Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Med Chem ; 269: 116324, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38520762

ABSTRACT

The development of cancer is influenced by several variables, including altered protein expression, and signaling pathways. Cancers are inherently heterogeneous and exhibit genetic and epigenetic aberrations; therefore, developing therapies that act on numerous biological targets is encouraged. To achieve this, two approaches are employed: combination therapy and dual/multiple targeting chemotherapeutics. Two enzymes, histone deacetylases (HDACs) and ribonucleotide reductase (RR), are crucial for several biological functions, including replication and repair of DNA, division of cells, transcription of genes, etc. However, it has been noted that different cancers exhibit abnormal functions of these enzymes. Potent inhibitors for each of these proteins have been extensively researched. Many medications based on these inhibitors have been successfully food and drug administration (FDA) approved, and the majority are undergoing various stages of clinical testing. This review discusses various studies of HDAC and RR inhibitors in combination therapy and dual-targeting chemotherapeutics.


Subject(s)
Neoplasms , Ribonucleotide Reductases , Humans , Ribonucleotide Reductases/therapeutic use , Histone Deacetylases/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Neoplasms/genetics
2.
Med Oncol ; 40(12): 353, 2023 Nov 11.
Article in English | MEDLINE | ID: mdl-37952032

ABSTRACT

3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP) has broad-spectrum antitumor activity. However, its role in osteosarcoma (OS) remains unclear. Therefore, this study explored the effects of 3-AP on OS in vitro and in vivo using three human OS cell lines (MG-63, U2-OS, and 143B) and a nude mice model generated by transplanting 143B cells. The cells and mice were treated with DMSO (control) or gradient concentrations of 3-AP. Then, various assays (e.g., cell counting kit-8, flow cytometry, immunohistochemistry, and western blotting) were performed to assess cell viability and apoptosis levels, as well as γH2A.X (DNA damage correlation), ribonucleotide reductase catalytic subunit M1 and M2 (RRM1 and RRM2, respectively) protein levels (iron-dependent correlation). 3-AP time- and dose-dependably suppressed growth and induced apoptosis in all three OS cell lines, and ferric ammonium citrate (FAC) blocked these effects. Moreover, 3-AP decreased RRM2 and total ribonucleotide reductase (RRM1 plus RRM2) protein expression but significantly increased γH2A.X expression; treatment did not affect RRM1 expression. Again, FAC treatment attenuated these effects. In vivo, the number of apoptotic cells in the tumor slices increased in the 3-AP-treated mice compared to the control mice. 3-AP treatment also decreased Ki-67 and p21 expression, suggesting inhibited OS growth. Furthermore, the expression of RRM1, RRM2, and transferrin receptor protein 1 (i.e., Tfr1) indicated that 3-AP inhibited OS growth via an iron-dependent pathway. In conclusion, 3-AP exhibits anticancer activity in OS by decreasing the activity of iron-dependent pathways, which could be a promising therapeutic strategy for OS.


Subject(s)
Bone Neoplasms , Osteosarcoma , Ribonucleotide Reductases , Humans , Animals , Mice , Iron/therapeutic use , Mice, Nude , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Ribonucleotide Reductases/therapeutic use , Cell Proliferation , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Cell Line, Tumor , Apoptosis
3.
Anticancer Agents Med Chem ; 23(17): 1958-1965, 2023.
Article in English | MEDLINE | ID: mdl-37565554

ABSTRACT

BACKGROUND: The development of chemotherapy resistance in prostate cancer (PCa) patients poses a significant obstacle to disease progression. Ribonucleotide reductase is a crucial enzyme for cell division and tumor growth. Triapine, an inhibitor of ribonucleotide reductase, has shown strong anti-tumor activity in various types of cancers. However, the effect of triapine on docetaxel-resistant (DR) human PCa cells has not been explored previously. AIM: This study aimed to examine the potential anti-proliferative effects of triapine in PC3-DR (docetaxel-resistant) cells. METHODS: Cell viability was determined by the MTT test, and apoptosis and cell cycle progression were analyzed by image-based cytometer. mRNA and protein expression were assessed by RT-qPCR and western blot, respectively. RESULTS: Triapine administration significantly reduced PC3 and PC3-DR cells' survival, while the cytotoxic effect was higher in PC3-DR cells. Cell death resulting from inhibition of ribonucleotide reductase was mediated by endoplasmic reticulum stress, induction of apoptosis, and cell cycle arrest. The findings were supported by the upregulation of caspases, Bax, Bak, P21, P27, P53, TNF-α, FAS, and FASL, and downregulation of Bcl2, Bcl-XL, cyclin-dependent kinase 2 (CDK2), CDK4, cyclins, and heat shock proteins expression. According to the data, the reduction of ABC transporter proteins and NF-ĸB expression may play a role in triapine-mediated cytotoxicity in docetaxel-resistant cells. CONCLUSION: Based on our findings, triapine emerges as a promising chemotherapeutic approach for combating docetaxel- resistant prostate cancer.


