Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Semin Cancer Biol ; 86(Pt 3): 899-913, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-34998944

RESUMO

The primary physiological process of respiration produces carbon dioxide (CO2) that reacts with water molecules which subsequently liberates bicarbonate (HCO-3) and protons. Carbonic anhydrases (CAs) are the primary catalyst involved in this conversion. More than 16 isoforms of human CAs show organ or subcellular specific activity. Dysregulation of each CA is associated with multiple pathologies. Out of these members, the overexpression of membrane-bound carbonic anhydrase IX (CAIX) is associated explicitly with hypoxic tumors or various solid cancers. CAIX helps tumors deal with higher CO2 by sequestering it with bicarbonate ions and helping cancer cells to grow in a comparatively hypoxic or acidic environment, thus acting as a pH adaptation switch. CAIX-mediated adaptations in cancer cells include angiogenesis, metabolic alterations, tumor heterogeneity, drug resistance, and regulation of cancer-specific chemokines. This review comprehensively collects and describe the cancer-specific expression mechanism and role of CAIX in cancer growth, progression, heterogeneity, and its structural insight to develop future combinatorial targeted cancer therapies.


Assuntos
Anidrases Carbônicas , Neoplasias , Humanos , Anidrase Carbônica IX/genética , Anidrase Carbônica IX/metabolismo , Dióxido de Carbono/metabolismo , Dióxido de Carbono/uso terapêutico , Anidrases Carbônicas/genética , Neoplasias/patologia , Antígenos de Neoplasias/metabolismo , Concentração de Íons de Hidrogênio , Quimiocinas/uso terapêutico
2.
Pharm Res ; 40(11): 2747-2758, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37833570

RESUMO

PURPOSE: There is growing interest in seeking pharmacological activation of neurolysin (Nln) for stroke treatment. Discovery of central nervous system drugs remains challenging due to the protection of the blood-brain barrier (BBB). The previously reported peptidomimetic Nln activators display unsatisfactory BBB penetration. Herein, we investigate the next generation of non-peptidomimetic Nln activators with high BBB permeability. METHODS: A BBB-mimicking model was used to evaluate their in vitro BBB permeability. Protein binding, metabolic stability, and efflux assays were performed to determine their unbound fraction, half-lives in plasma and brains, and dependence of BBB transporter P-glycoprotein (P-gp). The in vivo pharmacokinetic profiles were elucidated in healthy and stroke mice. RESULTS: Compounds KS52 and KS73 out of this generation exhibit improved peptidase activity and BBB permeability compared to the endogenous activator and previous peptidomimetic activators. They show reasonable plasma and brain protein binding, improved metabolic stability, and independence of P-gp-mediated efflux. In healthy animals, they rapidly distribute into brains and reach peak levels of 18.69% and 12.10% injected dose (ID)/ml at 10 min. After 4 h, their total brain concentrations remain 7.78 and 12.34 times higher than their A50(minimal concentration required for enhancing 50% peptidase activity). Moreover, the ipsilateral hemispheres of stroke animals show comparable uptake to the corresponding contralateral hemispheres and healthy brains. CONCLUSIONS: This study provides essential details about the pharmacokinetic properties of a new generation of potent non-peptidomimetic Nln activators with high BBB permeability and warrants the future development of these agents as potential neuroprotective pharmaceutics for stroke treatment.


Assuntos
Peptidomiméticos , Acidente Vascular Cerebral , Camundongos , Animais , Barreira Hematoencefálica/metabolismo , Peptidomiméticos/metabolismo , Metaloendopeptidases/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Permeabilidade
3.
J Cell Biochem ; 122(8): 897-910, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33829554

RESUMO

Cyclin-dependent kinase 6 (CDK6) is a member of serine/threonine kinase family, and its overexpression is associated with cancer development. Thus, it is considered as a potential drug target for anticancer therapies. This study showed the CDK6 inhibitory potential of vanillin using combined experimental and computational methods. Structure-based docking and 200 ns molecular dynamics simulation studies revealed that the binding of vanillin stabilizes the CDK6 structure and provides mechanistic insights into the binding mechanism. Enzyme inhibition and fluorescence-binding studies showed that vanillin inhibits CDK6 with an half maximal inhibitory concentration = 4.99 µM and a binding constant (K) 4.1 × 107 M-1 . Isothermal titration calorimetry measurements further complemented our observations. Studies on human cancer cell lines (MCF-7 and A549) showed that vanillin decreases cell viability and colonization properties. The protein expression studies have further revealed that vanillin reduces the CDK6 expression and induces apoptosis in the cancer cells. In conclusion, our study presents the CDK6-mediated therapeutic implications of vanillin for anticancer therapies.


