Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Adv Biol (Weinh) ; 6(9): e2200190, 2022 09.
Article in English | MEDLINE | ID: mdl-35925599

ABSTRACT

Oral squamous cell carcinoma (OSCC) patients suffer from poor survival due to metastasis or locoregional recurrence, processes that are both facilitated by perineural invasion (PNI). OSCC has higher rates of PNI than other cancer subtypes, with PNI present in 80% of tumors. Despite the impact of PNI on oral cancer prognosis and pain, little is known about the genes that drive PNI, which in turn drive pain, invasion, and metastasis. In this study, clinical data, preclinical, and in vitro models are leveraged to elucidate the role of neurotrophins in OSCC metastasis, PNI, and pain. The expression data in OSCC patients with metastasis, PNI, or pain demonstrate dysregulation of neurotrophin genes. TrkA and nerve growth factor receptor (NGFR) are focused, two receptors that are activated by NGF, a neurotrophin expressed at high levels in OSCC. It is demonstrated that targeted knockdown of these two receptors inhibits proliferation and invasion in an in vitro and preclinical model of OSCC, and metastasis, PNI, and pain. It is further determined that TrkA knockdown alone inhibits thermal hyperalgesia, whereas NGFR knockdown alone inhibits mechanical allodynia. Collectively the results highlight the ability of OSCC to co-opt different components of the neurotrophin pathway in metastasis, PNI, and pain.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Carcinoma, Squamous Cell/genetics , Humans , Mouth Neoplasms/genetics , Neoplasm Invasiveness/genetics , Neoplasm Recurrence, Local , Neoplastic Processes , Nerve Growth Factors , Nerve Tissue Proteins , Pain , Receptor Protein-Tyrosine Kinases , Receptor, Nerve Growth Factor , Receptor, trkA , Receptors, Nerve Growth Factor/genetics , Squamous Cell Carcinoma of Head and Neck
2.
Sci Rep ; 10(1): 20832, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33257729

ABSTRACT

Metastasis reduces survival in oral cancer patients and pain is their greatest complaint. We have shown previously that oral cancer metastasis and pain are controlled by the endothelin axis, which is a pathway comprised of the endothelin A and B receptors (ETAR and ETBR). In this study we focus on individual genes of the pathway, demonstrating that the endothelin axis genes are methylated and dysregulated in cancer tissue. Based on these findings in patients, we hypothesize that ETAR and ETBR play dichotomous roles in oral carcinogenesis and pain, such that ETAR activation and silenced ETBR expression result in increased carcinogenesis and pain. We test a treatment strategy that targets the dichotomous functions of the two receptors by inhibiting ETAR with macitentan, an ETAR antagonist approved for treatment of pulmonary hypertension, and re-expressing the ETBR gene with adenovirus transduction, and determine the treatment effect on cancer invasion (i.e., metastasis), proliferation and pain in vitro and in vivo. We demonstrate that combination treatment of macitentan and ETBR gene therapy inhibits invasion, but not proliferation, in cell culture and in a mouse model of tongue cancer. Furthermore, the treatment combination produces an antinociceptive effect through inhibition of endothelin-1 mediated neuronal activation, revealing the analgesic potential of macitentan. Our treatment approach targets a pathway shown to be dysregulated in oral cancer patients, using gene therapy and repurposing an available drug to effectively treat both oral cancer metastasis and pain in a preclinical model.


Subject(s)
Endothelins/genetics , Mouth Neoplasms/therapy , Neoplasm Metastasis/therapy , Adult , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Endothelins/metabolism , Endothelins/physiology , Female , HeLa Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mouth Neoplasms/metabolism , Neoplasm Invasiveness/genetics , Pain/metabolism , Pain/physiopathology , Pain Management/methods , Pyrimidines/pharmacology , Receptor, Endothelin A/genetics , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/genetics , Receptor, Endothelin B/metabolism , Sulfonamides/pharmacology
3.
Sci Rep ; 7(1): 9181, 2017 08 23.
Article in English | MEDLINE | ID: mdl-28835637

ABSTRACT

We propose a new mechanism of sensory modulation through cutaneous dopaminergic signalling. We hypothesize that dopaminergic signalling contributes to differential cutaneous sensitivity in darker versus lighter pigmented humans and mouse strains. We show that thermal and mechanical cutaneous sensitivity is pigmentation dependent. Meta-analyses in humans and mice, along with our own mouse behavioural studies, reveal higher thermal sensitivity in pigmented skin relative to less-pigmented or albino skin. We show that dopamine from melanocytes activates the D1-like dopamine receptor on primary sensory neurons. Dopaminergic activation increases expression of the heat-sensitive TRPV1 ion channel and reduces expression of the mechanically-sensitive Piezo2 channel; thermal threshold is lower and mechanical threshold is higher in pigmented skin.


