Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancer Cell Int ; 24(1): 278, 2024 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-39113040

RESUMO

Exosomes are extracellular vesicles well known for facilitating cell-to-cell communication by distributing essential macromolecules like proteins, DNA, mRNA, lipids, and miRNA. These vesicles are abundant in fluids distributed throughout the body, including urine, blood, saliva, and even bile. They are important diagnostic tools for breast, lung, gastrointestinal cancers, etc. However, their application as cancer biomarkers has not yet been implemented in most parts of the world. In this review, we discuss how OMICs profiling of exosomes can be practiced by substituting traditional imaging or biopsy methods for cancer detection. Previous methods like extensive imaging and biopsy used for screening were expensive, mostly invasive, and could not easily provide early detection for various types of cancer. Exosomal biomarkers can be utilized for routine screening by simply collecting body fluids from the individual. We anticipate that the use of exosomes will be brought to light by the success of clinical trials investigating their potential to enhance cancer detection and treatment in the upcoming years.

3.
ACS Appl Bio Mater ; 7(1): 44-58, 2024 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-38108852

RESUMO

Epithelial-mesenchymal transition (EMT) is a fundamental process driving cancer metastasis, transforming non-motile cells into a motile population that migrates to distant organs and forms secondary tumors. In recent years, cancer research has revealed a strong connection between exosomes and the EMT. Exosomes, a subpopulation of extracellular vesicles, facilitate cellular communication and dynamically regulate various aspects of cancer metastasis, including immune cell suppression, extracellular matrix remodeling, metastasis initiation, EMT initiation, and organ-specific metastasis. Tumor-derived exosomes (TEXs) and their molecular cargo, comprising proteins, lipids, nucleic acids, and carbohydrates, are essential components that promote EMT in cancer. TEXs miRNAs play a crucial role in reprogramming the tumor microenvironment, while TEX surface integrins contribute to organ-specific metastasis. Exosome-based cancer metastasis research offers a deeper understanding about cancer and an effective theranostic platform development. Additionally, various therapeutic sources of exosomes are paving the way for innovative cancer treatment development. In this Review, we spotlight the role of exosomes in EMT and their theranostic impact, aiming to inspire cancer researchers worldwide to explore this fascinating field in more innovative ways.


Assuntos
Exossomos , Vesículas Extracelulares , MicroRNAs , Neoplasias , Humanos , Exossomos/genética , Exossomos/metabolismo , Neoplasias/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Transição Epitelial-Mesenquimal , Microambiente Tumoral
4.
J Med Virol ; 95(10): e29135, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37792364

RESUMO

Personalized cancer immunotherapies, combined with nanotechnology (nano-vaccines), are revolutionizing cancer treatment strategies, explicitly targeting Human papilloma virus (HPV)-related cancers. Despite the availability of preventive vaccines, HPV-related cancers remain a global concern. Personalized cancer nano-vaccines, tailored to an individual's tumor genetic mutations, offer a unique and promising solution. Nanotechnology plays a critical role in these vaccines by efficiently delivering tumor-specific antigens, enhancing immune responses, and paving the way for precise and targeted therapies. Recent advancements in preclinical models have demonstrated the potential of polymeric nanoparticles and high-density lipoprotein-mimicking nano-discs in augmenting the efficacy of personalized cancer vaccines. However, challenges related to optimizing the nano-carrier system and ensuring safety in human trials persist. Excitingly, the integration of nanotechnology with Proteolysis-Targeting Chimeras (PROTACs) provides an additional avenue to enhance the effectiveness of personalized cancer treatment. PROTACs selectively degrade disease-causing proteins, amplifying the impact of nanotechnology-based therapies. Overcoming these challenges and leveraging the synergistic potential of nanotechnology, PROTACs, and Proteolysis-Targeting Antibodies hold great promise in pursuing novel and effective therapeutic solutions for individuals affected by HPV-related cancers.