Assuntos
Benzaldeídos , Neoplasias da Mama , Proliferação de Células/efeitos dos fármacos , Quinase 6 Dependente de Ciclina , Neoplasias Pulmonares , Simulação de Dinâmica Molecular , Proteínas de Neoplasias , Células A549 , Benzaldeídos/química , Benzaldeídos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Quinase 6 Dependente de Ciclina/química , Quinase 6 Dependente de Ciclina/metabolismo , Feminino , Células HEK293 , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/enzimologia , Células MCF-7 , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo
4.
Bioorg Med Chem ; 28(9): 115424, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32209296

RESUMO

Carbonic anhydrase IX (CAIX) is an emerging drug target for hypoxia associated cancers. To identify potent and selective inhibitors of CAIX, a small library of ferulic acid (FA) derivatives bearing triazole moiety has been designed, synthesized and evaluated against different human CA isoforms (CAII, CAVA & CAIX). Though most of the compounds showed CAIX inhibition in the micromolar range, compound 7i selectively inhibits CAIX in the nanomolar range (IC50 = 24 nM). In silico analysis revealed binding of 7i with the catalytically important amino acid residues of CAIX. Further, cell-based studies indicate that 7i inhibits the activity of CAIX, decreases the epithelial to mesenchymal transitions, induces apoptosis, inhibits cell migration and colonization potential of cancer cells. Taken together, these results emphasized the use of 7i as a prospective pharmacological lead molecule in CAIX targeted anticancer therapeutics.


Assuntos
Antineoplásicos/farmacologia , Inibidores da Anidrase Carbônica/farmacologia , Ácidos Cumáricos/farmacologia , Desenho de Fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Antígenos de Neoplasias , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Anidrase Carbônica IX , Inibidores da Anidrase Carbônica/síntese química , Inibidores da Anidrase Carbônica/química , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ácidos Cumáricos/síntese química , Ácidos Cumáricos/química , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Humanos , Estrutura Molecular , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade
5.
Bioorg Chem ; 98: 103754, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32200329

RESUMO

A diverse series of 1,2,4-oxadiazoles based substituted compounds were designed, synthesized and evaluated as anticancer agents targeting carbonic anhydrase IX (CAIX). Initial structure-activity analysis suggested that the thiazole/thiophene-sulfonamide conjugates of 1,2,4-oxadiazoles exhibited potent anticancer activities with low µM potencies. Compound OX12 exhibited antiproliferative activity (IC50 = 11.1 µM) along with appreciable inhibition potential for tumor-associated CAIX (IC50 = 4.23 µM) isoform. Therefore, OX12 was structurally optimized and its SAR oriented derivatives (OX17-27) were synthesized and evaluated. This iteration resulted in compound OX27 with an almost two-fold increase in antiproliferative effect (IC50 = 6.0 µM) comparable to the clinical drug doxorubicin and significantly higher potency against CAIX (IC50 = 0.74 µM). Additionally, OX27 treatment decreases the expression of CAIX, induces apoptosis and ROS production, inhibited colony formation and migration of colon cancer cells. Our studies provide preclinical rational for the further optimization of identified OX27 as a suitable lead for the possible treatment of CRC.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Oxidiazóis/farmacologia , Sulfonamidas/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular , Oxidiazóis/química , Relação Estrutura-Atividade , Sulfonamidas/química , Células Tumorais Cultivadas
6.
J Nat Prod ; 82(8): 2252-2261, 2019 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-31343173

RESUMO

Microtubule affinity regulating kinase 4 (MARK4) is a potential drug target for neuronal disorders and several types of cancers. Filtration of naturally occurring compound libraries using high-throughput screening and enzyme assay suggest α-mangostin is a potential inhibitor of MARK4. Structure-based docking and 100 ns molecular dynamics simulation revealed that the binding of α-mangostin stabilizes the MARK4 structure. Enzyme inhibition and binding studies showed that α-mangostin inhibited MARK4 in the submicromolar range with IC50 = 1.47 µM and binding constant (Ka) 5.2 × 107 M-1. Cell-based studies suggested that α-mangostin inhibited the cell viability (MCF-7 and HepG2), induced apoptosis, arrested the cell cycle in the G0/G1 phase, and reduced tau-phosphorylation. This study implicates MARK4 as a new target of α-mangostin, adding an additional lead molecule to the anticancer repertoire.


