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










Publication year range
1.
Cytotherapy ; 2024 May 10.
Article in English | MEDLINE | ID: mdl-38819362

ABSTRACT

Chimeric antigen receptor cell therapy is a successful immunotherapy for the treatment of blood cancers. However, hurdles in their manufacturing remain including efficient isolation and purification of the T-cell starting material. Herein, we describe a one-step separation based on inertial spiral microfluidics for efficient enrichment of T-cells in B-cell acute lymphoblastic leukemia (ALL) and B-cell chronic lymphocytic leukemia patient's samples. In healthy donors used to optimize the process, the lymphocyte purity was enriched from 65% (SD ± 0.2) to 91% (SD ± 0.06) and T-cell purity was enriched from 45% (SD ± 0.1) to 73% (SD ± 0.02). Leukemic samples had higher starting B-cells compared to the healthy donor samples. Efficient enrichment and recovery of lymphocytes and T-cells were achieved in ALL samples with B-cells, monocytes and leukemic blasts depleted by 80% (SD ± 0.09), 89% (SD ± 0.1) and 74% (SD ± 0.09), respectively, and a 70% (SD ± 0.1) T-cell recovery. Chronic lymphocytic leukemia samples had lower T-cell numbers, and the separation process was less efficient compared to the ALL. This study demonstrates the use of inertial microfluidics for T-cell enrichment and depletion of B-cell blasts in ALL, suggesting its potential to address a key bottleneck of the chimeric antigen receptor-T manufacturing workflow.

2.
R Soc Open Sci ; 11(4): 230370, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38577209

ABSTRACT

There is increasing recognition of the potential pleiotropic effects of melanin pigmentation, particularly on immunity, with reports of variation in haemoparasite infection intensity and immune responses between the morphs of colour-polymorphic bird species. In a population of the black sparrowhawk (Accipiter melanoleucus) in western South Africa, light morphs have a higher haemoparasite infection intensity, but no physiological effects of this are apparent. Here, we investigate the possible effects of haemoparasite infection on telomere length in this species and explore whether relative telomere length is associated with either plumage morph or sex. Using quantitative polymerase chain reaction analysis, we confirmed that dark morphs had a lower haemoparasite infection intensity than light morphs. However, we found no differences in telomere length associated with either the haemoparasite infection status or morph in adults, although males have longer telomeres than females. While differences in haemoparasite intensity between morphs are consistent with pleiotropic effects of melanin pigmentation in the black sparrowhawk, we found no evidence that telomere length was associated with haemoparasite infection. Further work is needed to investigate the implications of possible pleiotropic effects of plumage morph and their potential role in the maintenance of colour polymorphism in this species.

3.
Bioeng Transl Med ; 9(1): e10599, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38193128

ABSTRACT

Blockade of programmed cell death-1 (PD-1) is a transformative immunotherapy. However, only a fraction of patients benefit, and there is a critical need for broad-spectrum checkpoint inhibition approaches that both enhance the recruitment of cytotoxic immune cells in cold tumors and target resistance pathways. Indoleamine 2, 3-dioxygenase (IDO) small molecule inhibitors are promising but suboptimal tumor bioavailability and dose-limiting toxicity have limited therapeutic benefits in clinical trials. This study reports on a nanoformulation of the IDO inhibitor navoximod within polymeric nanoparticles prepared using a high-throughput microfluidic mixing device. Hydrophobic ion pairing addresses the challenging physicochemical properties of navoximod, yielding remarkably high loading (>10%). The nanoformulation efficiently inhibits IDO and, in synergy with PD-1 antibodies improves the anti-cancer cytotoxicity of T-cells, in vitro and in vivo. This study provides new insight into the IDO and PD-1 inhibitors synergy and validates hydrophobic ion pairing as a simple and clinically scalable formulation approach.