Assuntos
Vacinas Anticâncer , Neoplasias , Infecções por Papillomavirus , Humanos , Quimera de Direcionamento de Proteólise , Papillomavirus Humano , Infecções por Papillomavirus/complicações , Infecções por Papillomavirus/tratamento farmacológico , Proteólise , Neoplasias/terapia
5.
ACS Omega ; 8(40): 36614-36627, 2023 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-37841156

RESUMO

Ovarian cancer (OC) is a common gynecological cancer worldwide. Unfortunately, the lack of early detection methods translates into a substantial cohort of women grappling with the pressing health crisis. The discovery of extracellular vesicles (EVs) (their major subpopulation exosomes, microvesicles, and apoptotic bodies) has provided new insights into the understanding of cancer. Exosomes, a subpopulation of EVs, play a crucial role in cellular communication and reflect the cellular status under both healthy and pathological conditions. Tumor-derived exosomes (TEXs) dynamically influence ovarian cancer progression by regulating uncontrolled cell growth, immune suppression, angiogenesis, metastasis, and the development of drug and therapeutic resistance. In the field of OC diagnostics, TEXs offer potential biomarkers in various body fluids. On the other hand, exosomes have also shown promising abilities to cure ovarian cancer. In this review, we address the interlink between exosomes and ovarian cancer and explore their theragnostic signature. Finally, we highlight future directions of exosome-based ovarian cancer research.

6.
ACS Biomater Sci Eng ; 9(9): 5205-5221, 2023 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-37578350

RESUMO

Glioblastoma (GBM) is an aggressive type of cancer that has led to the death of a large population. The traditional approach fails to develop a solution for GBM's suffering life. Extensive research into tumor microenvironments (TME) indicates that TME extracellular vesicles (EVs) play a vital role in cancer development and progression. EVs are classified into microvacuoles, apoptotic bodies, and exosomes. Exosomes are the most highlighted domains in cancer research. GBM cell-derived exosomes participate in multiple cancer progression events such as immune suppression, angiogenesis, premetastatic niche formation (PMN), ECM (extracellular matrix), EMT (epithelial-to-mesenchymal transition), metastasis, cancer stem cell development and therapeutic and drug resistance. GBM exosomes also carry the signature of a glioblastoma-related status. The exosome-based GBM examination is part of the new generation of liquid biopsy. It also solved early diagnostic limitations in GBM. Traditional therapeutic approaches do not cross the blood-brain barrier (BBB). Exosomes are a game changer in GBM treatment and it is emerging as a potential platform for effective, efficient, and specific therapeutic development. In this review, we have explored the exosome-GBM interlink, the clinical impact of exosomes on GBM biomarkers, the therapeutics signature of exosomes in GBM, exosome-based research challenges, and future directions in GBM. Therefore, the GBM-derived exosomes offer unique therapeutic opportunities, which are currently under preclinical and clinical testing.


Assuntos
Exossomos , Vesículas Extracelulares , Glioblastoma , Humanos , Glioblastoma/terapia , Glioblastoma/diagnóstico , Glioblastoma/patologia , Exossomos/patologia , Medicina de Precisão , Vesículas Extracelulares/patologia , Diferenciação Celular , Microambiente Tumoral
7.
ACS Appl Bio Mater ; 6(7): 2576-2590, 2023 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-37314223

RESUMO

Cancer is a complex deadly disease that has caused a global health crisis in recent epochs. Colorectal cancer (CRC) is the third most common malignant gastrointestinal disease. It has led to high mortality due to early diagnostic failure. Extracellular vesicles (EVs) come with promising solutions for CRC. Exosomes (a subpopulation of EVs) play a vital role as signaling molecules in CRC tumor microenvironment. It is secreted from all active cells. Exosome-based molecular transport (DNA, RNA, proteins, lipids, etc.) transforms the recipient cell's nature. In CRC, tumor cell-derived exosomes (TEXs) regulate multiple events of CRC development and progression such as immunogenic suppression, angiogenesis, epithelial-mesenchymal transitions (EMT), physical changes in the extracellular matrix (ECM), and metastasis. Biofluid-circulated tumor-derived exosomes (TEXs) are a potential tool for CRC liquid biopsy. Exosome-based colorectal cancer detection creates a great impact in CRC biomarker research. The exosome-associated CRC theranostics approach is a state-of-the-art method. In this review, we address the CRC and exosomes complex associated with cancer development and progression, the impact of exosomes on CRC screening (diagnostic and prognostic biomarkers), and also highlight several exosomes with CRC clinical trials, as well as future directions of exosome-based CRC research. Hopefully, it will encourage several researchers to develop a potential exosome-based theranostic tool to fight CRC.


