Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Prod Res ; 37(7): 1161-1167, 2023 Apr.
Article in English | MEDLINE | ID: mdl-34720015

ABSTRACT

Highly efficient methodology was developed for the construction of functionalized Kojic acid involving Click linker via 1,3-dipolar cycloaddition and their cytotoxicity against MCF-7, MIAPaCa-2 and DU145 mammalian cell lines were evaluated. Preliminary studies on structure-activity-relationship (SAR) revealed that substitution at C-2 of kojic acid as well as C-5 of 1,2,3-triazole motif played a major role in the activity profile. Kojic acid 1,2,3-triazole analogue 3 b containing an alkyl chain (n = 6) exhibited two fold potent activity than the parent compound, kojic acid against MCF-7 and MIA PaCa-2 cell lines. It induced apoptosis in these cell lines via ID1/PARP1 mediated pathway. The structures of the new analogues of kojic acid 1,2,3-triazole were confirmed by the detailed spectroscopic data analysis.


Subject(s)
Antineoplastic Agents , Cytotoxins , Animals , Molecular Structure , Cytotoxins/pharmacology , Structure-Activity Relationship , Triazoles/chemistry , Antineoplastic Agents/chemistry , Cell Proliferation , Drug Screening Assays, Antitumor , Mammals
2.
Gene ; 853: 147092, 2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36464175

ABSTRACT

A family of inhibitors of cell differentiation or DNA-binding proteins, known as ID proteins (ID1-4), function as mighty transcription factors in various cellular processes, such as inhibiting differentiation, promoting cell-cycle progression, senescence, angiogenesis, tumorigenesis, and metastasis in cancer. Pancreatic cancer represents the deadliest cancer with the lowest survival rate of 10% due to the diagnosis at an advanced fatal stage and therapeutic resistance. Modestly, the only curative option for this lethal cancer is surgery but is done in less than 15-20% of patients because of the locally aggressive and early metastatic nature. Finding the earliest biomarkers and targeting the various hallmarks of pancreatic cancer can improve the treatment and survival of pancreatic cancer patients. Therefore, herein in this review, we explore in depth the potential roles of ID proteins function in hallmarks of pancreatic cancer, signaling pathways, and its oncogenic and tumor-suppressive effects. Hence, understanding the roles of dysregulated ID proteins would provide new insights into its function in pancreatic cancer tumorigenesis.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , DNA-Binding Proteins , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Pancreatic Neoplasms/genetics , Cell Differentiation , Carcinogenesis , Cell Transformation, Neoplastic , DNA , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms
3.
Biotechniques ; 73(1): 58-69, 2022 06.
Article in English | MEDLINE | ID: mdl-35775367

ABSTRACT

Western blotting (WB), also known as immunoblotting, is a well-known molecular biology method that biologists often use to investigate many features of the protein, ranging from basic protein analysis to disease detection. WB is simple, unique, rapid, widely used routine tool with easy interpretation and definite results. It is being used in various fields of science, research and development, diagnostic labs and hospitals. The principle of WB is to accomplish the separation of proteins based on molecular weight and charge. This review addresses in detail the individual steps involved in the WB technique, its troubleshooting, internal loading controls, total protein staining and its diverse applications in scientific research and clinical settings, along with its future perspectives.


Subject(s)
Biomedical Research , Proteins , Blotting, Western , Staining and Labeling
4.
Drug Dev Res ; 83(4): 910-926, 2022 06.
Article in English | MEDLINE | ID: mdl-35092073