4.
J Nanobiotechnology ; 21(1): 102, 2023 Mar 21.
Article in English | MEDLINE | ID: mdl-36945003

ABSTRACT

Disruption of the cell cycle is among the most effective approach to increase tumour cells' radio-sensitivity. However, the presence of dose-limiting side effects hampers the clinical use of tyrosine kinase inhibitors targeting the cell cycle. Towards addressing this challenge, we identified a bosutinib nanoformulation within high density lipoprotein nanoparticles (HDL NPs) as a promising radiosensitiser. Bosutinib is a kinase inhibitor clinically approved for the treatment of chronic myeloid leukemia that possesses radiosensitising properties through cell cycle checkpoint inhibition. We found that a remarkably high bosutinib loading (> 10%) within HDL NPs could be reliably achieved under optimal preparation conditions. The radiosensitisation activity of the bosutinib-HDL nanoformulation was first assessed in vitro in UM-SCC-1 head and neck squamous cell carcinoma (HNSCC) cells, which confirmed efficient disruption of the radiation induced G2/M cell cycle arrest. Interestingly, the bosutinib nanoformulation out-performed free bosutinib, likely because of the specific affinity of HDL NPs with tumour cells. The combination of bosutinib-HDL NPs and radiotherapy significantly controlled tumour growth in an immunocompetent murine HNSCC model. The bosutinib-HDL nanoformulation also enhanced the radiation induced immune response through the polarisation of tumour associated macrophages towards proinflammatory phenotypes.


Subject(s)
Antineoplastic Agents , Head and Neck Neoplasms , Animals , Mice , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Antineoplastic Agents/pharmacology , Aniline Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy
5.
Int J Pharm ; 622: 121845, 2022 Jun 25.
Article in English | MEDLINE | ID: mdl-35613652

ABSTRACT

Immune checkpoint inhibition with antibodies targeting the programmed cell death-1 (PD-1) pathway is a frontline cancer immunotherapy. Driven by the limited response rates and high off-target toxicity associated to monoclonal antibodies, small molecule inhibitors of PD-1 are under active investigation. Glycogen synthase kinase 3 (GSK3) is an up-stream regulator of PD-1 and small molecule GSK3 inhibitors have been shown to effectively reduce T-cell expression of PD-1 receptors. Towards harnessing the potent anticancer effects of GSK3 inhibition, we report here on the development of a nanoformulation within PEG-PLGA nanoparticles of the small molecule GSK3 inhibitor SB415286. The formulation physicochemical properties were optimised using a novel 3D printed microfluidic nanoprecipitation device and a hydrophobic ion pairing approach was used to increase the loading of the drug. The SB415286 nanoformulation efficiently inhibited PD-1 expression in chimeric antigen receptor (CAR)-T cells co-cultured with tumour cells expressing the CAR target, and improved their survival and proliferation. Treatment of the CAR-T cells with nanoformulation also increased the population of memory T-cells. The nanoformulation of small molecule inhibitor of the GSK3 pathway is a promising alternative to antibody-based checkpoint inhibition that warrants further studies.


Subject(s)
Antineoplastic Agents , Programmed Cell Death 1 Receptor , Antineoplastic Agents/therapeutic use , Glycogen , Glycogen Synthase Kinase 3 , Immunotherapy , Programmed Cell Death 1 Receptor/metabolism
6.
J Control Release ; 346: 98-109, 2022 06.
Article in English | MEDLINE | ID: mdl-35447296

ABSTRACT

Preclinical, clinical and epidemiologic studies have established the potent anticancer and radiosensitisation effects of HMG-CoA reductase inhibitors (statins). However, the low bioavailability of oral statin formulations is a key barrier to achieving effective doses within tumour. To address this issue and ascertain the radiosensitisation potential of simvastatin, we developed a parenteral high density lipoprotein nanoparticle (HDL NP) formulation of this commonly used statin. A scalable method for the preparation of the simvastatin-HDL NPs was developed using a 3D printed microfluidic mixer. This enables the production of litre scale amounts of particles with minimal batch to batch variation. Simvastatin-HDL NPs enhanced the radiobiological response in 2D/3D head and neck squamous cell carcinoma (HNSCC) in vitro models. The simvastatin-HDL NPs radiosensitisation was comparable to that of 10 and 5 times higher doses of free drug in 2D and 3D cultures, respectively, which could be partially explained by more efficient cellular uptake of the statin in the nanoformulation as well as by the inherent biological activity of the HDL NPs on the cholesterol pathway. The radiosensitising potency of the simvastatin-HDL nanoformulation was validated in an immunocompetent MOC-1 HNSCC tumour bearing mouse model. This data supports the rationale of repurposing statins through reformulation within HDL NPs. Statins are safe and readily available molecules including as generic, and their use as radiosensitisers could lead to much needed effective and affordable approaches to improve treatment of solid tumours.