Assuntos
Neoplasias Colorretais , Exossomos , Vesículas Extracelulares , Humanos , Exossomos/genética , Exossomos/metabolismo , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/metabolismo , Transdução de Sinais , Microambiente Tumoral
8.
Biomarkers ; 28(6): 502-518, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37352015

RESUMO

Breast cancer (BC) remains the most challenging global health crisis of the current decade, impacting a large population of females annually. In the field of cancer research, the discovery of extracellular vesicles (EVs), specifically exosomes (a subpopulation of EVs), has marked a significant milestone. In general, exosomes are released from all active cells but tumour cell-derived exosomes (TDXs) have a great impact (TDXs miRNAs, proteins, lipid molecules) on cancer development and progression. TDXs regulate multiple events in breast cancer such as tumour microenvironment remodelling, immune cell suppression, angiogenesis, metastasis (EMT-epithelial mesenchymal transition, organ-specific metastasis), and therapeutic resistance. In BC, early detection is the most challenging event, exosome-based BC screening solved the problem. Exosome-based BC treatment is a sign of the transforming era of liquid biopsy, it is also a promising therapeutic tool for breast cancer. Exosome research goes to closer precision oncology via a single exosome profiling approach. Our hope is that this review will serve as motivation for researchers to explore the field of exosomes and develop an efficient, and affordable theranostics approach for breast cancer.


Assuntos
Neoplasias da Mama , Exossomos , MicroRNAs , Feminino , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/genética , Neoplasias da Mama/terapia , Medicina de Precisão , Relevância Clínica , Exossomos/genética , Exossomos/patologia , Microambiente Tumoral/genética
10.
J Cell Mol Med ; 27(11): 1603-1607, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37183560

RESUMO

This short communication will enlighten the readers about the exosome and the epithelial-mesenchymal transition (EMT) related to several complicated events. It also highlighted the therapeutic potential of exosomes against EMT. Exosome toxicology, exosome heterogeneity, and a single exosome profiling approach are also covered in this article. In the future, exosomes could help us get closer to cancer vaccine and precision oncology.


Assuntos
Exossomos , Neoplasias , Humanos , Transição Epitelial-Mesenquimal , Transdução de Sinais , Medicina de Precisão
11.
Phytother Res ; 37(7): 3097-3120, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37246823

RESUMO

Phytoestrogens are plant secondary metabolite that is structurally and functionally similar to mammalian estrogens, which have been shown to have various health benefits in humans. Isoflavones, coumestans, and lignans are the three major bioactive classes of phytoestrogens. It has a complicated mechanism of action involving an interaction with the nuclear estrogen receptor isoforms ERα and ERß, with estrogen agonist and estrogen antagonist effects. Depending on their concentration and bioavailability in various plant sources, phytoestrogens can act as estrogen agonist or antagonists. Menopausal vasomotor symptoms, breast cancer, cardiovascular disease, prostate cancer, menopausal symptoms, and osteoporosis/bone health have all been studied using phytoestrogens as an additional standard hormone supplemental remedy. The botanical sources, techniques of identification, classification, side effects, clinical implications, pharmacological and therapeutic effects of their proposed mode of action, safety issues, and future directions for phytoestrogens have all been highlighted in this review.