Subject(s)
Dopamine/metabolism , Monophenol Monooxygenase/metabolism , Signal Transduction , Skin Physiological Phenomena , Skin Pigmentation , Animals , Humans , Melanocytes/metabolism , Mice , Receptors, Dopamine D1/metabolism , Sensory Receptor Cells/metabolism , Sensory Thresholds , Temperature
4.
J Pain ; 18(9): 1046-1059, 2017 09.
Article in English | MEDLINE | ID: mdl-28456745

ABSTRACT

Cancer patients in pain require high doses of opioids and quickly become opioid-tolerant. Previous studies have shown that chronic cancer pain as well as high-dose opioid use lead to mu-opioid receptor downregulation. In this study we explore downregulation of the mu-opioid receptor gene (OPRM1), as a mechanism for opioid tolerance in the setting of opioid use for cancer pain. We demonstrate in a cohort of 84 cancer patients that high-dose opioid use correlates with OPRM1 hypermethylation in peripheral leukocytes of these patients. We then reverse-translate our clinical findings by creating a mouse cancer pain model; we create opioid tolerance in the mouse cancer model to mimic opioid tolerance in the cancer patients. Using this model we determine the functional significance of OPRM1 methylation on cancer pain and opioid tolerance. We focus on 2 main cells within the cancer microenvironment: the cancer cell and the neuron. We show that targeted re-expression of mu-opioid receptor on cancer cells inhibits mechanical and thermal hypersensitivity, and prevents opioid tolerance, in the mouse model. The resultant analgesia and protection against opioid tolerance are likely due to preservation of mu-opioid receptor expression on the cancer-associated neurons. PERSPECTIVE: We demonstrate that epigenetic regulation of OPRM1 contributes to opioid tolerance in cancer patients, and that targeted gene therapy could treat cancer-induced nociception and opioid tolerance in a mouse cancer model.


Subject(s)
Analgesics, Opioid/therapeutic use , Cancer Pain/drug therapy , Cancer Pain/metabolism , DNA Methylation , Drug Tolerance/genetics , Receptors, Opioid, mu/metabolism , Adult , Aged , Analgesics, Opioid/pharmacology , Animals , Cancer Pain/genetics , Cell Line, Tumor , Cohort Studies , Drug Tolerance/physiology , Epigenesis, Genetic , Female , Humans , Male , Mice, Inbred BALB C , Middle Aged , Morphine/pharmacology , Morphine/therapeutic use , Neoplasm Transplantation , Neoplasms/genetics , Neoplasms/physiopathology , Nociceptive Pain/drug therapy , Nociceptive Pain/genetics , Nociceptive Pain/metabolism , Opioid-Related Disorders/genetics , Opioid-Related Disorders/metabolism , Pain Management/methods , Patient Education as Topic , Pharmacogenomic Testing , Receptors, Opioid, mu/genetics
5.
Pain ; 158(2): 240-251, 2017 02.
Article in English | MEDLINE | ID: mdl-28092646

ABSTRACT

Virus-mediated gene delivery shows promise for the treatment of chronic pain. However, viral vectors have cytotoxicity. To avoid toxicities and limitations of virus-mediated gene delivery, we developed a novel nonviral hybrid vector: HIV-1 Tat peptide sequence modified with histidine and cysteine residues combined with a cationic lipid. The vector has high transfection efficiency with little cytotoxicity in cancer cell lines including HSC-3 (human tongue squamous cell carcinoma) and exhibits differential expression in HSC-3 (∼45-fold) relative to HGF-1 (human gingival fibroblasts) cells. We used the nonviral vector to transfect cancer with OPRM1, the µ-opioid receptor gene, as a novel method for treating cancer-induced pain. After HSC-3 cells were transfected with OPRM1, a cancer mouse model was created by inoculating the transfected HSC-3 cells into the hind paw or tongue of athymic mice to determine the analgesic potential of OPRM1 transfection. Mice with HSC-3 tumors expressing OPRM1 demonstrated significant antinociception compared with control mice. The effect was reversible with local naloxone administration. We quantified ß-endorphin secretion from HSC-3 cells and showed that HSC-3 cells transfected with OPRM1 secreted significantly more ß-endorphin than control HSC-3 cells. These findings indicate that nonviral delivery of the OPRM1 gene targeted to the cancer microenvironment has an analgesic effect in a preclinical cancer model, and nonviral gene delivery is a potential treatment for cancer pain.


Subject(s)
Cancer Pain/therapy , Carcinoma, Squamous Cell/complications , Genetic Therapy/methods , Receptors, Opioid, mu/metabolism , Tongue Neoplasms/complications , Animals , Cancer Pain/metabolism , Cancer Pain/pathology , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Disease Models, Animal , Fibromatosis, Gingival/genetics , Fibromatosis, Gingival/metabolism , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Receptors, Opioid, mu/genetics , Tongue Neoplasms/genetics , Transfection
6.
Pain ; 156(5): 923-930, 2015 May.
Article in English | MEDLINE | ID: mdl-25734995

ABSTRACT

More than half of all cancer patients have significant pain during the course of their disease. The strategic localization of TMPRSS2, a membrane-bound serine protease, on the cancer cell surface may allow it to mediate signal transduction between the cancer cell and its extracellular environment. We show that TMPRSS2 expression is not only dramatically increased in the primary cancers of patients but TMPRSS2 immunopositivity is also directly correlated with cancer pain severity in these patients. TMPRSS2 induced proteolytic activity, activated trigeminal neurons, and produced marked mechanical hyperalgesia when administered into the hind paw of wild-type mice but not PAR2-deficient mice. Coculture of human cancer cells with murine trigeminal neurons demonstrated colocalization of TMPRSS2 with PAR2. These results point to a novel role for a cell membrane-anchored mediator in cancer pain, as well as pain in general.


Subject(s)
Head and Neck Neoplasms/complications , Pain/metabolism , Receptor, PAR-2/metabolism , Serine Endopeptidases/metabolism , Signal Transduction/drug effects , Animals , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/physiopathology , Humans , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pain/physiopathology , Proteolysis/drug effects , Receptor, PAR-2/deficiency , Receptor, PAR-2/genetics , Severity of Illness Index , Trigeminal Nucleus, Spinal/drug effects , Trigeminal Nucleus, Spinal/metabolism
7.
J Neurophysiol ; 113(9): 3345-55, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25787958

ABSTRACT

Differential thermal nociception across inbred mouse strains has genetic determinants. Thermal nociception is largely attributed to the heat/capsaicin receptor transient receptor potential vanilloid 1 (TRPV1); however, the contribution of this channel to the genetics of thermal nociception has not been revealed. In this study we compared TRPV1 expression levels and electrophysiological properties in primary sensory neurons and thermal nociceptive behaviors between two (C57BL/6 and BALB/c) inbred mouse strains. Using immunofluorescence and patch-clamp physiology methods, we demonstrated that TRPV1 expression was significantly higher in isolectin B4 (IB4)-positive trigeminal sensory neurons of C57BL/6 relative to BALB/c; the expression in IB4-negative neurons was similar between the strains. Furthermore, using electrophysiological cell classification (current signature method), we showed differences between the two strains in capsaicin sensitivity in IB4-positive neuronal cell types 2 and 13, which were previously reported as skin nociceptors. Otherwise electrophysiological membrane properties of the classified cell types were similar in the two mouse strains. In publicly available nocifensive behavior data and our own behavior data from the using the two mouse strains, C57BL/6 exhibited higher sensitivity to heat stimulation than BALB/c, independent of sex and anatomical location of thermal testing (the tail, hind paw, and whisker pad). The TRPV1-selective antagonist JNJ-17203212 inhibited thermal nociception in both strains; however, removing IB4-positive trigeminal sensory neurons with IB4-conjugated saporin inhibited thermal nociception on the whisker pad in C57BL/6 but not in BALB/c. These results suggest that TRPV1 expression levels in IB4-positive type 2 and 13 neurons contributed to differential thermal nociception in skin of C57BL/6 compared with BALB/c.


Subject(s)
Gene Expression Regulation , Glycoproteins/metabolism , Hyperalgesia/physiopathology , Nociception/physiology , Sensory Receptor Cells/metabolism , Skin/innervation , TRPV Cation Channels/genetics , Aminopyridines/pharmacology , Animals , Biophysical Phenomena/drug effects , Capsaicin/adverse effects , Female , Hyperalgesia/chemically induced , Membrane Potentials/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pain Measurement , Pain Threshold/physiology , Piperazines/pharmacology , Sensory Receptor Cells/classification , Sensory Receptor Cells/drug effects , Species Specificity , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Trigeminal Ganglion/cytology
8.
PLoS One ; 9(11): e112880, 2014.
Article in English | MEDLINE | ID: mdl-25391133

ABSTRACT

Cisplatin resistance in head and neck squamous cell carcinoma (HNSCC) reduces survival. In this study we hypothesized that methylation of key genes mediates cisplatin resistance. We determined whether a demethylating drug, decitabine, could augment the anti-proliferative and apoptotic effects of cisplatin on SCC-25/CP, a cisplatin-resistant tongue SCC cell line. We showed that decitabine treatment restored cisplatin sensitivity in SCC-25/CP and significantly reduced the cisplatin dose required to induce apoptosis. We then created a xenograft model with SCC-25/CP and determined that decitabine and cisplatin combination treatment resulted in significantly reduced tumor growth and mechanical allodynia compared to control. To establish a gene classifier we quantified methylation in cancer tissue of cisplatin-sensitive and cisplatin-resistant HNSCC patients. Cisplatin-sensitive and cisplatin-resistant patient tumors had distinct methylation profiles. When we quantified methylation and expression of genes in the classifier in HNSCC cells in vitro, we showed that decitabine treatment of cisplatin-resistant HNSCC cells reversed methylation and gene expression toward a cisplatin-sensitive profile. The study provides direct evidence that decitabine restores cisplatin sensitivity in in vitro and in vivo models of HNSCC. Combination treatment of cisplatin and decitabine significantly reduces HNSCC growth and HNSCC pain. Furthermore, gene methylation could be used as a biomarker of cisplatin-resistance.


Subject(s)
Antineoplastic Agents/therapeutic use , Azacitidine/analogs & derivatives , Carcinoma, Squamous Cell/drug therapy , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Head and Neck Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Animals , Azacitidine/therapeutic use , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA Methylation/drug effects , DNA Methylation/genetics , Decitabine , Drug Resistance, Neoplasm/genetics , Female , Head and Neck Neoplasms/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Transcriptome/drug effects , Transcriptome/genetics , Xenograft Model Antitumor Assays , Young Adult
9.
Clin Cancer Res ; 20(18): 4882-4893, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24963050

ABSTRACT

PURPOSE: In this study, we evaluated the analgesic potential of demethylating drugs on oral cancer pain. Although demethylating drugs could affect expression of many genes, we focused on the mu-opioid receptor (OPRM1) gene pathway, because of its role in pain processing. We determined the antinociceptive effect of OPRM1 re-expression in a mouse oral cancer model. EXPERIMENTAL DESIGN: Using a mouse oral cancer model, we determined whether demethylating drugs produced antinociception through re-expression of OPRM1. We then re-expressed OPRM1 with adenoviral transduction and determined if, and by what mechanism, OPRM1 re-expression produced antinociception. To determine the clinical significance of OPRM1 on cancer pain, we quantified OPRM1 methylation in painful cancer tissues and nonpainful contralateral normal tissues of patients with oral cancer, and nonpainful dysplastic tissues of patients with oral dysplasia. RESULTS: We demonstrated that OPRM1 was methylated in cancer tissue, but not normal tissue, of patients with oral cancer, and not in dysplastic tissues from patients with oral dysplasia. Treatment with demethylating drugs resulted in mechanical and thermal antinociception in the mouse cancer model. This behavioral change correlated with OPRM1 re-expression in the cancer and associated neurons. Similarly, adenoviral-mediated OPRM1 re-expression on cancer cells resulted in naloxone-reversible antinociception. OPRM1 re-expression on oral cancer cells in vitro increased ß-endorphin secretion from the cancer, and decreased activation of neurons that were treated with cancer supernatant. CONCLUSION: Our study establishes the regulatory role of methylation in cancer pain. OPRM1 re-expression in cancer cells produces antinociception through cancer-mediated endogenous opioid secretion. Demethylating drugs have an analgesic effect that involves OPRM1.


Subject(s)
Analgesics, Opioid/pharmacology , Carcinoma, Squamous Cell/complications , Mouth Neoplasms/complications , Pain/drug therapy , Pain/etiology , Receptors, Opioid, mu/genetics , Animals , Antineoplastic Agents/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cytidine/analogs & derivatives , Cytidine/pharmacology , DNA Methylation , Decitabine , Disease Models, Animal , Heterografts , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Neurons/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transduction, Genetic , Trigeminal Ganglion/metabolism
10.
J Neurosci ; 32(41): 14178-83, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-23055487

ABSTRACT

Targeted therapy to prevent the progression from acute to chronic pain in cancer patients remains elusive. We developed three novel cancer models in mice that together recapitulate the anatomical, temporal, and functional characteristics of acute and chronic head and neck cancer pain in humans. Using pharmacologic and genetic approaches in these novel cancer models, we identified the interaction between protease-activated receptor 2 (PAR2) and serine proteases to be of central importance. We show that serine proteases such as trypsin induce acute cancer pain in a PAR2-dependent manner. Chronic cancer pain is associated with elevated serine proteases in the cancer microenvironment and PAR2 upregulation in peripheral nerves. Serine protease inhibition greatly reduces the severity of persistent cancer pain in wild-type mice, but most strikingly, the development of chronic cancer pain is prevented in PAR2-deficient mice. Our results demonstrate a direct role for PAR2 in acute cancer pain and suggest that PAR2 upregulation may favor the development and maintenance of chronic cancer pain. Targeting the PAR2-serine protease interaction is a promising approach to the treatment of acute cancer pain and prevention of chronic cancer pain.


Subject(s)
Acute Pain/metabolism , Chronic Pain/metabolism , Disease Models, Animal , Head and Neck Neoplasms/metabolism , Receptor, PAR-2/physiology , Acute Pain/enzymology , Acute Pain/genetics , Animals , Cell Line, Tumor , Chronic Pain/enzymology , Chronic Pain/genetics , Female , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
11.
Anticancer Res ; 32(4): 1163-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22493345

ABSTRACT

We previously showed that within primary tumors there exist subpopulations of cells expressing stem cell markers. Using immunofluorescence and western blotting, we examined the expression of stem cell markers tumor-rejection antigen 1-60 (TRA1-60) and octamer-binding transcription factor 3/ 4 (OCT3/4) to determine their relationship with cell invasiveness. Six human oral cancer cell lines were examined and a direct correlation was found between expression of these stem cell markers and invasion. Poor expression of E-cadherin and increased expression of N-cadherin was also found in TRA1-60- and OCT3/4- expressing cells. Phosphorylation of the major signaling molecule mitogen activated protein kinase (MAPK) was greatest in the TRA1-60- and OCT3/4- expressing cells. These results suggest that expression of specific stem cell markers in tumors may help guide a clinician's choice of treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Mouth Neoplasms/pathology , Neoplasm Invasiveness , Cadherins/metabolism , Cell Line, Tumor , Enzyme Activation , Humans , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/metabolism
12.
J Pain ; 13(6): 524-31, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22483679

ABSTRACT

UNLABELLED: Cancer patients often suffer from pain and most will be prescribed µ-opioids. µ-opioids are not satisfactory in treating cancer pain and are associated with multiple debilitating side effects. Recent studies show that µ and δ opioid receptors are separately expressed on IB4 (-) and IB4 (+) neurons, which control thermal and mechanical pain, respectively. In this study we investigated IB4 (+) and IB4 (-) neurons in mechanical and thermal hypersensitivity in an orthotopic mouse oral cancer model. We used a δ opioid receptor agonist and a P2X(3) antagonist to target IB4 (+) neurons and to demonstrate that this subset plays a key role in cancer-induced mechanical allodynia, but not in thermal hyperalgesia. Moreover, selective removal of IB4 (+) neurons using IB4-saporin impacts cancer-induced mechanical but not thermal hypersensitivity. Our results demonstrate that peripherally administered pharmacological agents targeting IB4 (+) neurons, such as a selective δ-opioid receptor agonist or P2X(3) antagonist, might be useful in treating oral cancer pain. PERSPECTIVE: To clarify the mechanisms of oral cancer pain, we examined the differential role of IB4 (+) and IB4 (-) neurons. Characterization of these 2 subsets of putative nociceptors is important for further development of effective clinical cancer pain relief.


Subject(s)
Analgesics/pharmacology , Hyperalgesia/physiopathology , Lectins/metabolism , Neoplasms, Experimental/complications , Nociception/drug effects , Animals , Carcinoma, Squamous Cell/complications , Cell Line, Tumor , Disease Models, Animal , Humans , Hyperalgesia/etiology , Hyperalgesia/metabolism , Male , Mice , Mice, Inbred C57BL , Mouth Neoplasms/complications , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/physiopathology , Nociception/physiology , Pain/drug therapy , Pain/etiology , Pain/metabolism , Pain/physiopathology , Pain Threshold/drug effects , Pain Threshold/physiology , Receptors, Opioid, mu/metabolism
13.
J Calif Dent Assoc ; 40(12): 921-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23362664

ABSTRACT

Mucocutaneous melanoma has a five-year survival rate of less than 10 percent. The alphavbeta3 integrin promotes invasion, which requires actin reorganization by cofilin. The authors previously showed that cofilin and alphavbeta3 promote invasion. K1735 melanoma has several clones, each with different levels of alphavbeta3. The authors found that expression of alphavbeta3 suppresses activation of RhoA thus inhibiting LIMK1 phosphorylation of cofilin. This indicates that alphavbeta3 integrin suppresses the RhoA/ ROCK/LIMK1 pathway.


Subject(s)
Integrin alphaVbeta3/physiology , Lim Kinases/physiology , Melanoma/pathology , Signal Transduction/physiology , rho GTP-Binding Proteins/physiology , Actin Depolymerizing Factors/metabolism , Actin Depolymerizing Factors/physiology , Amides/pharmacology , Animals , Blotting, Western , Cell Line, Tumor , Cell Membrane/ultrastructure , Cell Nucleus/ultrastructure , Cytoskeleton/ultrastructure , Enzyme Activation , Enzyme Inhibitors/pharmacology , Genetic Vectors/genetics , Humans , Immunoprecipitation , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Lim Kinases/metabolism , Mice , Microscopy, Fluorescence , Neoplasm Invasiveness , Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/physiology , Phosphorylation , Pyridines/pharmacology , Transfection , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
14.
Pain ; 152(10): 2323-2332, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21782343

ABSTRACT

Endothelin-1 is a vasoactive peptide that activates both the endothelin A (ET(A)) and endothelin B (ET(B)) receptors, and is secreted in high concentrations in many different cancer environments. Although ET(A) receptor activation has an established nociceptive effect in cancer models, the role of ET(B) receptors on cancer pain is controversial. EDNRB, the gene encoding the ET(B) receptor, has been shown to be hypermethylated and transcriptionally silenced in many different cancers. In this study we demonstrate that EDNRB is heavily methylated in human oral squamous cell carcinoma lesions, which are painful, but not methylated in human oral dysplasia lesions, which are typically not painful. ET(B) mRNA expression is reduced in the human oral squamous cell carcinoma lesions as a consequence of EDNRB hypermethylation. Using a mouse cancer pain model, we show that ET(B) receptor re-expression attenuates cancer-induced pain. These findings identify EDNRB methylation as a novel regulatory mechanism in cancer-induced pain and suggest that demethylation therapy targeted at the cancer microenvironment has the potential to thwart pain-producing mechanisms at the source, thus freeing patients of systemic analgesic toxicity.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/therapy , DNA Methylation/genetics , Facial Pain/etiology , Facial Pain/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/therapy , Receptor, Endothelin B/genetics , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis
15.
Mol Cancer Ther ; 10(9): 1667-76, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21750223

ABSTRACT

Cancers often cause excruciating pain and rapid weight loss, severely reducing quality of life in cancer patients. Cancer-induced pain and cachexia are often studied and treated independently, although both symptoms are strongly linked with chronic inflammation and sustained production of proinflammatory cytokines. Because nerve growth factor (NGF) plays a cardinal role in inflammation and pain, and because it interacts with multiple proinflammatory cytokines, we hypothesized that NGF acts as a key endogenous molecule involved in the orchestration of cancer-related inflammation. NGF might be a molecule common to the mechanisms responsible for clinically distinctive cancer symptoms such as pain and cachexia as well as cancer progression. Here we reported that NGF was highly elevated in human oral squamous cell carcinoma tumors and cell cultures. Using two validated mouse cancer models, we further showed that NGF blockade decreased tumor proliferation, nociception, and weight loss by orchestrating proinflammatory cytokines and leptin production. NGF blockade also decreased expression levels of nociceptive receptors TRPV1, TRPA1, and PAR-2. Together, these results identified NGF as a common link among proliferation, pain, and cachexia in oral cancer. Anti-NGF could be an important mechanism-based therapy for oral cancer and its related symptoms.


Subject(s)
Cachexia/etiology , Carcinoma, Squamous Cell/complications , Carcinoma, Squamous Cell/metabolism , Mouth Neoplasms/complications , Mouth Neoplasms/metabolism , Nerve Growth Factor/metabolism , Pain/etiology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Nerve Growth Factor/antagonists & inhibitors , Pain/drug therapy , Pain Measurement/drug effects , RNA, Messenger/metabolism , Receptor, PAR-2/metabolism , Staining and Labeling , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism , Weight Loss/drug effects , Xenograft Model Antitumor Assays
16.
Anticancer Res ; 31(4): 1205-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21508366

ABSTRACT

This study shows that the expression of the extracellular matrix metalloproteinase inducer (EMMPRIN) in oral squamous cell carcinoma cells (SCC) depends upon activation of the Src Family kinaseFyn; and that EMMPRIN and ß6 form a complex that requires active Fyn and the full length ß6 integrin cytoplasmic domain. Fyn is also important for matrix remodeling as it regulates both matrix type 1 metalloproteinase (MT1-MMP) and tissue inhibitor of metalloproteinase-1 and -2 (TIMP1/2). The tumor promoter/suppressor caveolin-1, which associates with MT1-MMP, also requires FYN activation for expression. Lastly, EMMPRIN expression can act as a readout for the mitogen-activated protein kinase (MAPK) pathway, since when MAPK is blocked, so is the expression of EMMPRIN. In oral cancer, the activation of FYN occurs post ß6 integrin ligand binding. That the activation of FYN drives EMMPRIN expression and several important pathways associated with invasive oral SCC is now demonstrated.


Subject(s)
Basigin/metabolism , Carcinoma, Squamous Cell/metabolism , Gene Expression Regulation, Neoplastic , Mouth Neoplasms/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Blotting, Western , Caveolin 1/metabolism , Cell Line, Tumor , Humans , Immunoprecipitation , Matrix Metalloproteinase 14/metabolism , Mitogen-Activated Protein Kinases/metabolism
17.
Anticancer Res ; 30(7): 2591-6, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20682987

ABSTRACT

Oral squamous cell carcinoma (SCC) is an aggressive tumor with a poor 5-year survival rate. Oral SCC can undergo epithelial to mesenchymal transition (EMT). We previously showed that the epithelial integrin alphavbeta6 complexes with Fyn kinase in oral SCC to promote EMT. Using immunofluorescence microscopy and Western blotting, we evaluated whether the expression of specific markers of EMT were influenced by modulating serum concentration (ie. growth factors). The SCC cultures were grown under contrasting levels of serum. In low serum (1%), Fyn promoted EMT; whereas suppression of Fyn kinase promoted the epithelial phenotype. However, when the SCC cells were grown in 10% serum, activation of Fyn had the reverse effect. Lastly, cell migration was evaluated under low serum conditions (1% FBS). Activation of Fyn promoted SCC cell migration and its suppression thwarted SCC migration toward FN. These results indicate that the activation of Fyn kinase as well as local growth factor concentration modulate EMT in oral SCC.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/enzymology , Mouth Neoplasms/pathology , Proto-Oncogene Proteins c-fyn/biosynthesis , Blotting, Western , Cadherins/biosynthesis , Cadherins/metabolism , Cell Communication/physiology , Cell Movement/physiology , Culture Media , Enzyme Activation , Epithelial Cells/pathology , Humans , Keratins/biosynthesis , Mesoderm/pathology , Microscopy, Fluorescence , Proto-Oncogene Proteins c-fyn/metabolism , Serum , Signal Transduction
18.
Anticancer Res ; 29(6): 2043-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19528463

ABSTRACT

Oral squamous cell carcinoma (SCC) is composed of a heterogeneous population of cells which range anywhere from epithelial to mesenchymal in phenotype. Several oral cancer specimens with antibodies to TRA160, a marker of pluripotent cells, were screened. Compared with the well differentiated lesions, pluripotent cells were more numerous in specimens from poorly differentiated tumors. In vitro, the expression of TRA160 was much greater in invasive oral SCC9beta6 cells compared with the poorly invasive SCC9SN or SCC9beta6D1 cells, which express a truncated beta6. In vitro, pluripotent cells were instrumental in aggressively closing an experimental wound assay. Lastly, TRA-1-60+/beta6+ tumor cells which formed vascular-like structures in vivo were identified. SCC9beta6 cells formed interconnecting channels, whereas SCC9SN cells did not in an in vitro Matrigel angiogenesis assay. The results of this study clearly demonstrated the differential distribution of pluripotent stem cells in oral SCC and that the beta6 integrin may be an important regulatory component of the pluripotent phenotype.


Subject(s)
Antigens, Neoplasm/metabolism , Carcinoma, Squamous Cell/metabolism , Integrins/metabolism , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Antigens, Surface/metabolism , Blotting, Western , Carcinoma, Squamous Cell/pathology , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Neoplastic Stem Cells/pathology , Proteoglycans/metabolism , Tumor Cells, Cultured , Wound Healing
19.
Anticancer Res ; 29(1): 125-30, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19331141

ABSTRACT

In this study, we evaluated whether the forced expression of beta6 integrin would modulate the epithelial to mesenchymal transition (EMT). When the full length beta6 integrin was expressed in poorly invasive squamous cell carcinoma SCC9 cells, the resulting SCC9/6 cells acquired a fibroblast-like morphology, increased expression of the mesenchymal marker vimentin and reduced expression of the epithelial markers keratin and E-cadherin. SCC9beta6D1 cells, which express a truncated form of beta6 subunit lacking the C-terminal 11 amino acids (AA), retained their epithelial morphology and did not alter vimentin or E-cadherin expression. This suggests that the full-length beta6 subunit can induce EMT in oral SCC cells. We previously showed that expression of beta6 increases both MMP-3 activation and tenascin-C expression and we now show that both molecules are MEK dependent. These results also demonstrate that the terminal 11 AA of beta6 contain information important for establishing an epithelial to mesenchymal transition.


Subject(s)
Antigens, Neoplasm/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Integrins/metabolism , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Antigens, Neoplasm/biosynthesis , Cadherins/biosynthesis , Carcinoma, Squamous Cell/enzymology , Cell Line, Tumor , Enzyme Activation , Epithelial Cells/pathology , Humans , Integrins/antagonists & inhibitors , Integrins/biosynthesis , Keratins/biosynthesis , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase Inhibitors , Mesoderm/pathology , Mouth Neoplasms/enzymology , Tenascin/metabolism , Vimentin/biosynthesis
20.
J Calif Dent Assoc ; 37(12): 869-74, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20066875

ABSTRACT

Oral cancer is the sixth most frequent cancer worldwide. Prognosis for these patients remains poor. Recently, the epidermal growth factor receptor has been targeted as an adjunct to radiotherapy and surgery with limited success. The authors now present data suggesting that the alphanubeta6 integrin, which is a marker for aggressive oral cancer, may regulate epidermal growth factor receptor expression. The authors suggest perhaps targeting both alphanubeta6 and EGFR may provide additional benefits.


Subject(s)
Antigens, Neoplasm/physiology , Carcinoma, Squamous Cell/pathology , ErbB Receptors/physiology , Integrins/physiology , Mouth Neoplasms/pathology , Antibodies, Monoclonal , Antigens, Neoplasm/analysis , Biomarkers, Tumor/analysis , Blotting, Western , Cell Line, Tumor , Cell Membrane/pathology , ErbB Receptors/analysis , Extracellular Matrix/pathology , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Humans , Immunohistochemistry , Integrins/analysis , Neoplasm Invasiveness , Proto-Oncogene Proteins c-fyn/physiology , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...