Subject(s)
Head and Neck Neoplasms , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Animals , Cholesterol, HDL , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lipoproteins, HDL , Mice , Simvastatin/pharmacology , Simvastatin/therapeutic use , Squamous Cell Carcinoma of Head and Neck/drug therapy
7.
ACS Appl Mater Interfaces ; 14(14): 15881-15893, 2022 Apr 13.
Article in English | MEDLINE | ID: mdl-35357803

ABSTRACT

Intratumoral administration of immune checkpoint inhibitors, such as programmed cell death-1 antibodies (aPD-1), is a promising approach toward addressing both the low patients' responses and high off-target toxicity, but good preclinical results have not translated in phase I clinical studies as significant off-target toxicities were observed. We hypothesized that the nanoformulation of aPD-1 could alter both their loco-regional and systemic distribution following intratumoral administration. To test this hypothesis, we developed an aPD-1 nanoformulation (aPD-1 NPs) and investigated its biodistribution following intratumoral injection in an orthotopic mice model of head and neck cancer. Biodistribution analysis demonstrated a significantly lower distribution in off-target organs of the nanoformulated aPD-1 compared to free antibodies. On the other hand, both aPD-1 NPs and free aPD-1 yielded a significantly higher tumor and tumor draining lymph node accumulation than the systemically administrated free aPD-1 used as the current clinical benchmark. In a set of comprehensive in vitro biological studies, aPD-1 NPs effectively inhibited PD-1 expression on T-cells to a similar extent to free aPD-1 and efficiently potentiated the cytotoxicity of T-cells against head and neck cancer cells in vitro. Further studies are warranted to assess the potential of this intratumoral administration of aPD-1 nanoformulation in alleviating the toxicity and enhancing the tumor efficacy of immune checkpoint inhibitors.


Subject(s)
Head and Neck Neoplasms , Immune Checkpoint Inhibitors , Animals , Antibodies , Head and Neck Neoplasms/drug therapy , Humans , Immunotherapy/methods , Mice , T-Lymphocytes , Tissue Distribution
8.
Int J Pharm ; 617: 121585, 2022 Apr 05.
Article in English | MEDLINE | ID: mdl-35176332

ABSTRACT

Radiotherapy is one of the main treatment options for head and neck cancer patients. However, its clinical efficacy is hindered by both radiation induced side effects and radio-resistance. Radio-sensitising approaches with acceptable toxicity are being actively investigated. Among these, RNA therapeutics have great potentials as radio-sensitisers owing to their ability to target pathways specific to radio-resistance. However, their clinical translation is challenging due to delivery issues. Herein, we report the application of high-density lipoprotein nanoparticle (HDL NPs) as a biocompatible delivery system for a well-established radio-sensitising RNA, miR-34a. A simple/fast microfluidic based technique was used to prepare miR-34a-HDL NPs. Profiling of the radiation response in the UM-SCC-1 head and neck cancer cell line confirmed reduced metabolic activity and increased radiation induced apoptosis upon treatment with miR-34a-HDL NPs. The radio-sensitising properties of miR-34a-HDL NPs were further confirmed in a more biologically relevant co-culture spheroid model of head and neck cancer. Increased apoptotic activity and disrupted cell cycle were induced by miR-34a delivered by HDL NPs. The enhanced radio-biologic effects observed in both 2D and 3D models confirmed the utility of HDL NPs as an efficient delivery system for radio-sensitising RNA.


Subject(s)
Head and Neck Neoplasms , MicroRNAs , Nanoparticles , Cell Line, Tumor , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/radiotherapy , Humans , Lipoproteins, HDL , MicroRNAs/genetics
9.
Adv Biol (Weinh) ; 6(1): e2101018, 2022 01.
Article in English | MEDLINE | ID: mdl-34881810

ABSTRACT

Chimeric antigen receptor T (CAR-T) cell therapy is rapidly becoming a frontline cancer therapy. However, the manufacturing process is time-, labor- and cost-intensive, and it suffers from significant bottlenecks. Many CAR-T products fail to reach the viability release criteria set by regulators for commercial cell therapy products. This results in non-recoupable costs for the manufacturer and is detrimental to patients who may not receive their scheduled treatment or receive out-of-specification suboptimal formulation. It is demonstrated here that inertial microfluidics can, within minutes, efficiently deplete nonviable cells from low-viability CAR-T cell products. The percentage of viable cells increases from 40% (SD ± 0.12) to 71% (SD ± 0.09) for untransduced T cells and from 51% (SD ± 0.12) to 71% (SD ± 0.09) for CAR-T cells, which meets the clinical trials' release parameters. In addition, the processing of CAR-T cells formulated in CryStor yields a 91% reduction in the amount of the cryoprotectant dimethyl sulfoxide. Inertial microfluidic processing has no detrimental effects on the proliferation and cytotoxicity of CAR-T cells. Interestingly, ≈50% of T-regulatory and T-suppressor cells are depleted, suggesting the potential for inertial microfluidic processing to tune the phenotypical composition of T-cell products.


Subject(s)
Receptors, Chimeric Antigen , Cell- and Tissue-Based Therapy , Humans , Immunotherapy, Adoptive , Lymphocyte Count , Microfluidics
10.
Haematologica ; 105(6): 1517-1526, 2020 06.
Article in English | MEDLINE | ID: mdl-31413099

ABSTRACT

Telomerase is a ribonucleoprotein complex that maintains the length and integrity of telomeres, and thereby enables cellular proliferation. Understanding the regulation of telomerase in hematopoietic cells is relevant to the pathogenesis of leukemia, in which telomerase is constitutively activated, as well as bone marrow failure syndromes that feature telomerase insufficiency. Past studies showing high levels of telomerase in human erythroblasts and a prevalence of anemia in disorders of telomerase insufficiency provide the rationale for investigating telomerase regulation in erythroid cells. Here it is shown for the first time that the telomerase RNA-binding protein dyskerin (encoded by DKC1) is dramatically upregulated as human hematopoietic stem and progenitor cells commit to the erythroid lineage, driving an increase in telomerase activity in the presence of limiting amounts of TERT mRNA. It is also shown that upregulation of DKC1 was necessary for expansion of glycophorin A+ erythroblasts and sufficient to extend telomeres in erythroleukemia cells. Chromatin immunoprecipitation and reporter assays implicated GATA1-mediated transcriptional regulation of DKC1 in the modulation of telomerase in erythroid lineage cells. Together these results describe a novel mechanism of telomerase regulation in erythroid cells which contrasts with mechanisms centered on transcriptional regulation of TERT that are known to operate in other cell types. This is the first study to reveal a biological context in which telomerase is upregulated by DKC1 and to implicate GATA1 in telomerase regulation. The results from this study are relevant to hematopoietic disorders involving DKC1 mutations, GATA1 deregulation and/or telomerase insufficiency.


Subject(s)
Cell Cycle Proteins/metabolism , Erythroblasts/metabolism , GATA1 Transcription Factor/metabolism , Nuclear Proteins/metabolism , Telomerase , Cell Cycle Proteins/genetics , GATA1 Transcription Factor/genetics , Humans , Nuclear Proteins/genetics , Telomerase/genetics , Telomerase/metabolism , Up-Regulation
11.
Sci Adv ; 5(10): eaav4409, 2019 10.
Article in English | MEDLINE | ID: mdl-31616780

ABSTRACT

Telomerase is a ribonucleoprotein complex that catalyzes addition of telomeric DNA repeats to maintain telomeres in replicating cells. Here, we demonstrate that the telomerase protein hTERT performs an additional role at telomeres that is independent of telomerase catalytic activity yet essential for telomere integrity and cell proliferation. Short-term depletion of endogenous hTERT reduced the levels of heat shock protein 70 (Hsp70-1) and the telomere protective protein Apollo at telomeres, and induced telomere deprotection and cell cycle arrest, in the absence of telomere shortening. Short-term expression of hTERT promoted colocalization of Hsp70-1 with telomeres and Apollo and reduced numbers of deprotected telomeres, in a manner independent of telomerase catalytic activity. These data reveal a previously unidentified noncanonical function of hTERT that promotes formation of a telomere protective complex containing Hsp70-1 and Apollo and is essential for sustained proliferation of telomerase-positive cancer cells, likely contributing to the known cancer-promoting effects of both hTERT and Hsp70-1.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Neoplasms/metabolism , Telomerase/metabolism , Telomere/metabolism , Cell Line, Tumor , DNA Damage , Gene Expression Regulation , HSP70 Heat-Shock Proteins/genetics , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Neoplasms/genetics , Telomerase/genetics
12.
Article in English | MEDLINE | ID: mdl-31658973

ABSTRACT

Millions of women are exposed simultaneously to antiretroviral drugs (ARVs) and progestin-based hormonal contraceptives. Yet the reciprocal modulation by ARVs and progestins of their intracellular functions is relatively unexplored. We investigated the effects of tenofovir disoproxil fumarate (TDF) and dapivirine (DPV), alone and in the presence of select steroids and progestins, on cell viability, steroid-regulated immunomodulatory gene expression, activation of steroid receptors, and anti-HIV-1 activity in vitro Both TDF and DPV modulated the transcriptional efficacy of a glucocorticoid agonist via the glucocorticoid receptor (GR) in the U2OS cell line. In TZM-bl cells, DPV induced the expression of the proinflammatory interleukin 8 (IL-8) gene while TDF significantly increased medroxyprogesterone acetate (MPA)-induced expression of the anti-inflammatory glucocorticoid-induced leucine zipper (GILZ) gene. However, peripheral blood mononuclear cell (PBMC) and ectocervical explant tissue viability and gene expression results, along with TZM-bl HIV-1 infection data, are reassuring and suggest that TDF and DPV, in combination with dexamethasone (DEX) or MPA, do not reciprocally modulate key biological effects in primary cells and tissue. We show for the first time that TDF induces progestogen-independent activation of the progesterone receptor (PR) in a cell line. The ability of TDF and DPV to influence GR and PR activity suggests that their use may be associated with steroid receptor-mediated off-target effects. This, together with cell line and individual donor gene expression responses in the primary models, raises concerns that reciprocal modulation may cause side effects in a cell- and donor-specific manner in vivo.


Subject(s)
Anti-Retroviral Agents/adverse effects , Anti-Retroviral Agents/pharmacology , Receptors, Steroid/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Humans , Inflammation/metabolism , Peripheral Blood Stem Cells/drug effects , Peripheral Blood Stem Cells/metabolism , Progestins/metabolism , Pyrimidines/adverse effects , Pyrimidines/pharmacology , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/metabolism , Tenofovir/adverse effects , Tenofovir/pharmacology , Transcription Factors/metabolism
13.
Sci Rep ; 9(1): 4334, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30867477

ABSTRACT

The intramuscular progestin-only injectable contraceptive, depo-medroxyprogesterone acetate (DMPA-IM), is more widely used in Sub-Saharan Africa than another injectable contraceptive, norethisterone enanthate (NET-EN). Epidemiological data show a significant 1.4-fold increased risk of HIV-1 acquisition for DMPA-IM usage, while no such association is shown from limited data for NET-EN. We show that MPA, unlike NET, significantly increases R5-tropic but not X4-tropic HIV-1 replication ex vivo in human endocervical and ectocervical explant tissue from pre-menopausal donors, at physiologically relevant doses. Results support a mechanism whereby MPA, unlike NET, acts via the glucocorticoid receptor (GR) to increase HIV-1 replication in cervical tissue by increasing the relative frequency of CD4+ T cells and activated monocytes. We show that MPA, unlike NET, increases mRNA expression of the CD4 HIV-1 receptor and CCR5 but not CXCR4 chemokine receptors, via the GR. However, increased density of CD4 on CD3+ cells was not observed with MPA by flow cytometry of digested tissue. Results suggest that DMPA-IM may increase HIV-1 acquisition in vivo at least in part via direct effects on cervical tissue to increase founder R5-tropic HIV-1 replication. Our findings support differential biological mechanisms and disaggregation of DMPA-IM and NET-EN regarding HIV-1 acquisition risk category for use in high risk areas.


Subject(s)
Cervix Uteri/virology , Contraceptive Agents, Hormonal/pharmacology , HIV-1/pathogenicity , Medroxyprogesterone Acetate/pharmacology , Norethindrone/pharmacology , Contraceptive Agents, Hormonal/administration & dosage , Dose-Response Relationship, Drug , Female , HEK293 Cells , HIV Infections/transmission , HIV-1/physiology , Humans , In Vitro Techniques , Medroxyprogesterone Acetate/administration & dosage , RNA, Messenger/genetics , Receptors, CCR5/genetics , Risk Factors , Virus Replication/drug effects
14.
PLoS One ; 13(4): e0196043, 2018.
Article in English | MEDLINE | ID: mdl-29698514

ABSTRACT

High usage of progestin-only injectable contraceptives, which include the intramuscular injectables depo-medroxyprogesterone acetate (DMPA-IM, Depo-Provera) and norethisterone (NET) enanthate (NET-EN or Nur-Isterate), correlates worldwide with areas of high HIV-1 prevalence. Epidemiological data show a significant association between usage of DMPA-IM and increased HIV-1 acquisition but no such association from limited data for NET-EN. Whether MPA and NET have similar effects on HIV-1 acquisition and pathogenesis, and the relationship between these effects and the dose of MPA, are critical issues for women's health and access to suitable and safe contraceptives. We show for the first time that MPA, unlike NET, significantly increases HIV-1 replication in peripheral blood mononuclear cells (PBMCs) and a cervical cell line model. The results provide novel evidence for a biological mechanism whereby MPA, acting via the glucocorticoid receptor (GR), increases HIV-1 replication by at least in part increasing expression of the CCR5 HIV-1 coreceptor on target T-lymphocytes. MPA, unlike NET, also increases activation of T-cells and increases the CD4/CD8 ratio, suggesting that multiple mechanisms are involved in the MPA response. Our data offer strong support for different biological mechanisms for MPA versus NET, due to their differential GR activity. The dose-dependence of the MPA response suggests that significant effects are observed within the range of peak serum levels of progestins in DMPA-IM but not NET-EN users. Dose-response results further suggest that effects of contraceptives containing MPA on HIV-1 acquisition and disease progression may be critically dependent on dose, time after injection and intrinsic factors that affect serum concentrations in women.


Subject(s)
HIV-1/physiology , Medroxyprogesterone Acetate/pharmacology , Receptors, CCR5/metabolism , Receptors, Glucocorticoid/metabolism , Virus Replication/drug effects , CD4 Antigens/genetics , CD4 Antigens/metabolism , CD4-CD8 Ratio , Cell Line , Female , HEK293 Cells , HIV-1/drug effects , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Mifepristone/pharmacology , Norethindrone/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Receptors, CCR5/genetics , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/genetics , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Up-Regulation/drug effects
15.
Cancer Res ; 76(12): 3604-17, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27197171

ABSTRACT

The RNA-binding protein dyskerin, encoded by the DKC1 gene, functions as a core component of the telomerase holoenzyme as well as ribonuclear protein complexes involved in RNA processing and ribosome biogenesis. The diverse roles of dyskerin across many facets of RNA biology implicate its potential contribution to malignancy. In this study, we examined the expression and function of dyskerin in neuroblastoma. We show that DKC1 mRNA levels were elevated relative to normal cells across a panel of 15 neuroblastoma cell lines, where both N-Myc and c-Myc directly targeted the DKC1 promoter. Upregulation of MYCN was shown to dramatically increase DKC1 expression. In two independent neuroblastoma patient cohorts, high DKC1 expression correlated strongly with poor event-free and overall survival (P < 0.0001), independently of established prognostic factors. RNAi-mediated depletion of dyskerin inhibited neuroblastoma cell proliferation, including cells immortalized via the telomerase-independent ALT mechanism. Furthermore, dyskerin attenuation impaired anchorage-independent proliferation and tumor growth. Overexpression of the telomerase RNA component, hTR, demonstrated that this proliferative impairment was not a consequence of telomerase suppression. Instead, ribosomal stress, evidenced by depletion of small nucleolar RNAs and nuclear dispersal of ribosomal proteins, was the likely cause of the proliferative impairment in dyskerin-depleted cells. Accordingly, dyskerin suppression caused p53-dependent G1 cell-cycle arrest in p53 wild-type cells, and a p53-independent pathway impaired proliferation in cells with p53 dysfunction. Together, our findings highlight dyskerin as a new therapeutic target in neuroblastoma with crucial telomerase-independent functions and broader implications for the spectrum of malignancies driven by MYC family oncogenes. Cancer Res; 76(12); 3604-17. ©2016 AACR.


Subject(s)
Cell Cycle Proteins/physiology , Neuroblastoma/pathology , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-myc/physiology , Telomerase/physiology , Cells, Cultured , G1 Phase Cell Cycle Checkpoints , Humans , Ribosomes/physiology , Tumor Suppressor Protein p53/physiology
16.
Cell Mol Biol Lett ; 20(4): 626-46, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26274783

ABSTRACT

Extensive re-organisation of the actin cytoskeleton and changes in the expression of its binding proteins is a characteristic feature of cancer cells. Previously we have shown that the tropomyosin isoform Tpm3.1, an integral component of the actin cytoskeleton in tumor cells, is required for tumor cell survival. Our objective was to determine whether cancer cells devoid of Tpm3.1 would evade the tumorgenic effects induced by H-Ras transformation. The tropomyosin isoform (Tpm) expression profile of a range of cancer cell lines (21) demonstrates that Tpm3.1 is one of the most broadly expressed Tpm isoform. Consequently, the contribution of Tpm3.1 to the transformation process was functionally evaluated. Primary embryonic fibroblasts isolated from wild type (WT) and Tpm3.1 knockout (KO) mice were transduced with retroviral vectors expressing SV40 large T antigen and an oncogenic allele of the H-Ras gene, H-RasV12, to generate immortalized and transformed WT and KO MEFs respectively. We show that Tpm3.1 is required for growth factor-independent proliferation in the SV40 large T antigen immortalized MEFs, but this requirement is overcome by H-Ras transformation. Consistent with those findings, we found that Tpm3.1 was not required for anchorage independent growth or growth of H-Ras-driven tumors in a mouse model. Finally, we show that pERK and Importin 7 protein interactions are significantly decreased in the SV40 large T antigen immortalized KO MEFs but not in the H-Ras transformed KO cells, relative to control MEFs. The data demonstrate that H-Ras transformation overrides a requirement for Tpm3.1 in growth factor-independent proliferation of immortalized MEFs. We propose that in the SV40 large T antigen immortalized MEFs, Tpm3.1 is partly responsible for the efficient interaction between pERK and Imp7 resulting in cell proliferation, but this is overidden by Ras transformation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Genes, ras/genetics , Tropomyosin/genetics , Animals , Antigens, Polyomavirus Transforming/genetics , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fibroblasts/pathology , Humans , Karyopherins/genetics , Karyopherins/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tropomyosin/metabolism
17.
Methods Mol Biol ; 965: 215-31, 2013.
Article in English | MEDLINE | ID: mdl-23296661

ABSTRACT

The enzyme telomerase is activated in 80-90% of all human malignancies and immortal cell lines, where it functions to maintain the integrity of chromosomal-end structures called telomeres. Telomerase enzyme activity can be detected in whole cell lysates by a polymerase chain reaction (PCR)-based method referred to as the telomeric repeat amplification protocol (TRAP). The TRAP assay involves extension of an oligonucleotide through telomerase-mediated enzymatic addition of telomeric DNA repeats and subsequent PCR amplification of the extension products. While the TRAP assay as originally developed utilizes radioactively labelled nucleotides, protocols are provided herein for nonradioactive versions of the TRAP assay, with options for either qualitative assessment of TRAP products by polyacrylamide gel electrophoresis (standard TRAP), or quantitative analysis by real-time PCR (Q-TRAP). The Q-TRAP method poses the additional advantages of exquisite sensitivity, rapidity, and potential for adoption to a high-throughput format.


Subject(s)
Enzyme Assays/methods , Telomerase/metabolism , Cell Line , DNA Primers/genetics , Humans , Polymerase Chain Reaction , Repetitive Sequences, Nucleic Acid/genetics , Telomere/genetics
18.
Biol Chem ; 392(5): 439-48, 2011 May.
Article in English | MEDLINE | ID: mdl-21417953