Assuntos
Neoplasias da Mama , Isoflavonas , Neoplasias da Próstata , Animais , Masculino , Humanos , Fitoestrógenos/farmacologia , Fitoestrógenos/uso terapêutico , Estrogênios/uso terapêutico , Receptores de Estrogênio/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Próstata/tratamento farmacológico , Isoflavonas/farmacologia , Isoflavonas/uso terapêutico , Mamíferos/metabolismo
12.
ACS Biomater Sci Eng ; 9(2): 577-594, 2023 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-36621949

RESUMO

Exosomes are the phospholipid-membrane-bound subpopulation of extracellular vesicles derived from the plasma membrane. The main activity of exosomes is cellular communication. In cancer, exosomes play an important rolefrom two distinct perspectives, one related to carcinogenesis and the other as theragnostic and drug delivery tools. The outer phospholipid membrane of Exosome improves drug targeting efficiency. . Some of the vital features of exosomes such as biocompatibility, low toxicity, and low immunogenicity make it a more exciting drug delivery system. Exosome-based drug delivery is a new innovative approach to cancer treatment. Exosome-associated biomarker analysis heralded a new era of cancer diagnostics in a more specific way. This Review focuses on exosome biogenesis, sources, isolation, interrelationship with cancer and exosome-related cancer biomarkers, drug loading methods, exosome-based biomolecule delivery, advances and limitations of exosome-based drug delivery, and exosome-based drug delivery in clinical settings studies. The exosome-based understanding of cancer will change the diagnostic and therapeutic approach in the future.


Assuntos
Exossomos , Vesículas Extracelulares , Neoplasias , Humanos , Exossomos/metabolismo , Medicina de Precisão , Sistemas de Liberação de Medicamentos/métodos , Vesículas Extracelulares/metabolismo , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Fosfolipídeos/metabolismo , Fosfolipídeos/uso terapêutico
14.
Mol Cell ; 71(6): 1092-1104.e5, 2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30174291

RESUMO

Activation of class I phosphatidylinositol 3-kinase (PI3K) leads to formation of phosphatidylinositol-3,4,5-trisphophate (PIP3) and phosphatidylinositol-3,4-bisphophate (PI34P2), which spatiotemporally coordinate and regulate a myriad of cellular processes. By simultaneous quantitative imaging of PIP3 and PI34P2 in live cells, we here show that they have a distinctively different spatiotemporal distribution and history in response to growth factor stimulation, which allows them to selectively induce the membrane recruitment and activation of Akt isoforms. PI34P2 selectively activates Akt2 at both the plasma membrane and early endosomes, whereas PIP3 selectively stimulates Akt1 and Akt3 exclusively at the plasma membrane. These spatiotemporally distinct activation patterns of Akt isoforms provide a mechanism for their differential regulation of downstream signaling molecules. Collectively, our studies show that different spatiotemporal dynamics of PIP3 and PI34P2 and their ability to selectively activate key signaling proteins allow them to mediate class I PI3K signaling pathways in a spatiotemporally specific manner.


Assuntos
Imagem Óptica/métodos , Fosfatos de Fosfatidilinositol/fisiologia , Imagem Individual de Molécula/métodos , Animais , Linhagem Celular , Membrana Celular , Humanos , Fosfatos de Inositol , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Fosfatos de Fosfatidilinositol/metabolismo , Fosfatidilinositóis , Isoformas de Proteínas , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
15.
Mol Biosyst ; 12(3): 747-57, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26726010

