24-month-old mice. In marked contrast, in the same femurs, there were ~40% fewer hypertrophic chondrocytes of articular cartilage that expressed sclerostin-IR when comparing 24- vs. 4-month-old mice. Understanding the mechanism(s) that drive these divergent changes in sclerostin-IR may provide insight into understanding and treating the age-related decline of the skeleton.


Subject(s)
Aging/metabolism , Bone and Bones/cytology , Cartilage, Articular/cytology , Chondrocytes/metabolism , Glycoproteins/analysis , Osteocytes/metabolism , Adaptor Proteins, Signal Transducing , Animals , Bone Density , Immunohistochemistry , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C3H
18.
Pain ; 156(9): 1692-1702, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25993548

ABSTRACT

Tumor cells frequently metastasize to bone where they can generate cancer-induced bone pain (CIBP) that can be difficult to fully control using available therapies. Here, we explored whether PLX3397, a high-affinity small molecular antagonist that binds to and inhibits phosphorylation of colony-stimulating factor-1 receptor, the tyrosine-protein kinase c-Kit, and the FMS-like tyrosine kinase 3, can reduce CIBP. These 3 targets all regulate the proliferation and function of a subset of the myeloid cells including macrophages, osteoclasts, and mast cells. Preliminary experiments show that PLX3397 attenuated inflammatory pain after formalin injection into the hind paw of the rat. As there is an inflammatory component in CIBP, involving macrophages and osteoclasts, the effect of PLX3397 was explored in a prostate model of CIBP where skeletal pain, cancer cell proliferation, tumor metastasis, and bone remodeling could be monitored in the same animal. Administration of PLX3397 was initiated on day 14 after prostate cancer cell injection when the tumor was well established, and tumor-induced bone remodeling was first evident. Over the next 6 weeks, sustained administration of PLX3397 attenuated CIBP behaviors by approximately 50% and was equally efficacious in reducing tumor cell growth, formation of new tumor colonies in bone, and pathological tumor-induced bone remodeling. Developing a better understanding of potential effects that analgesic therapies have on the tumor itself may allow the development of therapies that not only better control the pain but also positively impact disease progression and overall survival in patients with bone cancer.


Subject(s)
Aminopyridines/therapeutic use , Analgesics/therapeutic use , Bone Neoplasms/complications , Pain/drug therapy , Pain/etiology , Pyrroles/therapeutic use , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Bone Neoplasms/secondary , Bone and Bones/pathology , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Dogs , Formaldehyde/toxicity , Male , Mice , Mice, Nude , Neoplasm Transplantation , Pain Measurement/drug effects , Prostatic Neoplasms/pathology , Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley
19.
J Neurosci ; 35(16): 6307-17, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25904784

ABSTRACT

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.


Subject(s)
Dopaminergic Neurons/physiology , Posterior Horn Cells/physiology , Receptors, Dopamine D1/physiology , Receptors, Dopamine D5/physiology , Receptors, Neurokinin-1/physiology , Animals , Benzazepines/pharmacology , Carrageenan/pharmacology , Dinoprostone/metabolism , Dinoprostone/pharmacology , Dopaminergic Neurons/drug effects , Hyperalgesia/chemically induced , Interleukin-6/pharmacology , Male , Mice , Posterior Horn Cells/drug effects , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D5/agonists , Receptors, Dopamine D5/antagonists & inhibitors , Serotonergic Neurons/physiology , Sulpiride/pharmacology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...