Assuntos
Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Xantonas/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Ligantes , Simulação de Acoplamento Molecular , Estrutura Secundária de Proteína , Espécies Reativas de Oxigênio/metabolismo
7.
J Ind Microbiol Biotechnol ; 46(12): 1669-1683, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31531745

RESUMO

Due to the potential toxicity of mercury, there is an immediate need to understand its uptake, transport and flux within living cells. Conventional techniques used to analyze Hg2+ are invasive, involve high cost and are less sensitive. In the present study, a highly efficient genetically encoded mercury FRET sensor (MerFS) was developed to measure the cellular dynamics of Hg2+ at trace level in real time. To construct MerFS, the periplasmic mercury-binding protein MerP was sandwiched between enhanced cyan fluorescent protein (ECFP) and venus. MerFS is pH stable, offers a measurable fluorescent signal and binds to Hg2+ with high sensitivity and selectivity. Mutant MerFS-51 binds with an apparent affinity (Kd) of 5.09 × 10-7 M, thus providing a detection range for Hg2+ quantification between 0.210 µM and 1.196 µM. Furthermore, MerFS-51 was targeted to Escherichia coli (E. coli), yeast and human embryonic kidney (HEK)-293T cells that allowed dynamic measurement of intracellular Hg2+ concentration with a highly responsive saturation curve, proving its potential application in cellular systems.


Assuntos
Transferência Ressonante de Energia de Fluorescência , Mercúrio/análise , Transporte Biológico , Sobrevivência Celular , Escherichia coli/química , Células HEK293 , Humanos , Espaço Intracelular/química , Saccharomyces cerevisiae/química
8.
Appl Microbiol Biotechnol ; 101(9): 3513-3536, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28361239

RESUMO

Chitin is a long unbranched polysaccharide, made up of ß-1,4-linked N-acetylglucosamine which forms crystalline fiber-like structure. It is present in the fungal cell walls, insect and crustacean cuticles, nematode eggshells, and protozoa cyst. We provide a critical appraisal on the chemical modifications of chitin and its derivatives in the context of their improved efficacy in medical applications without any side effect. Recent advancement in nanobiotechnology has helped to synthesize several chitin derivatives having significant biological applications. Here, we discuss the molecular diversity of chitin and its applications in enzyme immobilization, wound healing, packaging material, controlled drug release, biomedical imaging, gene therapy, agriculture, biosensor, and cosmetics. Also, we highlighted chitin and its derivatives as an antioxidant, antimicrobial agent, anticoagulant material, food additive, and hypocholesterolemic agent. We envisage that chitin and chitosan-based nanomaterials with their potential applications would augment nanobiotechnology and biomedical industries.


Assuntos
Materiais Biocompatíveis/química , Materiais Biocompatíveis/metabolismo , Produtos Biológicos/química , Produtos Biológicos/metabolismo , Quitina/química , Quitina/metabolismo , Nanoestruturas/química
9.
ACS Omega ; 9(24): 25945-25959, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38911744

RESUMO

Microtubule affinity-regulating kinase 4 (MARK4) is a serine-threonine kinase that phosphorylates microtubule-associated proteins (MAPs) and increases the microtubule dynamics. Due to its direct involvement in initiation, cell division, progression, and cancer metastasis, MARK4 is considered a potential therapeutic target. Here, we designed, synthesized, and characterized vanillin-isatin hybrids and evaluated their MARK4 inhibitory potential. All of the compounds strongly bind to MARK4 and interact closely with the active site residues. Finally, the compound VI-9 was selected for further investigation due to its high binding affinity and strong MARK4 inhibitory potential. Tau-phosphorylation assay has further confirmed that VI-9 significantly reduced the activity of MARK4. Compared with vanillin, VI-9 showed a better binding affinity and MARK4 inhibitory potential. Cell viability assays on human hepatocellular carcinoma (HCC) cell lines C3A and SNU-475 revealed that VI-9 inhibited their growth and proliferation. In addition, these compounds were nontoxic (up to 200 µM) for noncancerous (HEK-293) cells. Interestingly, VI-9 induces apoptosis and decreases the metastatic potential of the C3A and SNU-475 cell lines. The present work opens a newer avenue for vanillin-isatin hybrids and their derivatives in developing MARK4-targeted anticancer therapies.