RESUMO

Lamellipodin (Lpd) protein plays an important role in the formation of lamellipodial protrusion which is crucial in actin dynamics, cell polarity and motility. Lpd promotes actin polymerization with the help of members of the Ena/VASP family of actin regulators and tethering them to actin filaments. It is well documented that Lpd protein interacts with the membrane containing phosphatidylinositols through its pleckstrin homology (PH) domain and regulates several cellular functions and cell migration. However, the molecular mechanism that underlies how the PH domain of Lpd specifically gets recruited to phosphatidylinositols remains unclear. To understand their interaction properties, we quantitatively determined the binding parameters of the Lpd-PH domain employing a number of biophysical studies including surface plasmon resonance (SPR), fluorescence resonance energy transfer (FRET)-based competitive binding assay and monolayer penetration measurements. Our studies showed that the Lpd-PH domain strongly interacts with PI(3,4)P2 containing liposome without any membrane penetration. Mutational studies demonstrate that the presence of cationic residues within the phosphatidylinositol (PIP) binding site of the Lpd-PH domain is essential in membrane binding. The translocation patterns of the Lpd-PH domain and mutants in platelet-derived growth factor (PDGF) stimulated A549 cells are in good agreement with our in vitro binding measurements. Overall, these studies demonstrate an insight into how the Lpd-PH domain regulates cellular signals in a PI(3,4)P2 dependent manner.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Sequência de Aminoácidos , Becaplermina , Sítios de Ligação , Ligação Competitiva , Transferência Ressonante de Energia de Fluorescência , Proteínas de Fluorescência Verde/metabolismo , Humanos , Cinética , Lipossomos/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/isolamento & purificação , Proteínas Mutantes/metabolismo , Mutação/genética , Domínios Proteicos , Proteínas Proto-Oncogênicas c-sis/farmacologia , Ressonância de Plasmônio de Superfície , Temperatura
16.
Mol Biosyst ; 11(5): 1389-99, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25820877

RESUMO

The protein kinase C (PKC) family of proteins is an attractive drug target. Dysregulation of PKC-dependent signalling pathways is related to several human diseases like cancer, immunological and other diseases. We approached the problem of altering PKC activities by developing C1 domain-based PKC ligands. In this report γ-hydroxymethyl-γ-butyrolactone (HGL) substituents were investigated in an effort to develop small molecule-based PKC regulators with higher specificity for C1 domain than the endogenous diacylglycerols (DAGs). Extensive analysis of membrane-ligands interaction measurements revealed that the membrane-active compounds strongly interact with the lipid bilayers and the hydrophilic parts of compounds localize at the bilayer/water interface. The pharmacophores like hydroxymethyl, carbonyl groups and acyl-chain length of the compounds are crucial for their interaction with the C1 domain proteins. The potent compounds showed more than 17-fold stronger binding affinity for the C1 domains than DAG under similar experimental conditions. Nonradioactive kinase assay confirmed that these potent compounds have similar or better PKC dependent phosphorylation capabilities than DAG under similar experimental conditions. Hence, our findings reveal that these HGL analogues represent an attractive group of structurally simple C1 domain ligands that can be further structurally altered to improve their potencies.


Assuntos
4-Butirolactona/análogos & derivados , 4-Butirolactona/química , Modelos Moleculares , Domínios e Motivos de Interação entre Proteínas , Proteína Quinase C/química , 4-Butirolactona/metabolismo , Ativação Enzimática , Humanos , Cinética , Ligantes , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Lipossomos/química , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Estrutura Molecular , Ligação Proteica , Proteína Quinase C/metabolismo , Solubilidade , Relação Estrutura-Atividade
17.
Dalton Trans ; 41(25): 7573-81, 2012 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-22588369

RESUMO

Cu(II) complexes of three tridentate ligands, L(1), L(2) and L(3), [L(1), N-((1H-imidazole-2-yl)methyl)-2-(pyridine-2-yl)ethanamine; L(2), N-((1-methyl-1H-imidazole-2-yl)methyl)-2-(pyridine-2-yl)ethanamine; L(3), 2-(pyridine-2-yl)-N-((pyridine-2-yl)methyl)ethanamine] respectively, were synthesized and characterized. The single crystal X-ray structure of complex 1 reveals the pseudo octahedral coordination geometry around the copper center. Absorption and fluorescence experimental evidence show good DNA binding propensity (in the order of 10(5) M(-1)) of the complexes. Thermal denaturation and circular dichroism (CD) analyses reveal minor structural changes of calf thymus (CT) DNA in presence of complexes and groove and/or surface binding of the complexes to CT-DNA. Kinetic DNA cleavage assay shows pseudo-first-order kinetic reaction between the complex and supercoiled (SC) DNA. In addition, mechanistic SC DNA cleavage results show higher DNA cleavage activity in presence of reducing agent, due to the presence of hydroxyl radicals. In vitro cytotoxicity assay of the complexes demonstrate that the complexes have low toxicity for different cancer cell lines and IC(50) values were between 37 and 156 µM.


Assuntos
Cobre/química , DNA/química , Desoxirribonucleases/química , Neoplasias/tratamento farmacológico , Compostos Organometálicos/síntese química , Linhagem Celular Tumoral , Cristalografia por Raios X , DNA/metabolismo , Desoxirribonucleases/metabolismo , Humanos , Concentração Inibidora 50 , Ligantes , Estrutura Molecular , Compostos Organometálicos/química
18.
Chem Phys Lipids ; 165(3): 320-30, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22414757

RESUMO

The protein kinase C (PKC) family of serine/threonine kinases is an attractive drug target for the treatment of cancer and other diseases. Natural product curcumin is known to interact with PKC isoforms through the C1 domain and modulate PKC activity. The reported results demonstrate that the symmetric curcumin molecule might act as two separate units during its recognition of C1 domains. To understand the importance of the two halves of curcumin in PKC binding and to develop effective PKC regulators, we synthesized a series of alkyl cinnamates (1-8), characterized absorption and fluorescence properties and measured binding affinities with the C1b subdomains of PKC isoforms. The binding parameters of the monomeric compounds and liposomes containing compounds confirmed their interaction with the C1b subdomains of PKCδ and PKCθ. The molecular docking analysis with PKCδ and PKCθ C1b subdomains revealed that the alkyl cinnamates form hydrogen bond with the backbone of the protein at the same binding site as that of diacylglycerol and phorbol esters. The results show that the alkyl cinnamates bind to the activator binding site of PKCs and both methoxy and hydroxyl groups play important roles in the binding process.


Assuntos
Cinamatos/química , Cinamatos/farmacologia , Proteína Quinase C/química , Proteína Quinase C/metabolismo , Alquilação , Animais , Sítios de Ligação , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cinamatos/síntese química , Cinamatos/metabolismo , Diglicerídeos/metabolismo , Desenho de Fármacos , Ativação Enzimática/efeitos dos fármacos , Polarização de Fluorescência , Isoenzimas/química , Isoenzimas/metabolismo , Ligantes , Lipossomos/metabolismo , Modelos Moleculares , Ésteres de Forbol/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico/efeitos dos fármacos , Solubilidade
19.
Mol Biosyst ; 8(4): 1275-85, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22301880

RESUMO

The protein kinase C (PKC) family of serine/threonine kinases is an attractive drug target for the treatment of cancer and other diseases. Diacylglycerol (DAG), phorbol esters and others act as ligands for the C1 domain of PKC isoforms. Inspection of the crystal structure of the PKCδ C1b subdomain in complex with phorbol-13-O-acetate shows that one carbonyl group and two hydroxyl groups play pivotal roles in recognition of the C1 domain. To understand the importance of two hydroxyl groups of phorbol esters in PKC binding and to develop effective PKC activators, we synthesized DAG like diacyltetrols (DATs) and studied binding affinities with C1b subdomains of PKCδ and PKCθ. DATs, with the stereochemistry of natural DAGs at the sn-2 position, were synthesized from (+)-diethyl L-tartrate in four to seven steps as single isomers. The calculated EC(50) values for the short and long chain DATs varied in the range of 3-6 µM. Furthermore, the fluorescence anisotropy values of the proteins were increased in the presence of DATs in a similar manner to that of DAGs. Molecular docking of DATs (1b-4b) with PKCδ C1b showed that the DATs form hydrogen bonds with the polar residues and backbone of the protein, at the same binding site, as that of DAG and phorbol esters. Our findings reveal that DATs represent an attractive group of C1 domain ligands that can be used as research tools or further structurally modified for potential drug development.


Assuntos
Lipídeos/química , Proteína Quinase C/química , Linhagem Celular , Diglicerídeos/metabolismo , Descoberta de Drogas/métodos , Fluorescência , Humanos , Ligação de Hidrogênio , Ligantes , Lipídeos/síntese química , Espectroscopia de Ressonância Magnética/métodos , Modelos Moleculares , Ésteres de Forbol/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA