Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Cancers (Basel) ; 14(12)2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35740624

ABSTRACT

Hepatocellular carcinoma (HCC), a highly aggressive liver cancer, is a leading cause of cancer-related death. Tumor Treating Fields (TTFields) are electric fields that exert antimitotic effects on cancerous cells. The aims of the current research were to test the efficacy of TTFields in HCC, explore the underlying mechanisms, and investigate the possible combination of TTFields with sorafenib, one of the few front-line treatments for patients with advanced HCC. HepG2 and Huh-7D12 human HCC cell lines were treated with TTFields at various frequencies to determine the optimal frequency eliciting maximal cell count reduction. Clonogenic, apoptotic effects, and autophagy induction were measured. The efficacy of TTFields alone and with concomitant sorafenib was tested in cell cultures and in an orthotopic N1S1 rat model. Tumor volume was examined at the beginning and following 5 days of treatment. At study cessation, tumors were weighed and examined by immunohistochemistry to assess autophagy and apoptosis. TTFields were found in vitro to exert maximal effect at 150 kHz, reducing cell count and colony formation, increasing apoptosis and autophagy, and augmenting the effects of sorafenib. In animals, TTFields concomitant with sorafenib reduced tumor weight and volume fold change, and increased cases of stable disease following treatment versus TTFields or sorafenib alone. While each treatment alone elevated levels of autophagy relative to control, TTFields concomitant with sorafenib induced a significant increase versus control in tumor ER stress and apoptosis levels, demonstrating increased stress under the multimodal treatment. Overall, TTFields treatment demonstrated efficacy and enhanced the effects of sorafenib for the treatment of HCC in vitro and in vivo, via a mechanism involving induction of autophagy.

3.
Lung Cancer ; 160: 99-110, 2021 10.
Article in English | MEDLINE | ID: mdl-34482104

ABSTRACT

OBJECTIVES: Tumor Treating Fields (TTFields) are low intensity, intermediate frequency, alternating electric fields with antimitotic effects on cancerous cells. TTFields concomitant with pemetrexed and a platinum agent are approved in the US and EU as first line therapy for unresectable, locally advanced or metastatic malignant pleural mesothelioma (MPM). The goal of the current study was to characterize the mechanism of action of TTFields in MPM cell lines and animal models. METHODS: Human MPM cell lines MSTO-211H and NCI-H2052 were treated with TTFields to determine the frequency that elicits maximal cytotoxicity. The effect of TTFields on DNA damage and repair, and the cytotoxic effect of TTFields in combination with cisplatin and/or pemetrexed were examined. Efficacy of TTFields concomitant with cisplatin and pemetrexed was evaluated in orthotopic IL-45 and subcutaneous RN5 murine models. RESULTS: TTFields at a frequency of 150 kHz demonstrated the highest cytotoxicity to MPM cells. Application of 150 kHz TTFields resulted in increased formation of DNA double strand breaks, elevated expression of DNA damage induced cell cycle arrest proteins, and reduced expression of Fanconi Anemia (FA)-BRCA DNA repair pathway proteins. Co-treatment of TTFields with cisplatin or pemetrexed significantly increased treatment efficacy versus each modality alone, with additivity and synergy exhibited by the TTFields-pemetrexed and TTFields-cisplatin combinations, respectively. In animal models, tumor volume was significantly lower for the TTFields-cisplatin-pemetrexed combination compared to control, accompanied by increased DNA damage within the tumor. CONCLUSION: This research demonstrated that the efficacy of TTFields for the treatment of MPM is associated with reduced expression of FA-BRCA pathway proteins and increased DNA damage. This mechanism of action is consistent with the observed synergism for TTFields-cisplatin vs additivity for TTFields-pemetrexed, as cisplatin-induced DNA damage is repaired via the FA-BRCA pathway.


Subject(s)
Fanconi Anemia , Lung Neoplasms , Mesothelioma, Malignant , Animals , Cisplatin , Humans , Lung Neoplasms/drug therapy , Mice , Pemetrexed
4.
Front Oncol ; 11: 670809, 2021.
Article in English | MEDLINE | ID: mdl-34249709

ABSTRACT

BACKGROUND: Tumor Treating Fields (TTFields) therapy is a non-invasive, loco-regional, anti-mitotic treatment modality that targets rapidly dividing cancerous cells, utilizing low intensity, alternating electric fields at cancer-cell-type specific frequencies. TTFields therapy is approved for the treatment of newly diagnosed and recurrent glioblastoma (GBM) in the US, Europe, Israel, Japan, and China. The favorable safety profile of TTFields in patients with GBM is partially attributed to the low rate of mitotic events in normal, quiescent brain cells. However, specific safety evaluations are warranted at locations with known high rates of cellular proliferation, such as the torso, which is a primary site of several of the most aggressive malignant tumors. METHODS: The safety of delivering TTFields to the torso of healthy rats at 150 or 200 kHz, which were previously identified as optimal frequencies for treating multiple torso cancers, was investigated. Throughout 2 weeks of TTFields application, animals underwent daily clinical examinations, and at treatment cessation blood samples and internal organs were examined. Computer simulations were performed to verify that the targeted internal organs of the torso were receiving TTFields at therapeutic intensities (≥ 1 V/cm root mean square, RMS). RESULTS: No treatment-related mortality was observed. Furthermore, no significant differences were observed between the TTFields-treated and control animals for all examined safety parameters: activity level, food and water intake, stools, motor neurological status, respiration, weight, complete blood count, blood biochemistry, and pathological findings of internal organs. TTFields intensities of 1 to 2.5 V/cm RMS were confirmed for internal organs within the target region. CONCLUSIONS: This research demonstrates the safety of therapeutic level TTFields at frequencies of 150 and 200 kHz when applied as monotherapy to the torso of healthy rats.

5.
Cancer Immunol Immunother ; 69(7): 1191-1204, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32144446

ABSTRACT

Tumor-treating fields (TTFields) are alternating electric fields in a specific frequency range (100-300 kHz) delivered to the human body through transducer arrays. In this study, we evaluated whether TTFields-mediated cell death can elicit antitumoral immunity and hence would be effectively combined with anti-PD-1 therapy. We demonstrate that in TTFields-treated cancer cells, damage-associated molecular patterns including high-mobility group B1 and adenosine triphosphate are released and calreticulin is exposed on the cell surface. Moreover, we show that TTFields treatment promotes the engulfment of cancer cells by dendritic cells (DCs) and DCs maturation in vitro, as well as recruitment of immune cells in vivo. Additionally, our study demonstrates that the combination of TTFields with anti-PD-1 therapy results in a significant decline of tumor volume and increase in the percentage of tumor-infiltrating leukocytes in two tumor models. In orthotopic lung tumors, these infiltrating leukocytes, specifically macrophages and DCs, showed elevated expression of PD-L1. Compatibly, cytotoxic T-cells isolated from these tumors demonstrated increased production of IFN-γ. In colon cancer tumors, T-cells infiltration was significantly increased following long treatment duration with TTFields plus anti-PD-1. Collectively, our results suggest that TTFields therapy can induce anticancer immune response. Furthermore, we demonstrate robust efficacy of concomitant application of TTFields and anti-PD-1 therapy. These data suggest that integrating TTFields with anti-PD-1 therapy may further enhance antitumor immunity, hence achieve better tumor control.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Hepatocellular/therapy , Carcinoma, Lewis Lung/therapy , Electric Stimulation Therapy/methods , Immunogenic Cell Death , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Apoptosis , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Proliferation , Combined Modality Therapy , Female , Humans , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Cell Death Dis ; 9(11): 1074, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30341282

ABSTRACT

Tumor Treating Fields (TTFields), an approved treatment modality for glioblastoma, are delivered via non-invasive application of low-intensity, intermediate-frequency, alternating electric fields. TTFields application leads to abnormal mitosis, aneuploidy, and increased cell granularity, which are often associated with enhancement of autophagy. In this work, we evaluated whether TTFields effected the regulation of autophagy in glioma cells. We found that autophagy is upregulated in glioma cells treated with TTFields as demonstrated by immunoblot analysis of the lipidated microtubule-associated protein light chain 3 (LC3-II). Fluorescence and transmission electron microscopy demonstrated the presence of LC3 puncta and typical autophagosome-like structures in TTFields-treated cells. Utilizing time-lapse microscopy, we found that the significant increase in the formation of LC3 puncta was specific to cells that divided during TTFields application. Evaluation of selected cell stress parameters revealed an increase in the expression of the endoplasmic reticulum (ER) stress marker GRP78 and decreased intracellular ATP levels, both of which are indicative of increased proteotoxic stress. Pathway analysis demonstrated that TTFields-induced upregulation of autophagy is dependent on AMP-activated protein kinase (AMPK) activation. Depletion of AMPK or autophagy-related protein 7 (ATG7) inhibited the upregulation of autophagy in response to TTFields, as well as sensitized cells to the treatment, suggesting that cancer cells utilize autophagy as a resistance mechanism to TTFields. Combining TTFields with the autophagy inhibitor chloroquine (CQ) resulted in a significant dose-dependent reduction in cell growth compared with either TTFields or CQ alone. These results suggest that dividing cells upregulate autophagy in response to aneuploidy and ER stress induced by TTFields, and that AMPK serves as a key regulator of this process.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy , Brain Neoplasms/pathology , Electric Stimulation/methods , Glioblastoma/pathology , Up-Regulation , Adenosine Triphosphate/metabolism , Aneuploidy , Animals , Autophagosomes/metabolism , Autophagy-Related Protein 7/antagonists & inhibitors , Brain Neoplasms/therapy , Cell Line, Tumor , Cell Survival , Electric Stimulation Therapy , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Glioblastoma/therapy , Heat-Shock Proteins/metabolism , Humans , Lysosomes/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Mitosis , Rats , Vascular Endothelial Growth Factor A
7.
J Vis Exp ; (123)2017 05 04.
Article in English | MEDLINE | ID: mdl-28518093

ABSTRACT

Tumor Treating Fields (TTFields) are an effective treatment modality delivered via the continuous, noninvasive application of low-intensity (1-3 V/cm), alternating electric fields in the frequency range of several hundred kHz. The study of TTFields in tissue culture is carried out using the TTFields in vitro application system, which allows for the application of electric fields of varying frequencies and intensities to ceramic Petri dishes with a high dielectric constant (Ɛ > 5,000). Cancerous cell lines plated on coverslips at the bottom of the ceramic Petri dishes are subjected to TTFields delivered in two orthogonal directions at various frequencies to facilitate treatment outcome tests, such as cell counts and clonogenic assays. The results presented in this report demonstrate that the optimal frequency of the TTFields with respect to both cell counts and clonogenic assays is 200 kHz for both ovarian and glioma cells.


Subject(s)
Colony-Forming Units Assay/methods , Electric Stimulation Therapy , Electricity , Glioma/therapy , Ovarian Neoplasms/therapy , Antineoplastic Protocols , Cell Line, Tumor , Female , Humans , Treatment Outcome
8.
Int J Cancer ; 139(12): 2850-2858, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27561100

ABSTRACT

Long-term survival rates for advanced ovarian cancer patients have not changed appreciably over the past four decades; therefore, development of new, effective treatment modalities remains a high priority. Tumor Treating Fields (TTFields), a clinically active anticancer modality utilize low-intensity, intermediate frequency, alternating electric fields. The goal of this study was to evaluate the efficacy of combining TTFields with paclitaxel against ovarian cancer cells in vitro and in vivo. In vitro application of TTFields on human ovarian cancer cell lines led to a significant reduction in cell counts as compared to untreated cells. The effect was found to be frequency and intensity dependent. Further reduction in the number of viable cells was achieved when TTFields treatment was combined with paclitaxel. The in vivo effect of the combined treatment was tested in mice orthotopically implanted with MOSE-LTICv cells. In this model, combined treatment led to a significant reduction in tumor luminescence and in tumor weight as compared to untreated mice. The feasibility of effective local delivery of TTFields to the human abdomen was examined using finite element mesh simulations performed using the Sim4life software. These simulations demonstrated that electric fields intensities inside and in the vicinity of the ovaries of a realistic human computational phantom are about 1 and 2 V/cm pk-pk, respectively, which is within the range of intensities required for TTFields effect. These results suggest that prospective clinical investigation of the combination of TTFields and paclitaxel is warranted.


Subject(s)
Antineoplastic Agents/pharmacology , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Combined Modality Therapy , Disease Models, Animal , Female , Humans , Mice , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/therapy , Treatment Outcome , Xenograft Model Antitumor Assays
9.
Sci Rep ; 5: 18046, 2015 Dec 11.
Article in English | MEDLINE | ID: mdl-26658786

ABSTRACT

Tumor Treating Fields (TTFields) are low intensity, intermediate frequency, alternating electric fields. TTFields are a unique anti-mitotic treatment modality delivered in a continuous, noninvasive manner to the region of a tumor. It was previously postulated that by exerting directional forces on highly polar intracellular elements during mitosis, TTFields could disrupt the normal assembly of spindle microtubules. However there is limited evidence directly linking TTFields to an effect on microtubules. Here we report that TTFields decrease the ratio between polymerized and total tubulin, and prevent proper mitotic spindle assembly. The aberrant mitotic events induced by TTFields lead to abnormal chromosome segregation, cellular multinucleation, and caspase dependent apoptosis of daughter cells. The effect of TTFields on cell viability and clonogenic survival substantially depends upon the cell division rate. We show that by extending the duration of exposure to TTFields, slowly dividing cells can be affected to a similar extent as rapidly dividing cells.


Subject(s)
Chromosome Segregation/physiology , Mitosis/physiology , Neoplasms/pathology , Spindle Apparatus/pathology , Animals , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/physiology , Electricity , Humans , MCF-7 Cells , Microtubules/metabolism , Microtubules/pathology , Neoplasms/metabolism , Rats , Rats, Inbred F344 , Tubulin/metabolism
10.
Autophagy ; 11(12): 2387-9, 2015.
Article in English | MEDLINE | ID: mdl-26571129

ABSTRACT

HPSE (heparanase) is the predominant enzyme in mammals capable of cleaving heparan sulfate, an activity highly implicated in cellular invasion and tumor metastasis. HPSE expression is induced in many types of cancer and increased HPSE levels are most often associated with increased tumor metastasis and reduced patient survival post operation. In addition, HPSE induction is associated with progression of the primary tumors but the mechanism(s) underlying tumor expansion by HPSE have not been sufficiently resolved. Our results establish a role for heparanase in modulating autophagy in normal and malignant cells, thereby conferring growth advantages as well as resistance to chemotherapy.


Subject(s)
Autophagy/physiology , Glucuronidase/physiology , Neoplasm Proteins/physiology , Animals , Female , Humans
11.
Cancer Res ; 75(18): 3946-57, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26249176

ABSTRACT

Heparanase is the only enzyme in mammals capable of cleaving heparan sulfate, an activity implicated in tumor inflammation, angiogenesis, and metastasis. Heparanase is secreted as a latent enzyme that is internalized and subjected to proteolytic processing and activation in lysosomes. Its role under normal conditions has yet to be understood. Here, we provide evidence that heparanase resides within autophagosomes, where studies in heparanase-deficient or transgenic mice established its contributions to autophagy. The protumorigenic properties of heparanase were found to be mediated, in part, by its proautophagic function, as demonstrated in tumor xenograft models of human cancer and through use of inhibitors of the lysosome (chloroquine) and heparanase (PG545), both alone and in combination. Notably, heparanase-overexpressing cells were more resistant to stress and chemotherapy in a manner associated with increased autophagy, effects that were reversed by chloroquine treatment. Collectively, our results establish a role for heparanase in modulating autophagy in normal and malignant cells, thereby conferring growth advantages under stress as well as resistance to chemotherapy. Cancer Res; 75(18); 3946-57. ©2015 AACR.


Subject(s)
Autophagy/physiology , Glucuronidase/physiology , Neoplasm Proteins/physiology , Amino Acids/deficiency , Animals , Antineoplastic Agents/pharmacology , Carcinoma/pathology , Cell Division , Cell Line, Tumor , Cells, Cultured , Chloroquine/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Female , Fibroblasts/enzymology , Glioma/pathology , Heterografts , Humans , Mechanistic Target of Rapamycin Complex 1 , Membrane Lipids/metabolism , Mice , Mice, Knockout , Mice, SCID , Mice, Transgenic , Multiprotein Complexes/metabolism , Phagosomes/enzymology , Pharyngeal Neoplasms/pathology , Phosphatidylethanolamines/metabolism , Rats , TOR Serine-Threonine Kinases/metabolism , Tumor Stem Cell Assay
12.
Semin Thromb Hemost ; 41(2): 244-54, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25682080

ABSTRACT

Heparanase is an endo-ß-glucuronidase that enzymatically cleaves heparan sulfates (HS) and heparan sulfate proteoglycan (HSPG) structures. Heparanase expression levels by tumors were correlated with cell invasion, angiogenic activity, and poor prognosis. Heparanase can also possess pro-tumorigenic effects independent of its enzymatic activity. Using human melanoma MV3 cells, we demonstrate that latent heparanase activates in a tightly temporary-regulated manner the binding function of the integrin very late antigen-4 (VLA-4), an important component in the metastatic spread of melanoma cells. shRNA-mediated knockdown of syndecan-4 (SDC-4) indicated that this proteoglycan is the key element to convey heparanase binding via focal adhesion complex formation, detected by vinculin staining, to an upregulated VLA-4 binding function. This inside-out signaling pathway of VLA-4 involved activated FAK and Akt, but apparently not PKCα/δ. VLA-4, however, appears representative of other integrins which together impact the heparanase/integrin activation axis in tumorigenicity. Biosensor measurements provided an insight as to how heparin can interfere with this activation process. While low-molecular-weight heparin (LMWH) cannot replace heparanase bound to SDC-4, LMWH can compete with SDC-4 binding of heparanase. Since blockade of heparanase by LMWH has functional consequences for reduced VLA-4 binding, latent heparanase appears as a novel, so far unnoticed target of heparin, underlying its antimetastatic activity.


Subject(s)
Drug Delivery Systems , Heparin Lyase/metabolism , Heparin, Low-Molecular-Weight/pharmacology , Integrin alpha4beta1/metabolism , Melanoma/drug therapy , Melanoma/metabolism , Neoplasm Proteins/metabolism , Cell Line, Tumor , Enzyme Activation/drug effects , Enzyme Activation/genetics , Gene Knockdown Techniques , HEK293 Cells , Heparin Lyase/genetics , Humans , Integrin alpha4beta1/genetics , Melanoma/genetics , Melanoma/pathology , Neoplasm Metastasis , Neoplasm Proteins/genetics
13.
Cell Mol Life Sci ; 71(22): 4457-70, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24788042

ABSTRACT

Heparanase activity plays a decisive role in cell dissemination associated with cancer metastasis. Cellular uptake of heparanase is considered a pre-requisite for the delivery of latent 65-kDa heparanase to lysosomes and its subsequent proteolytic processing and activation into 8- and 50-kDa protein subunits by cathepsin L. Heparan sulfate proteoglycans, and particularly syndecan, are instrumental for heparanase uptake and activation, through a process that has been shown to occur independent of rafts. Nevertheless, the molecular mechanism underlying syndecan-mediated internalization outside of rafts is unclear. Here, we examined the role of syndecan-1 cytoplasmic domain in heparanase processing, utilizing deletion constructs lacking the entire cytoplasmic domain (Delta), the conserved (C1 or C2), or variable (V) regions. Heparanase processing was markedly increased following syndecan-1 over-expression; in contrast, heparanase was retained at the cell membrane and its processing was impaired in cells over-expressing syndecan-1 deleted for the entire cytoplasmic tail. We have next revealed that conserved domain 2 (C2) and variable (V) regions of syndecan-1 cytoplasmic tail mediate heparanase processing. Furthermore, we found that syntenin, known to interact with syndecan C2 domain, and α actinin are essential for heparanase processing.


Subject(s)
Actinin/metabolism , Glucuronidase/metabolism , Syndecan-1/metabolism , Syntenins/metabolism , Actinin/antagonists & inhibitors , Actinin/genetics , Animals , Cell Line, Tumor , Cell Membrane/metabolism , HEK293 Cells , Humans , Mice , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering/metabolism , Syndecan-1/chemistry , Syndecan-1/genetics , Syntenins/antagonists & inhibitors , Syntenins/genetics
14.
J Leukoc Biol ; 95(4): 677-88, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24319286

ABSTRACT

Heparanase is an endo-ß-glucuronidase that specifically cleaves the saccharide chains of HSPGs, important structural and functional components of the ECM. Cleavage of HS leads to loss of the structural integrity of the ECM and release of HS-bound cytokines, chemokines, and bioactive angiogenic- and growth-promoting factors. Our previous study revealed a highly significant correlation of HPSE gene SNPs rs4693608 and rs4364254 and their combination with the risk of developing GVHD. We now demonstrate that HPSE is up-regulated in response to pretransplantation conditioning, followed by a gradual decrease thereafter. Expression of heparanase correlated with the rs4693608 HPSE SNP before and after conditioning. Moreover, a positive correlation was found between recipient and donor rs4693608 SNP discrepancy and the time of neutrophil and platelet recovery. Similarly, the discrepancy in rs4693608 HPSE SNP between recipients and donors was found to be a more significant factor for the risk of aGVHD than patient genotype. The rs4693608 SNP also affected HPSE gene expression in LPS-treated MNCs from PB and CB. Possessors of the AA genotype exhibited up-regulation of heparanase with a high ratio in the LPS-treated MNCs, whereas individuals with genotype GG showed down-regulation or no effect on HPSE gene expression. HPSE up-regulation was mediated by TLR4. The study emphasizes the importance of rs4693608 SNP for HPSE gene expression in activated MNCs, indicating a role in allogeneic stem cell transplantation, including postconditioning, engraftment, and GVHD.


Subject(s)
Glucuronidase/genetics , Hematopoietic Stem Cell Transplantation , Lipopolysaccharides/pharmacology , Polymorphism, Single Nucleotide , Transplantation Conditioning , Adolescent , Adult , Aged , Female , Genotype , Glucuronidase/physiology , Graft vs Host Disease/etiology , Graft vs Host Disease/genetics , Humans , Leukocytes, Mononuclear/enzymology , Male , Middle Aged , Toll-Like Receptor 4/physiology , Transplantation, Homologous
15.
J Biol Chem ; 285(36): 28010-9, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20576607

ABSTRACT

Heparanase activity is highly implicated in cell dissemination associated with tumor metastasis, angiogenesis, and inflammation. Heparanase expression is induced in many hematological and solid tumors, associated with poor prognosis. Heparanase homolog, termed heparanase 2 (Hpa2), was cloned based on sequence homology. Detailed characterization of Hpa2 at the biochemical, cellular, and clinical levels has not been so far reported, and its role in normal physiology and pathological disorders is obscure. We provide evidence that unlike heparanase, Hpa2 is not subjected to proteolytic processing and exhibits no enzymatic activity typical of heparanase. Notably, the full-length Hpa2c protein inhibits heparanase enzymatic activity, likely due to its high affinity to heparin and heparan sulfate and its ability to associate physically with heparanase. Hpa2 expression was markedly elevated in head and neck carcinoma patients, correlating with prolonged time to disease recurrence (follow-up to failure; p = 0.006) and inversely correlating with tumor cell dissemination to regional lymph nodes (N-stage; p = 0.03). Hpa2 appears to restrain tumor metastasis, likely by attenuating heparanase enzymatic activity, conferring a favorable outcome of head and neck cancer patients.


Subject(s)
Glucuronidase/antagonists & inhibitors , Glucuronidase/metabolism , Heparitin Sulfate/metabolism , Amino Acid Sequence , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Glucuronidase/chemistry , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Molecular Sequence Data , Neoplasm Metastasis , Protein Binding , Protein Transport
16.
J Cell Sci ; 122(Pt 6): 807-12, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19223395

ABSTRACT

RIC-3 is a member of a conserved family of proteins that affect nicotinic acetylcholine receptor maturation. In yeast and in vitro, BATH-42, a BTB- and MATH-domain-containing protein, interacts with RIC-3. BATH-42 is also known to interact with the CUL-3 ubiquitin ligase complex. Loss of BATH-42 function leads to increased RIC-3 expression and decreased activity of nicotinic acetylcholine receptors in Caenorhabditis elegans vulva muscles. Increased expression of RIC-3 is deleterious for activity and distribution of nicotinic acetylcholine receptors, and thus the effects of BATH-42 loss of function on RIC-3 expression explain the associated reduction in receptor activity. Overexpression of BATH-42 is also detrimental to nicotinic acetylcholine receptor function, leading to decreased pharyngeal pumping. This effect depends on the C-terminus of RIC-3 and on CUL-3. Thus, our work suggests that BATH-42 targets RIC-3 to degradation via CUL-3-mediated ubiquitylation. This demonstrates the importance of regulation of RIC-3 levels, and identifies a mechanism that protects cells from the deleterious effects of excess RIC-3.


Subject(s)
Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Receptors, Nicotinic/metabolism , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/drug effects , Female , Levamisole/pharmacology , Muscles/cytology , Muscles/drug effects , Muscles/metabolism , Mutation/genetics , Pharynx/cytology , Pharynx/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Transport/drug effects , Vulva/cytology , Vulva/drug effects , Vulva/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...