ABSTRACT

Pancreatic cancer (PC) is among the most lethal cancers and is resistant to existing therapies, which highlights the need for new and alternative therapeutic treatments. Autophagy is emerging as one of the alternative cell death mechanisms and is well known to cross-talk with apoptosis. Autophagy can act as a viable option to treat highly resistant PC. The current study investigates and provides insight into the autophagic and apoptotic cell death induced by quinoline derivative 2-(6-methoxynaphthalen-2-yl)quinolin-4-amine (6MN-4-AQ) in PC cell lines PANC-1 and MIA PaCa-2. Treatment with 6MN-4-AQ reduced cell viability in concentration dependent manner (2-16 µM) and inhibited the clonogenic potential of PC cells at a concentration of 4 µM for 24 h. Further, we found that 6MN-4-AQ induced both apoptosis and autophagic cell death simultaneously. We identified that 6MN-4-AQ induced autophagic cell death by forming cytoplasmic vacuoles, the elevation of autophagy flux, increase in LC3-II, Beclin-1 protein expression, and degradation of p62. Moreover, 6MN-4-AQ induced apoptosis via Caspase-3 activation and cleavage of PARP in PC cells. Upon investigating the underlying mechanism associated with 6MN-4-AQ induced cell death, it was observed that 6MN-4-AQ treatment is able to suppress the Akt/mTOR pathway and induced ER stress leading to the induction of autophagy. Also, 6MN-4-AQ treatment suppressed epithelial to mesenchymal transition by reducing the protein expression of SLUG, snail, and vimentin. Subsequently, 6MN-4-AQ inhibited cell migration and invasion by down regulating MMP-7 and MMP-9 protein expression, suggesting that 6MN-4-AQ may serve as a plausible therapeutic agent for PC.


Subject(s)
Pancreatic Neoplasms , Quinolines , Amines , Apoptosis , Autophagy , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Humans , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Quinolines/pharmacology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Pancreatic Neoplasms
5.
Drug Dev Res ; 83(3): 605-614, 2022 05.
Article in English | MEDLINE | ID: mdl-34612529

ABSTRACT

Noscapine is a phthalide isoquinoline alkaloid present in the latex of Papaver somniferum and has demonstrated potent antitumor activity in various cancer models. Structural changes in the core molecule of noscapine architecture have produced a number of potent analogs. We have recently synthesized the novel noscapine analogs (3, 4, and 5) with different functional groups appended at ninth position of natural noscapine. The anticancer activity of these compounds has been investigated using various human cancer cell lines such as HeLa (cervical cancer), DU-145 (prostate cancer), MCF-7 (breast cancer), and IMR-32 (neuroblastoma). One of the compounds in this series, 9-ethynyl noscapine (5), has demonstrated good anticancer activity against HeLa cells. Biological studies demonstrated that compound 5 decreased cell viability and colony formation in HeLa cells in a concentration dependent manner. To further uncover the mechanism in detail, we evaluated compound 5 effect on cell cycle progression, microtubule dynamics, and apoptosis. Cell cycle and western blotting analysis revealed that 9-ethynyl noscapine treatment resulted in cell cycle arrest at G2/M and decreased CDK1 and cyclinB1 protein expression. We also observed that 9-ethynyl noscapine (5) treatment leads to disruption in tubulin polymerization and induction of apoptosis by decreasing expression of bcl2, pro-caspase 3, and activation of cytochrome C. Taken together, our results indicate that 9-ethynyl noscapine (5) effectively supresses the growth of cervical cancer cells (HeLa) by disrupting tubulin polymerization, cell cycle progression leading to apoptosis.


Subject(s)
Antineoplastic Agents , Noscapine , Uterine Cervical Neoplasms , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Female , G2 Phase Cell Cycle Checkpoints , HeLa Cells , Humans , Male , Noscapine/pharmacology , Polymerization , Tubulin/chemistry , Tubulin/metabolism , Tubulin/pharmacology , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
6.
Mol Biol Rep ; 48(5): 4973-4979, 2021 May.
Article in English | MEDLINE | ID: mdl-34125328

ABSTRACT

Neuroblastoma (NB) is the most common childhood cancer that arises from the sympathetic nervous system. NB is characterized by poor prognosis. One of the strategies to control NB is activating the differentiation process in undifferentiated NB cells. Many differentiating agents including 13-cis-retinoic acid (RA) led to disappointing results. In the current study, we investigated the effect of Quisinostat/JNJ-26481585(JNJ) on NB SK-N-SH cells differentiation. The SK-N-SH cell differentiation was observed by morphology and neurite length measurement. The cell cycle arrest was determined by FACS analysis. The relative levels of autophagy marker LC3-II, neuronal markers ßIII-tubulin and Eno-2, cell cycle related proteins cyclin D1 and CDK 4 were detected by western blotting. JNJ induces differentiation in SK-N-SH cells, as evident by the morphological features and expression of neuronal markers, ßIII-tubulin and Eno-2. Cell cycle arrest at G1 phase was confirmed by a decrease in the expression of cyclin D1 and CDK 4. Furthermore, we also observed that autophagy plays an important role in JNJ induced cell differentiation of SK-N-SH cells. We demonstrated that autophagy is induced upon JNJ treatment and is important for the neuronal differentiation of human SK-N-SH cells.


Subject(s)
Autophagy/drug effects , Cell Differentiation/drug effects , Hydroxamic Acids/pharmacology , Neuroblastoma , Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Child , Humans , Neural Stem Cells/drug effects , Neuroblastoma/drug therapy , Neuroblastoma/metabolism
7.
Life Sci ; 277: 119504, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33872660

ABSTRACT

The role of genetic and epigenetic factors in tumor initiation and progression is well documented. Histone deacetylases (HDACs), histone methyl transferases (HMTs), and DNA methyl transferases. (DNMTs) are the main proteins that are involved in regulating the chromatin conformation. Among these, histone deacetylases (HDAC) deacetylate the histone and induce gene repression thereby leading to cancer. In contrast, histone acetyl transferases (HATs) that include GCN5, p300/CBP, PCAF, Tip 60 acetylate the histones. HDAC inhibitors are potent drug molecules that can induce acetylation of histones at lysine residues and induce open chromatin conformation at tumor suppressor gene loci and thus resulting in tumor suppression. The key processes regulated by HDAC inhibitors include cell-cycle arrest, chemo-sensitization, apoptosis induction, upregulation of tumor suppressors. Even though FDA approved drugs are confined mainly to haematological malignancies, the research on HDAC inhibitors in glioblastoma multiforme and triple negative breast cancer (TNBC) are providing positive results. Thus, several combinations of HDAC inhibitors along with DNA methyl transferase inhibitors and histone methyl transferase inhibitors are in clinical trials. This review focuses on how HDAC inhibitors regulate the expression of coding and non-coding genes with specific emphasis on their anti-cancer potential.


Subject(s)
Histone Deacetylase Inhibitors/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Acetylation , Apoptosis/drug effects , Cell Cycle/drug effects , Chromatin/metabolism , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Epigenomics/methods , Gene Expression/drug effects , Histone Acetyltransferases/metabolism , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Neoplasms/metabolism , Protein Processing, Post-Translational/drug effects
8.
Chem Biol Interact ; 343: 109433, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33689707

ABSTRACT

Pancreatic cancer is one of the most malignant cancers around the world. The co-occurrence of mutation in KRAS and p53 makes it highly aggressive, proliferative, metastatic, and resistant to apoptotic cell death. Therefore, there is a need to trigger an alternate mechanism of cancer cell death in apoptosis-resistant pancreatic cancer. Autophagic cell death could be an alternate viable option for treatment in such cases. Thus, the identification of small molecules as autophagy modulators with potent anticancer efficacy would be of great importance in pancreatic cancer. The present study investigates fluorinated thiazolidionol (FTZ) driven autophagy modulation, underlying mechanism, and regulation of critical sentinels of oncogenic signaling in pancreatic cancer cells. We identified that FTZ triggered autophagic cell death in pancreatic cancer cells, independent of apoptosis evidenced by an increase in cytoplasmic vacuoles formation, autophagy flux, LC3-II expression, and p62 degradation. Further, the crucial events of apoptosis i.e., Caspase-3 activation and PARP cleavage, were not observed, indicating the non-occurrence of apoptotic cell death. Moreover, FTZ was able to activate AMPK and suppress PI3k/Akt/mTOR as well as MEK/ERK, the key oncogenic signaling pathways in cancer cells. Furthermore, treatment with FTZ suppressed migration, invasion, and angiogenesis in pancreatic cancer cells. Studies in vivo revealed significant regression of tumors by FTZ in nude mice model. Overall, our study demonstrates that FTZ induces autophagic cell death in pancreatic cancer cells independent of apoptosis, which is accompanied by AMPK activation and suppression of critical sentinels of oncogenic signaling in pancreatic cancer cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/therapeutic use , Autophagic Cell Death/drug effects , MAP Kinase Signaling System/drug effects , Neoplasms/drug therapy , Thiazoles/therapeutic use , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice, Nude , Thiazoles/pharmacology , Xenograft Model Antitumor Assays
9.
Biomed Pharmacother ; 127: 110128, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32305697

ABSTRACT

BACKGROUND: Amiodarone represents a principal antiarrhythmic pharmaceutical drug available in the market for the treatment of ventricular arrhythmias. However, despite its better efficacy, the usage of amiodarone is associated with extracardiac toxicity. The human body evolved a system of cytochrome P450 enzymes which play an essential role in the metabolism of harmful foreign substances. Therefore, CYP enzymes may either lead to the elimination or degradation of the leftover drug residues. OBJECTIVE: The present study focused on successful utilization of Saccharomyces cerevisiae strain OBS2 with probiotic- cum- therapeutic potential and expressing in silico enhanced human cytochrome P4503A4 for the degradation of leftover drug residues of amiodarone in vitro and in vivo. METHODOLOGY: In this study, cytochrome P4503A4 (1W0E) was taken as a template and the predicted 3D model of mutant CYP3A4 was developed using different bioinformatics tools. Selected mutant (Glu165Asp) protein was reverse translated and transcribed into cDNA sequence. The cDNA of CYP3A4 was synthesized, cloned into p427TEF construct and transformed into Saccharomyces cerevisiae OBS2. The degradation of leftover drug residues of amiodarone in vitro and in vivo using recombinant Saccharomyces cerevisiae OBS2 expressing CYP3A4 was evaluated. RESULT: The CYP3A4 activity in recombinant probiotic yeast was observed as 108 IU/mL and in vitro degradation of leftover drug residues of amiodarone was observed as 66.32 %. Whereas, in vivo degradation of leftover drug residues of amiodarone was observed as 72.61 % along with recovery of organ damage in histopathological studies of the animal model. CONCLUSION: Saccharomyces cerevisiae OBS2 expressing CYP3A4 can be used for probiotic and therapeutic applications.


Subject(s)
Amiodarone/metabolism , Cytochrome P-450 CYP3A/metabolism , Drug Residues/metabolism , Probiotics/therapeutic use , Saccharomyces cerevisiae/genetics , Animals , Cloning, Molecular , Cytochrome P-450 CYP3A/genetics , Male , Rats , Rats, Wistar
10.
Mol Cell Biochem ; 468(1-2): 21-34, 2020 May.
Article in English | MEDLINE | ID: mdl-32146648