10.
ACS Med Chem Lett ; 15(4): 510-517, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38628788

RESUMO

The peptidase neurolysin (Nln) has been validated as a potential target for developing therapeutics for ischemic stroke (IS). Overexpression of Nln in a mouse model of IS provides significant cerebroprotection, leading to reduced infarction size and edema volume. Pharmacological inhibition of Nln in the post-stroke brain worsens neurological outcomes. A virtual screen identified dipeptide small-molecule activators of Nln. Optimization studies resulted in a class of peptidomimetic compounds with promising activity. However, these compounds still possessed an amide bond that compromised their stability in plasma and the brain. Herein, we report the synthesis and characterization of a series of amide bioisosteres based on our peptidomimetic leads. Imidazole-based bioisosteres afford scaffolds with increased potency to activate Nln combined with enhanced mouse plasma stability and significantly better brain permeability over the original dipeptide hits.

11.
J Biomol Struct Dyn ; 40(7): 3144-3154, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-33183174

RESUMO

Inhibitors of carbonic anhydrase (CAIs) hold promise for addressing various diseases, including cancer, diabetes, and other metabolic syndromes. CAV is the only isoform present in the mitochondria and is considered a potential drug target for obesity. In this work, we have developed C2, and C4 substituted oxazole-5(4H)-one derivatives as a new scaffold for the selective inhibition of human carbonic anhydrase VA (hCAVA). Synthesized compounds were characterized by 1H NMR, 13C NMR, and LC-MS mass spectrometry and subsequently evaluated for in vitro hCAVA inhibition. Two compounds, 4 and 5 showed a considerably higher binding affinity for hCAVA in comparison to the hCAII. Further, cell-based studies showed that these compounds decrease the expression of CAVA and GLUT4 in adipocytes and non-toxic to HEK293 cells. The present work opens a platform for the use of oxazole-5(4H)-ones and holds promise for further refinement of potent and selective hCAVA inhibitors.Communicated by Ramaswamy H. Sarma.


Assuntos
Anidrases Carbônicas , Diabetes Mellitus , Anidrase Carbônica IX , Inibidores da Anidrase Carbônica/química , Inibidores da Anidrase Carbônica/farmacologia , Células HEK293 , Humanos , Obesidade/tratamento farmacológico , Oxazolona/uso terapêutico , Relação Estrutura-Atividade
12.
Int J Biol Macromol ; 161: 596-604, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-32535203

RESUMO

Microtubule affinity regulating kinase (MARK4) is considered as a potential drug target for diabetes, cancer, and neurodegenerative diseases. Since the role of MARK4 in the phosphorylation of tau protein and subsequently Alzheimer's disease has been established, therefore, we have investigated the binding affinity and MARK4 inhibitory potential of cholic acid (CHA) using both computational and spectroscopic methods. Molecular docking suggested a strong binding of CHA to the functionally important residues of MARK4. We further performed 500 ns molecular dynamics simulation which suggested the MARK4-CHA system was quite stable throughout the simulation trajectory. CHA potential binds to the MARK4 with a binding constant (K) of 107 M-1 at 288 K. Further, MARK4 activity was inhibited by CHA with an IC50 = 5.5 µM. Further insights into the thermodynamic parameters suggested that MARK4-CHA complex formation is driven by both electrostatic and van der Waals interactions. Overall study provides a rationale to use CHA in the drug development via MARK4 inhibition, towards possible therapeutic implications in neurodegenerative diseases.


Assuntos
Ácido Cólico/química , Doenças Neurodegenerativas/tratamento farmacológico , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Humanos , Microtúbulos/química , Simulação de Acoplamento Molecular/métodos , Simulação de Dinâmica Molecular , Fosforilação/efeitos dos fármacos , Ligação Proteica
13.
Cells ; 9(2)2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-32033110

RESUMO

Telomere comprises the ends of eukaryotic linear chromosomes and is composed of G-rich (TTAGGG) tandem repeats which play an important role in maintaining genome stability, premature aging and onsets of many diseases. Majority of the telomere are replicated by conventional DNA replication, and only the last bit of the lagging strand is synthesized by telomerase (a reverse transcriptase). In addition to replication, telomere maintenance is principally carried out by two key complexes known as shelterin (TRF1, TRF2, TIN2, RAP1, POT1, and TPP1) and CST (CDC13/CTC1, STN1, and TEN1). Shelterin protects the telomere from DNA damage response (DDR) and regulates telomere length by telomerase; while, CST govern the extension of telomere by telomerase and C strand fill-in synthesis. We have investigated both structural and biochemical features of shelterin and CST complexes to get a clear understanding of their importance in the telomere maintenance. Further, we have analyzed ~115 clinically important mutations in both of the complexes. Association of such mutations with specific cellular fault unveils the importance of shelterin and CST complexes in the maintenance of genome stability. A possibility of targeting shelterin and CST by small molecule inhibitors is further investigated towards the therapeutic management of associated diseases. Overall, this review provides a possible direction to understand the mechanisms of telomere borne diseases, and their therapeutic intervention.


Assuntos
Doença/genética , Mutação/genética , Nucleoproteínas/química , Homeostase do Telômero , Proteínas de Ligação a Telômeros/química , Animais , Humanos , Modelos Biológicos , Complexo Shelterina
14.
Biomedicines ; 8(5)2020 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-32422877

RESUMO

Design and development of potential pyruvate dehydrogenase kinase 3 (PDK3) inhibitors have gained attention because of their possible therapeutic uses in lung cancer therapy. In the present study, the binding affinity of naturally occurring alkaloids, hordenine, vincamine, tryptamine, cinchonine, and colcemid was measured with PDK3. The molecular docking and fluorescence binding studies suggested that all these compounds show a considerable binding affinity for PDK3. Among them, the affinity of hordenine to the PDK3 was excellent (K = 106 M-1) which was further complemented by isothermal titration calorimetric measurements. Hordenine binds in the active site pocket of PDK3 and forms a significant number of non-covalent interactions with functionally important residues. All-atom molecular dynamics (MD) simulation study suggested that the PDK3-hordenine complex is stabilized throughout the trajectory of 100ns and leads to fewer conformational changes. The enzyme inhibition studies showed that hordenine inhibits the activity of PDK3 with an IC50 value of 5.4 µM. Furthermore, hordenine showed a cytotoxic effect on human lung cancer cells (A549 and H1299) with an admirable IC50 value. However, it did not inhibit the growth of HEK293 cells up to 200 µM, indicating its non-toxicity to non-cancerous cell lines. In summary, our findings provide the basis for the therapeutic implication of hordenine and its derivatives in lung cancer and PDK3-related diseases after required in vivo validation.

15.
Sci Rep ; 10(1): 10300, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32587267

RESUMO

Microtubule affinity regulating kinase (MARK4) is a potential drug target for different types of cancer as it controls the early step of cell division. In this study, we have screened a series of natural compounds and finally identified rosmarinic acid (RA) as a potential inhibitor of MARK4. Molecular docking and 500 ns all-atom simulation studies suggested that RA binds to the active site pocket of MARK4, forming enough number of non-covalent interactions with critical residues and MARK4-RA complex is stable throughout the simulation trajectory. RA shows an excellent binding affinity to the MARK4 with a binding constant (K) of 107 M-1. Furthermore, RA significantly inhibits MARK4 activity (IC50 = 6.204 µM). The evaluation of enthalpy change (∆H) and entropy change (∆S) suggested that the MARK4-RA complex formation is driven by hydrogen bonding and thus complexation process is seemingly specific. The consequence of MARK4 inhibition by RA was further evaluated by cell-based tau-phosphorylation studies, which suggested that RA inhibited the phosphorylation of tau. The treatment of cancer cells with RA significantly controls cell growth and subsequently induces apoptosis. Our study provides a rationale for the therapeutic evaluation of RA and RA-based inhibitors in MARK4 associated cancers and other diseases.


Assuntos
Cinamatos/farmacologia , Depsídeos/farmacologia , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cinamatos/química , Cinamatos/uso terapêutico , Depsídeos/química , Depsídeos/uso terapêutico , Células HEK293 , Humanos , Simulação de Acoplamento Molecular , Neoplasias/patologia , Fosforilação/efeitos dos fármacos , Ligação Proteica , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas tau/metabolismo , Ácido Rosmarínico
16.
Sci Rep ; 9(1): 1676, 2019 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-30737440

RESUMO

Microtubule affinity regulating kinase 4 (MARK4) becomes a unique anti-cancer drug target as its overexpression is responsible for different types of cancers. In quest of novel, effective MARK4 inhibitors, some acridone derivatives were synthesized, characterized and evaluated for inhibitory activity against human MARK4. Among all the synthesized compounds, three (7b, 7d and 7f) were found to have better binding affinity and enzyme inhibition activity in µM range as shown by fluorescence binding, ITC and kinase assays. Here we used functional assays of selected potential lead molecules with commercially available panel of 26 kinases of same family. A distinctive kinase selectivity profile was observed for each compound. The selective compounds were identified with submicromolar cellular activity against MARK4. Furthermore, in vitro antitumor evaluation against cancerous cells (MCF-7 and HepG2) revealed that compounds 7b, 7d and 7f inhibit cell proliferation and predominantly induce apoptosis in MCF-7 cells, with IC50 values of 5.2 ± 1.2 µM, 6.3 ± 1.2 µM, and 5.8 ± 1.4 µM respectively. In addition, these compounds significantly upsurge the oxidative stress in cancerous cells. Our observations support our approach for the synthesis of effective inhibitors against MARK4 that can be taken forward for the development of novel anticancer molecules targeting MARK4.


Assuntos
Acridonas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Acridonas/química , Sítios de Ligação , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células HEK293 , Células Hep G2 , Humanos , Células MCF-7 , Modelos Moleculares , Simulação de Acoplamento Molecular , Estrutura Molecular , Conformação Proteica , Inibidores de Proteínas Quinases/química
17.
Eur J Med Chem ; 163: 840-852, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30579124

RESUMO

Microtubule affinity-regulating kinase 4 (MARK4) is a potential drug target as the same is found to be over expressed in several types of cancers. In search of effective MARK4 inhibitors, we have synthesized and characterized Isatin-triazole hydrazones (9a-i) and evaluated their inhibitory potential. Of all the compounds, 9g showed better binding affinity and enzyme inhibition potential in sub micromolar range. Human serum albumin (HSA) binding assay suggested an easy transportation of 9g in blood stream due to its binding affinity. In vitro anticancer studies performed on MCF-7, MDA-MB-435s and HepG2 cells using 9g showed inhibition of cell proliferation and cell migration. Further, 9g induces apoptosis in these cancerous cells, with IC50 values of 6.22, 9.94 and 8.14 µM, respectively. Putatively, 9g seems to cause oxidative stress resulting in apoptosis. Functional assay of 9g with a panel of 26 kinases showed MARK4 specific profile. In conclusion, 9g seems to possess an effective inhibitory potential towards MARK4 adding an additional repertoire to anticancer therapeutics.


Assuntos
Hidrazonas/uso terapêutico , Isatina/uso terapêutico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Triazóis/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Hep G2 , Humanos , Hidrazonas/química , Hidrazonas/farmacologia , Isatina/química , Isatina/farmacologia , Células MCF-7 , Metástase Neoplásica/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , Triazóis/química , Triazóis/farmacologia
18.
Curr Protein Pept Sci ; 19(9): 909-923, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28799503

RESUMO

Obesity is a metabolic syndrome leading to several health problems such as hypertension, heart attack, type II diabetes, and even cancer. Carbonic anhydrase VA (CAVA) is a mitochondrial enzyme which is directly associated with the glucose homeostasis and considered as a promising target for obesity and other associated diseases in humans. So far, numerous inhibitors have been designed to inhibit the catalytic activity of CAVA with an assumption for its possible therapeutic uses against type II diabetes and other metabolic diseases. Among these, sulphonamide inhibitors and various non-classical inhibitors are extensively used. The focus of this review is to understand the mechanism and role CAVA in glucose homeostasis to ascertain as a potential drug target of obesity. We have further highlighted different types of inhibitors and their mode of binding and possible consequences with an aim to investigate possible therapeutic used for the treatment of obesity and associated diseases. Along with classical inhibitors, various non-classical inhibitors have proved to be potential inhibitors of CAV which may be employed to combat obesity. Certain phytochemicals are utilized as therapeutic molecules to fight obesity. These phytochemicals have been discussed in detail here.


Assuntos
Fármacos Antiobesidade/uso terapêutico , Inibidores da Anidrase Carbônica/uso terapêutico , Anidrase Carbônica V/antagonistas & inibidores , Obesidade/tratamento farmacológico , Sulfonamidas/uso terapêutico , Desenho de Fármacos , Humanos , Mitocôndrias/metabolismo , Estrutura Molecular , Obesidade/enzimologia , Compostos Fitoquímicos/uso terapêutico
19.
Int J Biol Macromol ; 106: 840-850, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28830777

RESUMO

To find potential inhibitors of human carbonic anhydrase IX (CAIX), we have successfully deigned, synthesized and characterized three p-toluene sulphonylhydrazone derivatives (1-3). Molecular docking studies provided the structural basis of CAIX inhibition and a deeper insight into the protein-ligand interactions. p-Toluene sulphonylhydrazone derivatives show a well organized conformational compatibility with the active site of CAIX. The protein-ligand complex was stabilized by several non-covalent interactions offered by residues present in the active site cavity. The actual binding affinity of synthesized compounds with CAIX was experimentally measured by fluorescence and isothermal titration calorimetry (ITC). Results of both fluorescence binding and ITC measurements show the binding affinity of p-Toluene sulphonylhydrazone derivatives to the CAIX in the µM range. CAIX enzyme inhibition assay showed the IC50 values in nM range. Though all the three compounds (1-3) showed a good binding with CAIX, compound 2 showed the best inhibition of CAIX activity. These compounds were non-toxic on normal cell lines (HEK-293) and significantly inhibit the proliferation of hypoxic cancer cells. All compounds induce apoptosis in the hypoxic cancer cells. These compounds may be further exploited as promising therapeutic agents to control the hypoxia-induced tumors.


Assuntos
Anidrase Carbônica IX/antagonistas & inibidores , Inibidores da Anidrase Carbônica/farmacologia , Hidrazonas/química , Neoplasias/tratamento farmacológico , Anidrase Carbônica IX/química , Inibidores da Anidrase Carbônica/síntese química , Inibidores da Anidrase Carbônica/química , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células HEK293 , Humanos , Hidrazonas/síntese química , Hidrazonas/farmacologia , Simulação de Acoplamento Molecular , Neoplasias/enzimologia , Tolueno/síntese química , Tolueno/química , Tolueno/farmacologia , Hipóxia Tumoral/efeitos dos fármacos
20.
Sci Rep ; 7(1): 9470, 2017 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-28842631

RESUMO

Microtubule affinity regulating kinase 4 (MARK4) is a Ser/Thr kinase belonging to AMPK-like family, has recently become an important drug target against cancer and neurodegenerative disorders. In this study, we have evaluated different natural dietary polyphenolics including rutin, quercetin, ferulic acid, hesperidin, gallic acid and vanillin as MARK4 inhibitors. All compounds are primarily binds to the active site cavity of MARK4. In silico observations were further complemented by the fluorescence-binding studies and isothermal titration calorimetry (ITC) measurements. We found that rutin and vanillin bind to MARK4 with a reasonably high affinity. ATPase and tau-phosphorylation assay further suggesting that rutin and vanillin inhibit the enzyme activity of MARK4 to a great extent. Cell proliferation, ROS quantification and Annexin-V staining studies are clearly providing sufficient evidences for the apoptotic potential of rutin and vanillin. In conclusion, rutin and vanillin may be considered as potential inhibitors for MARK4 and further exploited to design novel therapeutic molecules against MARK4 associated diseases.


Assuntos
Polifenóis/química , Polifenóis/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/química , Apoptose/efeitos dos fármacos , Sítios de Ligação , Proliferação de Células , Suplementos Nutricionais , Ligação de Hidrogênio , Modelos Moleculares , Conformação Molecular , Estrutura Molecular , Fosforilação , Ligação Proteica , Espécies Reativas de Oxigênio/metabolismo , Relação Estrutura-Atividade , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA