Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Pain ; 158(8): 1609-1621, 2017 08.
Article in English | MEDLINE | ID: mdl-28715356

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) continues to be one of the deadliest human malignancies and is associated with excruciating pain, which is a serious complication and severely impacts the quality of life in patients. In human patients, poor survival prognosis is linked to remarkable remodeling of intrapancreatic nerves, which, in turn, is correlated to increased pain intensity. Understanding mechanisms underlying pain associated with PDAC has been hampered by the lack of animal models which replicate all germane aspects of the disease and importantly, enable analyses of pain associated with PDAC. In this study, we describe an immunocompetent orthotopic mouse model of PDAC involving intrapancreatic growth of K8484 mouse PDAC cells, which reliably exhibits a large number of key characteristics of human PDAC, including its unique histopathology and neuroplastic changes. We observed that tumor-bearing mice demonstrated significant abdominal mechanical hypersensitivity to von Frey stimuli as well as on-going pain in the conditioned place preference paradigm. Moreover, a myriad of other behavioral tests revealed that indicators of overall well-being were significantly reduced in tumor-bearing mice as compared to sham mice. Morphological and immunohistochemical analyses revealed structural remodeling in several different types of sensory and autonomic nerve fibers. Finally, perineural invasion of tumor cells, a cardinal manifestation in human PDAC, was also observed in our orthotopic mouse model. Thus, we describe a validated tumor model for quantitatively testing hypersensitivity and pain in PDAC, which lays a crucial basis for interrogating tumor-nerve interactions and the molecular and cellular mechanisms underlying pain in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/complications , Pain/physiopathology , Pancreatic Neoplasms/complications , Animals , Biomarkers, Tumor/analysis , Cell Line, Tumor , Cell Proliferation/physiology , Mice , Neuronal Plasticity/physiology
2.
Cancer Cell ; 27(6): 780-96, 2015 Jun 08.
Article in English | MEDLINE | ID: mdl-26058077

ABSTRACT

Cancer pain is a debilitating disorder and a primary determinant of the poor quality of life. Here, we report a non-vascular role for ligands of the Vascular Endothelial Growth Factor (VEGF) family in cancer pain. Tumor-derived VEGF-A, PLGF-2, and VEGF-B augment pain sensitivity through selective activation of VEGF receptor 1 (VEGFR1) expressed in sensory neurons in human cancer and mouse models. Sensory-neuron-specific genetic deletion/silencing or local or systemic blockade of VEGFR1 prevented tumor-induced nerve remodeling and attenuated cancer pain in diverse mouse models in vivo. These findings identify a therapeutic potential for VEGFR1-modifying drugs in cancer pain and suggest a palliative effect for VEGF/VEGFR1-targeting anti-angiogenic tumor therapies.


Subject(s)
Neoplasms/pathology , Pain/metabolism , Sensory Receptor Cells/metabolism , Vascular Endothelial Growth Factor Receptor-1/biosynthesis , Angiogenesis Inhibitors/pharmacology , Animals , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasms/metabolism , Pain/drug therapy , Pain/pathology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/pathology , Up-Regulation , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors
4.
EMBO Mol Med ; 5(11): 1740-58, 2013 11.
Article in English | MEDLINE | ID: mdl-24039159

ABSTRACT

Cancer pain remains a major challenge and there is an urgent demand for the development of specific mechanism-based therapies. Various diseases are associated with unique signatures of expression of microRNAs (miRNAs), which reveal deep insights into disease pathology. Using a comprehensive approach combining genome-wide miRNA screening, molecular and in silico analyses with behavioural approaches in a clinically relevant model of metastatic bone-cancer pain in mice, we now show that tumour-induced conditions are associated with a marked dysregulation of 57 miRNAs in sensory neurons corresponding to tumour-affected areas. By establishing protocols for interference with disease-induced miRNA dysregulation in peripheral sensory neurons in vivo, we functionally validate six dysregulated miRNAs as significant modulators of tumour-associated hypersensitivity. In silico analyses revealed that their predicted targets include key pain-related genes and we identified Clcn3, a gene encoding a chloride channel, as a key miRNA target in sensory neurons, which is functionally important in tumour-induced nociceptive hypersensitivity in vivo. Our results provide new insights into endogenous gene regulatory mechanisms in cancer pain and open up attractive and viable therapeutic options.


Subject(s)
Genome , MicroRNAs/genetics , Neoplasms/genetics , Pain/genetics , Animals , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred C3H , MicroRNAs/metabolism , Neoplasms/metabolism , Pain/metabolism , Sensory Receptor Cells/metabolism
5.
Pain ; 154(12): 2801-2812, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23973358

ABSTRACT

The rich diversity of lipids and the specific signalling pathways they recruit provides tremendous scope for modulation of biological functions. Lysophosphatidylinositol (LPI) is emerging as a key modulator of cell proliferation, migration, and function, and holds important pathophysiological implications due to its high levels in diseased tissues, such as in cancer. Here we report a novel role for LPI in sensitization of peripheral sensory neurons, which was evident as exaggerated sensitivity to painful and innocuous pressure. Histopathological analyses indicated lack of involvement of myelin pathology and immune cell recruitment by LPI. Using pharmacological and conditional genetic tools in mice, we delineated receptor-mediated from non-receptor-mediated effects of LPI and we observed that GPR55, which functions as an LPI receptor when heterologously expressed in mammalian cells, only partially mediates LPI-induced actions in the context of pain sensitization in vivo; we demonstrate that, in vivo, LPI functions by activating Gα(13) as well as Gα(q/11) arms of G-protein signalling in sensory neurons. This study thus reports a novel pathophysiological function for LPI and elucidates underlying molecular mechanisms.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Lysophospholipids/physiology , Nociception/physiology , Sensory Receptor Cells/physiology , Signal Transduction/physiology , Animals , Dose-Response Relationship, Drug , GTP-Binding Proteins/metabolism , Lysophospholipids/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nociception/drug effects , Phospholipids/pharmacology , Phospholipids/physiology , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects
6.
Mol Pain ; 6: 18, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20346101

ABSTRACT

BACKGROUND: On-going pain is one of the most debilitating symptoms associated with a variety of chronic pain disorders. An understanding of mechanisms underlying on-going pain, i.e. stimulus-independent pain has been hampered so far by a lack of behavioural parameters which enable studying it in experimental animals. Ultrasound vocalizations (USVs) have been proposed to correlate with pain evoked by an acute activation of nociceptors. However, literature on the utility of USVs as an indicator of chronic pain is very controversial. A majority of these inconsistencies arise from parameters confounding behavioural experiments, which include novelty, fear and stress due to restrain, amongst others. RESULTS: We have developed an improved assay which overcomes these confounding factors and enables studying USVs in freely moving mice repetitively over several weeks. Using this improved assay, we report here that USVs increase significantly in mice with bone metastases-induced cancer pain or neuropathic pain for several weeks, in comparison to sham-treated mice. Importantly, analgesic drugs which are known to alleviate tumour pain or neuropathic pain in human patients significantly reduce USVs as well as mechanical allodynia in corresponding mouse models. CONCLUSIONS: We show that studying USVs and mechanical allodynia in the same cohort of mice enables comparing the temporal progression of on-going pain (i.e. stimulus-independent pain) and stimulus-evoked pain in these clinically highly-relevant forms of chronic pain.


Subject(s)
Neoplasms/pathology , Neuralgia/pathology , Pain Measurement/methods , Ultrasonics , Animals , Chronic Disease , Female , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...