Subject(s)
Prostatic Neoplasms , Ribonucleotide Reductases , Male , Humans , Docetaxel/pharmacology , Docetaxel/therapeutic use , Ribonucleotide Reductases/pharmacology , Ribonucleotide Reductases/therapeutic use , Apoptosis , Prostatic Neoplasms/metabolism , Endoplasmic Reticulum Stress , Cell Line, Tumor
4.
Sci Adv ; 9(20): eade7236, 2023 05 19.
Article in English | MEDLINE | ID: mdl-37196077

ABSTRACT

During therapy, adaptations driven by cellular plasticity are partly responsible for driving the inevitable recurrence of glioblastoma (GBM). To investigate plasticity-induced adaptation during standard-of-care chemotherapy temozolomide (TMZ), we performed in vivo single-cell RNA sequencing in patient-derived xenograft (PDX) tumors of GBM before, during, and after therapy. Comparing single-cell transcriptomic patterns identified distinct cellular populations present during TMZ therapy. Of interest was the increased expression of ribonucleotide reductase regulatory subunit M2 (RRM2), which we found to regulate dGTP and dCTP production vital for DNA damage response during TMZ therapy. Furthermore, multidimensional modeling of spatially resolved transcriptomic and metabolomic analysis in patients' tissues revealed strong correlations between RRM2 and dGTP. This supports our data that RRM2 regulates the demand for specific dNTPs during therapy. In addition, treatment with the RRM2 inhibitor 3-AP (Triapine) enhances the efficacy of TMZ therapy in PDX models. We present a previously unidentified understanding of chemoresistance through critical RRM2-mediated nucleotide production.


Subject(s)
Brain Neoplasms , Drug Resistance, Neoplasm , Glioblastoma , Ribonucleotide Reductases , Humans , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Ribonucleotide Reductases/genetics , Ribonucleotide Reductases/therapeutic use , Temozolomide/pharmacology , Temozolomide/therapeutic use , Drug Resistance, Neoplasm/genetics
5.
Parasitol Int ; 91: 102632, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35870741

ABSTRACT

The kinetoplastid protozoan parasite, Trypanosoma evansi causes a fatal disease condition known as Surra in equines throughout the globe. Disease condition being acute in nature, entrust a huge economic and health impact on the equine industry. Till date, quinapyramine methyl sulphate (QPS) is the first line of treatment and a panacea for the T. evansi infection in equines. Still after the >70 years of its discovery, there is no clue about the mode of action of QPS in T. evansi. The establishment of in vitro cultivation of T. evansi in HMI-9 media has provided opportunity to study the alteration in mRNA expression of parasite on exposure to the drug. With this research gap, the present study aimed to investigate the relative mRNA expression of 13 important drug target genes to elucidate the anti-trypanosomal activity of QPS against T. evansi. The IC50 of QPS against a pony isolate of T. evansi was determined as 276.4 nM(147.21 ng/ mL) in the growth inhibitory assay. The in vitro cultured T. evansi population were further exposed to IC50 of QPS and their relative mRNA expression was studied at 12 h, 24 h and 48 h interval.The mRNA expression of several genes such as hexokinase, trypanothione reductase, aurora kinase, oligopeptidase B and ribonucleotide reductase II were found refractory (non-significant, p > 0.1234) to the exposure of QPS. Significant up-regulation of trans-sialidase (p < 0.0001), ESAG8 (p < 0.0021), ribonucleotide reductase I (p < 0.0001), ornithine decarboxylase (p < 0.0001), topoisomerase II (p < 0.0021) and casein kinase I (p < 0.0021) were recorded after exposure with QPS. The arginine kinase 1 and calcium ATPase I showed highly significant (p < 0.0001) down-regulation in the drug kinetics. Therefore, the arginine kinase 1 and calcium ATPase I can be explored further to elucidate the trypanocidal activity of QPS. The preliminary data generated provide the potential of arginine kinase 1 and calcium ATPase I mRNA mediated pathway of trypanocidal action of QPS. Further, transcriptomics approach is required to investigate the possible mechanism of action of drugs at molecular level against the targeted organism.


Subject(s)
Arginine Kinase , Ribonucleotide Reductases , Trypanocidal Agents , Trypanosoma , Trypanosomiasis , Animals , Arginine Kinase/metabolism , Arginine Kinase/therapeutic use , Gene Expression , Horses , Quinolinium Compounds , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleotide Reductases/metabolism , Ribonucleotide Reductases/therapeutic use , Sulfuric Acid Esters , Trypanocidal Agents/metabolism , Trypanosomiasis/drug therapy , Trypanosomiasis/veterinary
6.
Clin Diagn Lab Immunol ; 11(3): 437-45, 2004 May.
Article in English | MEDLINE | ID: mdl-15138167

ABSTRACT

The development of effective prophylactic and therapeutic vaccines against genital herpes has proven problematic. Difficulties are associated with the complexity of the virus life cycle (latency) and our relatively poor understanding of the mechanism of immune control of primary and recurrent disease. The types of effector cells and the mechanisms responsible for their activation and regulation are particularly important. Studies from my and other laboratories have shown that recurrent disease is prevented by virus-specific T helper 1 (Th1) cytokines (viz., gamma interferon) and activated innate immunity. Th2 cytokines (viz., interleukin-10 [IL-10]) and regulatory (suppressor) T cells downregulate this immune profile, thereby allowing unimpeded replication of reactivated virus and recurrent disease. Accordingly, an effective therapeutic vaccine must induce Th1 immunity and be defective in Th2 cytokine production, at least IL-10. These concepts are consistent with the findings of the most recent clinical trials, which indicate that (i) a herpes simplex virus type 2 (HSV-2) glycoprotein D (gD-2) vaccine formulated with a Th1-inducing adjuvant has prophylactic activity in HSV-2- and HSV-1-seronegative females, an activity attributed to the adjuvant function, and (ii) a growth-defective HSV-2 mutant (ICP10DeltaPK), which is deleted in the Th2-polarizing gene ICP10PK, induces Th1 immunity and has therapeutic activity in both genders. The ICP10DeltaPK vaccine prevents recurrent disease in 44% of treated subjects and reduces the frequency and severity of recurrences in the subjects that are not fully protected. Additional studies to evaluate these vaccines are warranted.


Subject(s)
Herpes Simplex Virus Vaccines/immunology , Herpesvirus 2, Human/immunology , Animals , Antigens, Viral/immunology , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Female , Herpes Genitalis/immunology , Herpes Genitalis/prevention & control , Herpes Genitalis/therapy , Herpes Simplex Virus Vaccines/adverse effects , Herpes Simplex Virus Vaccines/therapeutic use , Humans , Immunotherapy, Active , Male , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/therapeutic use , Ribonucleotide Reductases/genetics , Ribonucleotide Reductases/immunology , Ribonucleotide Reductases/therapeutic use , Th1 Cells/immunology , Vaccines, Attenuated/immunology , Vaccines, DNA/immunology , Vaccines, Inactivated/immunology , Vaccines, Subunit/immunology , Viral Envelope Proteins/immunology
7.
Radiat Oncol Investig ; 5(2): 62-71, 1997.
Article in English | MEDLINE | ID: mdl-9303059

ABSTRACT

Gemcitabine, a cytidine nucleoside analogue, has schedule-dependent antitumor activity in vitro and in vivo. Gemcitabine also has dose- and time-dependent radiosensitization properties in vitro. Thus it may have therapeutic application in combination with radiation. The aims of this study were to investigate whether gemcitabine could enhance radiation-induced tumor regrowth delay in a human squamous carcinoma (FaDu) xenograft in nude mice and to examine the effect of gemcitabine on radiation-induced apoptosis in in vivo tumors. Radiation was given locally to the tumors twice daily in 2 Gy fractions over 2 weeks for 5 days/week. Significant regrowth delay enhancement was observed which was dependent on gemcitabine schedule. Effective schedules using maximum tolerated gemcitabine doses were twice weekly and once weekly, but not daily. Significant toxicity occurred with radiation plus twice weekly gemcitabine, but enhancement was seen using gemcitabine doses well below the maximum tolerated dose. Both gemcitabine and radiation led to apoptotic cell death, but this was not increased when both treatments were combined. These results indicate that gemcitabine may be of therapeutic value as a radiation enhancer in the treatment of human cancers. Preliminary studies suggest that increased apoptotic cell death is not a mechanism leading to this enhancement.


Subject(s)
Carcinoma, Squamous Cell/radiotherapy , Deoxycytidine/analogs & derivatives , Radiation-Sensitizing Agents/therapeutic use , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Division/drug effects , Cell Division/radiation effects , Combined Modality Therapy , Deoxycytidine/administration & dosage , Deoxycytidine/therapeutic use , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Administration Schedule , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Radiation-Sensitizing Agents/administration & dosage , Ribonucleotide Reductases/administration & dosage , Ribonucleotide Reductases/antagonists & inhibitors , Ribonucleotide Reductases/therapeutic use , Transplantation, Heterologous , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...