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.
Cell ; 185(11): 1943-1959.e21, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35545089

ABSTRACT

Parthanatos-associated apoptosis-inducing factor (AIF) nuclease (PAAN), also known as macrophage migration inhibitor factor (MIF), is a member of the PD-D/E(X)K nucleases that acts as a final executioner in parthanatos. PAAN's role in Parkinson's disease (PD) and whether it is amenable to chemical inhibition is not known. Here, we show that neurodegeneration induced by pathologic α-synuclein (α-syn) occurs via PAAN/MIF nuclease activity. Genetic depletion of PAAN/MIF and a mutant lacking nuclease activity prevent the loss of dopaminergic neurons and behavioral deficits in the α-syn preformed fibril (PFF) mouse model of sporadic PD. Compound screening led to the identification of PAANIB-1, a brain-penetrant PAAN/MIF nuclease inhibitor that prevents neurodegeneration induced by α-syn PFF, AAV-α-syn overexpression, or MPTP intoxication in vivo. Our findings could have broad relevance in human pathologies where parthanatos plays a role in the development of cell death inhibitors targeting the druggable PAAN/MIF nuclease.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Parkinson Disease , Animals , Brain/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Endonucleases/metabolism , Mice , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/metabolism
2.
Angew Chem Int Ed Engl ; 58(48): 17158-17162, 2019 11 25.
Article in English | MEDLINE | ID: mdl-31591797

ABSTRACT

Glucose transporters play an essential role in cancer cell proliferation and survival and have been pursued as promising cancer drug targets. Using microarrays of a library of new macrocycles known as rapafucins, which were inspired by the natural product rapamycin, we screened for new inhibitors of GLUT1. We identified multiple hits from the rapafucin 3D microarray and confirmed one hit as a bona fide GLUT1 ligand, which we named rapaglutin A (RgA). We demonstrate that RgA is a potent inhibitor of GLUT1 as well as GLUT3 and GLUT4, with an IC50 value of low nanomolar for GLUT1. RgA was found to inhibit glucose uptake, leading to a decrease in cellular ATP synthesis, activation of AMP-dependent kinase, inhibition of mTOR signaling, and induction of cell-cycle arrest and apoptosis in cancer cells. Moreover, RgA was capable of inhibiting tumor xenografts in vivo without obvious side effects. RgA could thus be a new chemical tool to study GLUT function and a promising lead for developing anticancer drugs.


Subject(s)
Antineoplastic Agents/chemistry , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Macrolides/pharmacology , Small Molecule Libraries/chemistry , A549 Cells , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Drug Screening Assays, Antitumor , Humans , MCF-7 Cells , Macrolides/chemistry , Molecular Structure , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Protein Array Analysis , Signal Transduction , Sirolimus/chemistry , Structure-Activity Relationship , TOR Serine-Threonine Kinases/metabolism , Tacrolimus/chemistry , Tacrolimus Binding Proteins
3.
Bioorg Chem ; 87: 484-494, 2019 06.
Article in English | MEDLINE | ID: mdl-30927589

ABSTRACT

A series of diindolylmethanes (5a-t) were designed, synthesized, and examined for their cytotoxicity against four human cancer cell lines like prostate (DU-145), lung (A549), breast (MCF-7) and cervical cancer (HeLa). These results revealed that among all the hybrids, two (5k and 5r) were identified and exhibited significant cytotoxic effect against A549 cancer cells with IC50 values of 1.65 ±â€¯0.3 and 1.80 ±â€¯0.8 µM respectively. To investigate the reasons for the cytotoxic activity, the conventional biological assays were carried out with 5k and 5r on the A549 cancer cells. Both hybrids led to the arrest of A549 cell lines at the G2/M phase of the cell cycle and strongly induced apoptosis. Further the apoptotic effects of 5k and 5r were confirmed by ROS, annexin-V FITC, and mitochondrial membrane potential. Moreover, structure-activity relationships were elucidated with various substitutions on these hybrids.


Subject(s)
Apoptosis/drug effects , Imidazoles/pharmacology , Indoles/pharmacology , Pyridines/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Imidazoles/chemistry , Indoles/chemistry , Membrane Potential, Mitochondrial/drug effects , Molecular Structure , Pyridines/chemistry , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
4.
Eur J Med Chem ; 126: 36-51, 2017 Jan 27.
Article in English | MEDLINE | ID: mdl-27744185

ABSTRACT

A series of imidazo[2,1-b]thiazole linked triazole conjugates were synthesized by using Huisgen 1,3-dipolar cyclo-addition reaction (click chemistry approach) and evaluated for their antiproliferative activity against some human cancer cell lines like, HeLa (cervical), DU-145 (prostate), A549 (lung), MCF-7 (breast) and HepG2 (liver). Among them, Conjugates 4g and 4h demonstrated a significant antiproliferative effect against human lung cancer cells (A549) with IC50 values of 0.92 and 0.78 µM respectively. Flow cytometric analysis revealed that these conjugates induced cell cycle arrest in G2/M phase in A549 lung cancer cells. The tubulin polymerization assay and immunofluorescence analysis showed that these conjugates effectively inhibit microtubule assembly in cell free and cell based (A549) experiment respectively. Moreover, the apoptosis inducing properties were evaluated by Hoechst staining, mitochondrial membrane potential and Annexin V-FITC assay. Further, western blot analysis was performed for proapoptotic protein Bax and antiapoptotic protein Bcl-2 and the results demonstrated that there was up regulation of Bax and down regulation of Bcl-2 suggesting that these compounds induced apoptosis in human lung cancer cells, A549.


Subject(s)
Drug Design , Microtubules/drug effects , Thiazoles/chemical synthesis , Thiazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Drug Screening Assays, Antitumor , Humans , Microtubules/metabolism , Models, Molecular , Protein Structure, Quaternary , Structure-Activity Relationship , Thiazoles/chemistry , Tubulin/chemistry , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology
5.
Food Chem Toxicol ; 87: 1-11, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26615871

ABSTRACT

We have synthesized a novel quinazolinone chalcone derivative (QC) and first time reported its in-vitro and in-vivo anticancer potential. It inhibited the cell proliferation of different cancer cell lines like PC-3, Panc-1, Mia-Paca-2, A549, MCF-7 and HCT-116. It induces apoptosis as measured by several biological endpoints such as apoptotic body formation, evident by Hoechst and scanning electron microscopy, enhanced annexinV-FITC binding of the cells, increased sub-G0 cell fraction, loss of mitochondrial membrane potential (Δψm), reduction of Bcl-2/Bax ratio, activation of caspase-9, caspase-3 and PARP-1 (poly-ADP Ribose polymerase) cleavage in HCT-116 cells. In spite of apoptosis, QC significantly hammers the downstream and upstream signaling cascade of PI3K/Akt/mTOR pathway and cell cycle regulator Skp-2, p21 and p27. Interestingly, QC induces the S and G2/M phase of HCT-116 cells at experimental doses. QC inhibits the tumor growth of Ehrlich ascites carcinoma (EAC), Ehrlich tumor (ET, solid) and sarcoma-180(solid) mice models. Furthermore, it was found to be non-toxic as no animal mortality (0/7) occurred during experimental doses. The present study provides an insight of anticancer potential of QC, which may be useful in managing and treating cancer.


Subject(s)
Apoptosis/drug effects , Chalcones/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinazolinones/pharmacology , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chalcones/chemistry , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Molecular Structure , Neoplasms, Experimental/drug therapy , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Quinazolinones/chemistry , Random Allocation , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics
6.
Org Biomol Chem ; 12(48): 9864-80, 2014 Dec 28.
Article in English | MEDLINE | ID: mdl-25354805

ABSTRACT

A series of imidazo[1,5-a]pyridine-benzimidazole hybrids (5a­aa) were prepared and evaluated for their cytotoxic activity against a panel of sixty human tumor cell lines. Among them compounds 5d and 5l showed significant cytotoxic activity with GI50 values ranging from 1.06 to 14.9 µM and 0.43 to 7.73 µM, respectively. Flow cytometric analysis revealed that these compounds arrest the cell cycle at G2/M phase and induced cell death by apoptosis. The tubulin polymerization assay (IC50 of 5d is 3.25 µM and 5l is 1.71 µM) and immunofluorescence analysis showed that these compounds effectively inhibited the microtubule assembly in human breast cancer cells (MCF-7). Further, the apoptotic effects of compounds were confirmed by Hoechst staining, mitochondrial membrane potential, cytochrome c release, ROS generation, caspase 9 activation and DNA fragmentation analysis. After treatment with these compounds for 48 h, p-PTEN and p-AKT levels were markedly decreased. Moreover, these compounds did not significantly inhibit the normal human embryonic kidney cells, HEK-293. The molecular docking simulations predicted the binding interactions of 5d and 5l with colchicine binding site of the tubulin, which is in compliance with the antiproliferative activity data.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyridines/pharmacology , Signal Transduction/drug effects , Tubulin Modulators/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Benzimidazoles/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Phosphatidylinositol 3-Kinases/metabolism , Polymerization/drug effects , Protein Kinases/chemical synthesis , Protein Kinases/chemistry , Protein Kinases/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/chemistry , Structure-Activity Relationship , Tubulin/metabolism
7.
Eur J Med Chem ; 46(6): 2427-35, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21481986

ABSTRACT

A series of oxindole derivatives of imidazo[1,5-a]pyrazines were prepared and confirmed by 1H NMR, mass and HRMS data. These compounds were evaluated for their anticancer activity against a panel of 52 human tumor cell lines derived from nine different cancer types: leukemia, lung, colon, CNS, melanoma, ovarian, renal, prostate and breast. Among them compound 7l showed significant anticancer activity with GI50 values ranging from 1.54 to 13.0 µM. Cell cycle arrest was observed in G0/G1 phase upon treatment of A549 cells with 6.5 µM (IC50) concentration of compound 7l and induced apoptosis. This was confirmed by Annexin V-FITC as well as DNA fragmentation analysis and interestingly this compound (7l) did not affect the normal cells.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Indoles/chemistry , Pyrazines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Molecular Structure , Oxindoles , Pyrazines/chemical synthesis , Pyrazines/chemistry , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...