Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Prostate ; 78(2): 128-139, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29131367

ABSTRACT

BACKGROUND: Nerves are key factors in prostate cancer (PCa), but the functional role of innervation in prostate cancer is poorly understood. PCa induced neurogenesis and perineural invasion (PNI), are associated with aggressive disease. METHOD: We denervated rodent prostates chemically and physically, before orthotopically implanting cancer cells. We also performed a human neoadjuvant clinical trial using botulinum toxin type A (Botox) and saline in the same patient, before prostatectomy. RESULT: Bilateral denervation resulted in reduced tumor incidence and size in mice. Botox treatment in humans resulted in increased apoptosis of cancer cells in the Botox treated side. A similar denervation gene array profile was identified in tumors arising in denervated rodent prostates, in spinal cord injury patients and in the Botox treated side of patients. Denervation induced exhibited a signature gene profile, indicating translation and bioenergetic shutdown. Nerves also regulate basic cellular functions of non-neoplastic epithelial cells. CONCLUSION: Nerves play a role in the homeostasis of normal epithelial tissues and are involved in prostate cancer tumor survival. This study confirms that interactions between human cancer and nerves are essential to disease progression. This work may make a major impact in general cancer treatment strategies, as nerve/cancer interactions are likely important in other cancers as well. Targeting the neural microenvironment may represent a therapeutic approach for the treatment of human prostate cancer.


Subject(s)
Botulinum Toxins, Type A/pharmacology , Denervation/methods , Prostate , Prostatic Neoplasms , Acetylcholine Release Inhibitors/pharmacology , Animals , Disease Models, Animal , Disease Progression , Energy Metabolism , Male , Mice , Neoplasm Invasiveness , Prostate/innervation , Prostate/pathology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Tumor Burden , Tumor Microenvironment/physiology
2.
Hum Pathol ; 52: 182-9, 2016 06.
Article in English | MEDLINE | ID: mdl-26980040

ABSTRACT

Pancreatic cancer (PaCA) is a deadly disease with few systemic therapeutic options. The head of the pancreas is the most innervated part and most common location of cancer. However, little is known about the contribution of the nerve-cancer interaction to facilitate pancreatic progression. To quantify PaCA axonogenesis, we used a 3-dimensional in vitro neurogenesis model. In addition, neurogenesis in human PaCA was analyzed using PGP9.5 immunohistochemistry, deconvolution imaging, and image segmentation and analysis. There was a significant increase of the total area of neurites in the in vitro coculture with dorsal root ganglia group than control. The nerve density in PaCA tissue was significantly higher than normal pancreatic tissue. To study the functional role of nerves in PaCA, male athymic nude (Nu-Nu) mice were divided into 3 groups: (A) animals were coinjected with MIA PaCa-2 cells and 20U/kg weight units of Botulinum toxin (Botox) (n=10); (B) first injected with Botox and 6weeks later MIA PaCa-2 cancer cells (n=4); and (C) control animals were injected with equivalent amounts of saline fluid (n=9). Animals were sacrificed 6weeks later. Tumor size and apoptotic count (terminal deoxynucleotidyl transferase dUTP nick-end labeling) were measured. Tumor size was decreased and apoptotic rate increased in Botox-treated PaCA. Our data indicate that neural microenvironment may play an important role in the progression of PaCA. It may lead to novel nerve-targeted coadjuvant therapies for PaCA.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Ganglia, Spinal/pathology , Neurogenesis , Neurons/pathology , Pancreatic Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers, Tumor/metabolism , Botulinum Toxins, Type A/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Humans , Male , Mice, Nude , Mice, Transgenic , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Time Factors , Tumor Burden , Tumor Microenvironment , Ubiquitin Thiolesterase/metabolism , Xenograft Model Antitumor Assays
3.
Hum Pathol ; 45(7): 1365-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24768607

ABSTRACT

Perineural invasion is a symbiotic relationship between cancer cells and nerves and is most frequently seen in "neurotropic" cancers such as prostate cancer. It results in increased perineural space cancer cell growth and decreased apoptosis and induces nerve growth. Tissue microarrays were constructed from 640 radical prostatectomy specimens with prostate cancer. The perineural diameter was measured as previously described. Multiple biomarkers have been previously performed on this tissue microarray cohort, and all data were kept in the same database. The biomarker results database was queried for correlations between perineural invasion diameter and tissue biomarkers. Increased perineural invasion diameter correlated with increased proliferation of prostate cancer cells and with apoptosis. It also correlated with proteins involved in survival pathways such as nuclear factor κB, c-Myc, phosphorylated AKT, and its downstream effector FHKR, but not with GSK. Unlike nerve density, it did not correlate with decreased PTEN expression. Increased perineural invasion diameter was associated with higher levels of hormonal receptors such as androgen receptor, but not estrogen receptor. Also associated with perineural invasion diameter were coregulators and corepressors including SRC1 and TIF2. Perineural invasion diameter had the strongest correlation with tumor volume (ρ = 0.579, P = .000), not identified with nerve density. These data demonstrate that perineural invasion has the same biologic correlations as neural density. However, we found a distinct and very strong correlation with increased tumor volume. These data confirm that perineural invasion is the ultimate and most successful interaction between cancer cells and nerve fibers, resulting in increased tumor growth.


Subject(s)
Neoplasm Invasiveness/pathology , Peripheral Nerves/pathology , Prostate/pathology , Prostatic Neoplasms/pathology , Apoptosis , Biomarkers, Tumor/metabolism , Humans , Male , Peripheral Nerves/metabolism , Prostate/metabolism , Prostate/surgery , Prostatectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/surgery , Retrospective Studies , Tumor Microenvironment
4.
Hum Pathol ; 45(7): 1358-64, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24767770

ABSTRACT

Cancer-related axonogenesis and neurogenesis are recently described biologic phenomena. Our previously published data showed that nerve density and the number of neurons in the parasympathetic ganglia are increased in prostate cancer (PCa) and associated with aggressive disease. Tissue microarrays were constructed from 640 radical prostatectomy specimens with PCa. Anti-protein gene product 9.5 (PGP 9.5) antibodies were used to identify and quantify nerve density. Protein expression was objectively analyzed using deconvolution imaging, image segmentation, and image analysis. Data were correlated with clinicopathological variables and tissue biomarkers available in our database. Nerve density, as measured by PGP 9.5 expression, had a weak but significant positive correlation with the lymph node status (ρ = 0.106; P = .0275). By Cox univariate analysis, PGP 9.5 was a predictor of time to biochemical recurrence, but not on multivariate analysis. Increased nerve density correlated with increased proliferation of PCa cells. It also correlated with expression of proteins involved in survival pathways (Phosphorylated alpha serine/threonine-protein kinase, NFκB, GSK-2, PIM-2, c-Myc, SKP-2, SRF, P27n, PTEN), with increased levels of hormonal regulation elements (androgen receptor, estrogen receptor α), and coregulators and repressors (SRC-1, SRC-2, AIB-1, DAX). Axonogenesis is a recently described phenomenon of paramount importance in the biology of PCa. Although the degree of axonogenesis is predictive of aggressive behavior in PCa, it does not add to the information present in current models on multivariate analysis. We present data that corroborate that axonogenesis is involved in biologic processes such as proliferation of PCa, through activation of survival pathways and interaction with hormonal regulation.


Subject(s)
Axons/physiology , Neurogenesis/physiology , Prostate/metabolism , Prostatic Neoplasms/physiopathology , Axons/metabolism , Axons/pathology , Gene Expression Regulation, Neoplastic , Humans , Male , Prognosis , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Time Factors , Ubiquitin Thiolesterase/metabolism
5.
Clin Cancer Res ; 19(22): 6101-11, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24097862

ABSTRACT

BACKGROUND: Semaphorin 4F (S4F) has roles in embryologic axon guidance and is expressed in adults. S4F is involved in cancer-induced neurogenesis. METHODS: Prostate cells were transfected with S4F retrovirus. Cells and controls were used for a bromodeoxyuridine (BrdUrd) incorporation assay (proliferation) and in vitro scratch and Matrigel Transwell chamber invasion assay (migration). Monoclonal antibodies were developed using baculovirus-expressed recombinant GST-S4F and used to immunostain tissue microarrays. Slides were imaged using deconvolution and analyzed using tissue segmentation. Data were correlated with clinicopathologic parameters, other biomarkers and survival analysis conducted. Heterogeneity of S4F expression was analyzed with unsupervised clustering algorithms. RESULTS: Proliferation rates measured by BrdUrd incorporation were higher in all S4F-transfected cells. S4F overexpression was associated with increased motility of the cancer cells. S4F expression was overexpressed in high-grade prostatic intraepithelial neoplasia/prostate cancer than normal epithelium. S4F expression correlated with seminal vesicle invasion. Patients with high values of S4F in prostate cancer cytoplasm are at significantly higher risk of biochemical recurrence, by univariate and multivariate analyses. S4F cytoplasmic expression in prostate cancer cells also correlates with nerve density in prostate cancer and perineural invasion diameter. Correlations were identified with NF-κB and inversely with apoptosis in perineural invasion. CONCLUSION: These data show that S4F is significantly involved in human prostate cancer progression. S4F is a key regulator of the interactions between nerves in the tumor microenvironment and cancer cells. Because of the importance of cancer nerve interaction in the biology of cancer and its clinical implication, S4F can be considered a major therapeutic target. Clin Cancer Res; 19(22); 6101-11. ©2013 AACR.


Subject(s)
Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuroepithelial Cells/metabolism , Neurogenesis/genetics , Prostatic Neoplasms/genetics , Semaphorins/metabolism , Apoptosis/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Male , Membrane Proteins/genetics , NF-kappa B/genetics , Neoplasm Invasiveness/genetics , Nerve Tissue Proteins/genetics , Seminal Vesicles/cytology , Seminal Vesicles/pathology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...