ABSTRACT

Neuroblastoma (NB) is the common pediatric tumor of the sympathetic nervous system characterized by poor prognosis. Owing to the challenges such as high tumor heterogeneity, multidrug resistance, minimal residual disease, etc., there is an immediate need for exploring new therapeutic strategies and effective treatments for NB. Herein, in the current study, we explored the unexplored response of NB cells to the second-generation histone deacetylase inhibitor (HDACi) JNJ-26481585(JNJ) and the lysosomotropic agent, Chloroquine (CQ) alone and upon JNJ/CQ treatment as a plausible therapeutic. We identify that while JNJ alone induced autophagy in NB cells, JNJ/CQ treatment decreased the viability and proliferation of NB cells in vitro by switching from autophagy to apoptosis. Further we found that autophagy inhibition by CQ pre-treatment led to the generation of ROS and a decrease in the mitochondrial membrane potential (MMP) that subsequently caused caspase-3-mediated apoptotic cell death in NB cells. Corroborating the above observations, we found that the ROS scavenger N-acetylcysteine (NAC) countered caspase-3 activity and the cells were rescued from apoptosis. Finally, these observations establish that JNJ/CQ treatment resulted in cell death in NB cells by triggering the formation of ROS and disruption of MMP, suggesting that modulation of JNJ-induced autophagy by CQ represents a promising new therapeutic approach in NB.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Chloroquine/pharmacology , Hydroxamic Acids/pharmacology , Membrane Potential, Mitochondrial/drug effects , Neuroblastoma/drug therapy , Peripheral Nervous System Neoplasms/drug therapy , Acetylcysteine/pharmacology , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Free Radical Scavengers/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , Peripheral Nervous System Neoplasms/genetics , Peripheral Nervous System Neoplasms/metabolism , Reactive Oxygen Species/metabolism
11.
ACS Omega ; 4(21): 19382-19398, 2019 Nov 19.
Article in English | MEDLINE | ID: mdl-31763563

ABSTRACT

Noscapine, a phthalide isoquinoline alkaloid isolated from the opium poppy Papaver somniferum, is traditionally being used as an anticough drug. With a safe in vitro toxicological profile, noscapine and its analogues have been explored to show microtubule-regulating properties and anticancer activity against various mammalian cancer cell lines. Since then, our group and other research groups worldwide are working on developing new noscapinoids to tap its potential as the leading drug molecule. With our continuing efforts, we herein present synthesis and anticancer evaluation of a series of imidazothiazole-coupled noscapinoids 7a-o and 11a-o. Natural α-noscapine was N-demethylated to nornoscapine 4 and then reacted with 4-(chloromethyl) thiazole-2-amine. The resultant noscapinoid 5 was coupled with various bromomethyl ketones 10a-o to give N-imidazothiazolyl noscapinoids 7a-o in very good yields. Similarly, natural α-noscapine 1 was O-demethylated using sodium azide/sodium iodide, reacted with 4-(chloromethyl)thiazole-2-amine, and coupled with bromomethyl ketones 10a-o to result in O-imidazothiazolyl noscapinoids 11a-o. All the new analogues 7a-o and 11a-o were fully characterized by their NMR and mass spectral analysis. In vitro cytotoxicity assay was performed for compounds 5, 7a-o, 9, and 11a-o against four different cancer cell lines: HeLa (cervical), MIA PaCa-2 (pancreatic), SK-N-SH (neuroblastoma), and DU145 (prostate cancer). Among these conjugates, 5, 7a, 9, 11b, 11c, 11e, and 11o showed potent cytotoxicity with low IC50 values. Further, flow cytometry analysis revealed that MIA PaCa-2 cells treated with these compounds induced cell cycle G2/M-phase arrest. In addition, Western blot analysis revealed that the cells treated with these conjugates accumulate tubulin in the soluble fraction and also elevate cyclin-B1 protein expression levels. Moreover, the conjugates also increased the expression of caspase-3 and PARP levels which is indicative of apoptotic cell death. In silico molecular docking studies showed several noncovalent interactions like van der Waals and hydrogen-bonding with tubulin protein and with good binding energy. The results indicated that these noscapine analogues may serve as novel compounds that can possibly inhibit tubulin protein and can be considered for further optimization as a clinical candidate for treating pancreatic cancer.

12.
Int J Biol Macromol ; 114: 906-921, 2018 Jul 15.
Article in English | MEDLINE | ID: mdl-29605257

ABSTRACT

Low molecular weight antimicrobial polypeptides were extracted and purified from the young fresh leaves of Azadirachta indica (neem). The total protein extracted was precipitated with 15% TCA-Acetone. The total purified proteins yielded from the two extraction methods were 122.33±2.21 and 115.09±1.88mg/g of the total fresh weight. The SDS-PAGE analysis identified the presence of eight low molecular weight polypeptide bands. The antimicrobial activity of the resolved bands was detected by Polyacrylamide gel-Agar overlay diffusion assay (PAG-ADA). Their broad-spectrum bactericidal activity was confirmed using the same technique and found three low molecular weight bands from 11 to 14kDa collectively exhibiting superior bactericidal activities against Staphylococcus aureus, methicillin-resistant Staphylococcus aureus (MRSA), Staphylococcus epidermis, Enterococcus faecalis, Pseudomonas aeruginosa and fungicidal activity against Candida tropicalis. The FTIR spectrum of the protein bands depicted the presence of hydroxyl and carbonyl groups in the protein bands. These polypeptides were characterized by MALDI-TOF/TOF analysis. Further, the purified protein extract was found to be active against HELA, BT-549 and Neuro-2a cell lines with IC50 value of 74.03±2.31, 64.82±1.64, 238.32±2.12 and 109.94±2.96, 59.61±0.75 for 24h and 48h, respectively. The results of present study indicate that these polypeptides exhibit broad spectrum antimicrobial and anticancer activity and can therefore be explored for their therapeutic potential.


Subject(s)
Anti-Infective Agents , Antineoplastic Agents, Phytogenic , Azadirachta/chemistry , Bacteria/growth & development , Neoplasms/drug therapy , Plant Leaves/chemistry , Plant Proteins , Anti-Infective Agents/chemistry , Anti-Infective Agents/isolation & purification , Anti-Infective Agents/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , HeLa Cells , Humans , Neoplasms/metabolism , Neoplasms/pathology , Plant Proteins/chemistry , Plant Proteins/isolation & purification , Plant Proteins/pharmacology
13.
Curr Top Med Chem ; 17(22): 2523-2537, 2017.
Article in English | MEDLINE | ID: mdl-28056738

ABSTRACT

Microtubules form crucial dynamic structural cellular components of the cell and are composed of the alpha beta tubulin heterodimers. Microtubules are involved in a wide variety of functions in the cell such as attribution to cell shape, motility, intracellular trafficking and mitotic spindle formation. Owing to these reasons, tubulin and microtubules have gained significant interest as important targets for cancer therapy. A review of the existing microtubule targeting drugs specifies that these agents can be categorised into two of the major categories: Microtubule stabilizing agents such as paclitaxel, docetaxel, epothilones, and discodermolide which bind to the tubulin polymer and stabilize the microtubules, microtubule destabilizing agents such as vinca alkaloids, colchicine and combretastatins which bind to tubulin dimers and cause destabilization. These agents ultimately alter the equilibrium between tubulin and microtubule resulting in disruption of mitotic spindle, thereby effecting a critical transition in the cell cycle, leading to cell death. Further, clinical studies of these agents are limited by toxicity effects and emergence of drug resistance. The hybrid drugs are a combination of two or more drugs wherein pharmacophores are incorporated into a single molecule to interact with multiple targets and enhance the cytotoxic action with minimal side effects. Such hybrid regimens can improve therapeutic efficacy and reduce drug toxicity. Therefore, studies on new hybrids with such biological properties form important part in chemistry. In this review, we present an overview of various recent hybrids of colchicines, combretastatin, phodophyllotoxin, etc generated by combination among themselves through linkers or with other pharmacophores and their properties like tubulin stabilization and tubulin destabilization. We also attempted to provide chemistry, toxicity, resistance, side effects of these molecular hybrids acting as microtubule targeting drugs.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Microtubules/drug effects , Neoplasms/drug therapy , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Antineoplastic Agents/chemical synthesis , Cell Cycle/drug effects , Humans , Microtubules/metabolism , Neoplasms/pathology , Protein Stability/drug effects , Tubulin/drug effects , Tubulin/metabolism , Tubulin Modulators/chemical synthesis
14.
Curr Drug Targets ; 18(16): 1836-1851, 2017 Nov 30.
Article in English | MEDLINE | ID: mdl-27557820

ABSTRACT

BACKGROUND: Prohibitin (PHB) is overtly conserved evolutionarily and ubiquitously expressed protein with pleiotropic functions in diverse cellular compartments. However, regulation and function of these proteins in different cells, tissues and in various diseases is different as evidenced by expression of these proteins which is found to be reduced in heart diseases, kidney diseases, lung disease, Crohn's disease and ulcerative colitis but this protein is highly expressed in diverse cancers. The mechanism by which this protein acts at the molecular level in different subcellular localizations or in different cells or tissues in different conditions (diseases or normal) has remained poorly understood. There are several studies reported to understand and decipher PHB's role in diseases and/or cancers of ovary, lung, stomach, thyroid, liver, blood, prostrate, gastric, esophagus, glioma, breast, bladder etc. where PHB is shown to act through mechanisms by acting as oncogene, tumor suppressor, antioxidant, antiapoptotic, in angiogenesis, autophagy etc. OBJECTIVE: This review specifically gives attention to the functional role and regulatory mechanism of PHB proteins in cardiovascular health and diseases and its associated implications. Various molecular pathways involved in PHB function and its regulation are analyzed. CONCLUSION: PHB is rapidly emerging as a critical target molecule for cardiovascular signaling. Progress in delineating CVD and mechanisms of PHB in diverse molecular pathways is essential for determining when and how PHB targeted therapy might be feasible. In this regard, new therapies targeting PHB may best be applied in the future together with molecular profiling of CVD for clinical stratification of disease diagnosis and prognosis.


Subject(s)
Cardiovascular Diseases/metabolism , Neoplasms/metabolism , Repressor Proteins/metabolism , Animals , Cardiovascular Diseases/genetics , Gene Expression Regulation , Humans , Neoplasms/genetics , Oxidative Stress , Prohibitins , Repressor Proteins/genetics , Signal Transduction
15.
J Transl Med ; 11: 130, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23706090

ABSTRACT

BACKGROUND: The present study aimed at using a proteomics based approach to: a) analyze and contrast the proteome of the healthy and isoproterenol induced hypertrophied hearts and b) identify potential biomarkers for diagnosis of cardiac hypertrophy. METHODS: Male Sprague Dawley (SD) rats were administered isoproterenol (ISO, 5 mg/kg, sc, once daily) for 14 days to induce cardiac hypertrophy. There was a significant (p<0.05) increase (~ 55%) in the heart weight to tail length ratio after 14 days of treatment and cardiac hypertrophy was evidenced by significant increase of ß-MHC and ANP, two indicative markers of cardiac hypertrophy, in the treated heart compared to that of control. Following confirmation of hypertrophy, 2DE of the tissue samples was done followed by MS/MS analysis of the protein spots to obtain a proteomic view for identification of novel biomarkers. RESULTS: Several important proteins were identified by proteomics analysis. They belong to the major functional categories such as cholesterol and protein metabolism, muscle contraction and development, transport, TCAcycle, ATP-biosynthesis, chaperone, signal transduction, DNA synthesis and ubiquitinisation. Careful examination of these protein spots by image analysis led to the successful identification of 7 differentially expressed proteins in the diseased sample. Further extension of this work for validation of differential expression of these proteins was also achieved by RTPCR and western blotting. CONCLUSIONS: Our results demonstrate characteristic protein expression profile in control and hypertrophy condition in SD rats and also expand the existing knowledge on differentially expressed proteins in hypertrophy. The study signifies the importance of reduced expression of a novel protein such as Prohibitin (PHB) which may be associated with the cardiomyocytes growth and cardiac hypertrophy. However, further work is necessary to confirm the role of PHB in human heart and its potential role in diagnostic and therapeutic intervention in the clinic.


Subject(s)
Cardiomegaly/metabolism , Isoproterenol/adverse effects , Proteomics/methods , Repressor Proteins/metabolism , Animals , Biomarkers/metabolism , Cardiomegaly/chemically induced , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Heart/physiopathology , Male , Myocytes, Cardiac/cytology , Prohibitins , Proteome , Rats , Rats, Sprague-Dawley , Signal Transduction , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...