ABSTRACT

AP-1, a transcription factor comprised primarily of Jun and Fos family proteins, regulates genes involved in proliferation, differentiation and oncogenesis. Previous studies demonstrated that elevated expression of Jun and Fos family member proteins is associated with numerous human cancers and in cancer-relevant biological processes. In this study we used a dominant-negative mutant of c-Jun, Tam67, which interferes with the functional activity of all AP-1 complexes, to investigate the requirement of AP-1 in the proliferation and cell cycle progression of cervical cancer cells. Transient and stable expression of Tam67 in CaSki cervical cancer cells resulted in decreased AP-1 activity that correlated with a significant inhibition of cell proliferation and anchorage-independent colony formation. Inhibiting AP-1 activity resulted in a two-fold increase in cells located in the G(2)/M phase of the cell cycle and an accompanying increase in the expression of the cell cycle regulatory protein, p21. The increase in p21 was associated with a decrease in HPV E6 expression and an increase in p53. Importantly, blocking the induction of p21 in CaSki-Tam67-expressing cells accelerated their proliferation rate to that of CaSki, implicating p21 as a key player in the growth arrest induced by Tam67. Our results suggest a role for AP-1 in the proliferation, G(2)/M progression and inhibition of p21 expression in cervical cancer.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Transcription Factor AP-1/metabolism , Uterine Cervical Neoplasms/metabolism , Blotting, Western , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , HeLa Cells , Humans , Peptide Fragments/genetics , Peptide Fragments/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor AP-1/genetics , Uterine Cervical Neoplasms/genetics
19.
Future Oncol ; 6(5): 769-89, 2010 May.
Article in English | MEDLINE | ID: mdl-20465390

ABSTRACT

Over the past two decades, it has become increasingly apparent that telomerase-mediated telomere maintenance plays a crucial role in hematopoiesis. Supporting evidence is underscored by recent findings of mutations in genes involved in telomerase-mediated telomere maintenance that contribute to the pathogenesis of bone marrow failure syndromes. More recently described telomere-independent functions of telomerase are also likely to contribute to both normal hematopoiesis and hematologic diseases. The high levels of telomerase detected in aggressive leukemias have fueled fervent investigation into diverse approaches to targeting telomerase in hematologic malignancies. Successful preclinical investigations that employed genetic strategies, oligonucleotides, small-molecule inhibitors and immunotherapy have resulted in a rapid translation to clinical trials. Further investigation of telomere-independent functions of telomerase and detailed preclinical studies of telomerase inhibition in both normal and malignant hematopoiesis will be invaluable for refining treatments to effectively and safely exploit telomerase as a therapeutic target in hematologic malignancies.


Subject(s)
Hematologic Neoplasms/genetics , Telomerase/physiology , Animals , Hematologic Neoplasms/metabolism , Humans , Telomere/metabolism
20.
Mol Cancer Ther ; 6(9): 2535-43, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17876050

ABSTRACT

Marine invertebrates, algae, and microorganisms are prolific producers of novel secondary metabolites. Some of these secondary metabolites have the potential to be developed as chemotherapeutic agents for the treatment of a wide variety of diseases, including cancer. We describe here the mechanism leading to apoptosis of esophageal cancer cell lines in the presence of triprenylated toluquinones and toluhydroquinones originally isolated from the Arminacean nudibranch Leminda millecra. Triprenylated toluquinone-induced and toluhydroquinone-induced cell death is mediated via apoptosis after a cell cycle block. Molecular events include production of reactive oxygen species (ROS), followed by induction and activation of c-Jun (AP1) via c-Jun-NH2-kinase-mediated and extracellular signal-regulated kinase-mediated pathways. Partial resistance to these compounds could be conferred by the ROS scavengers Trolox and butylated hydroxyanisol, a c-Jun-NH2-kinase inhibitor, and inhibition of c-Jun with a dominant negative mutant (TAM67). Interestingly, the levels of ROS produced varied between compounds, but was proportional to the ability of each compound to kill cells. Because cancer cells are often more susceptible to ROS, these compounds present a plausible lead for new antiesophageal cancer treatments and show the potential of the South African marine environment to provide new chemical entities with potential clinical significance.


Subject(s)
Apoptosis/drug effects , Esophageal Neoplasms/drug therapy , Hydroquinones/pharmacology , Mollusca/chemistry , Quinones/pharmacology , Reactive Oxygen Species/metabolism , Animals , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Free Radical Scavengers/pharmacology , Humans , Hydroquinones/chemistry , Mitogen-Activated Protein Kinases/metabolism , Necrosis , Proto-Oncogene Proteins c-jun/metabolism , Quinones/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...