Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 205
Filtrar
1.
ACS Appl Bio Mater ; 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39287553

RESUMEN

The intricate healing mechanism of chronic wounds and their multitude of healing-related obstacles, such as infections, compromised cellular processes, and impediments to the healing process, pose a significant healthcare problem. Exploration of metal oxide nanoparticles, such as yttrium oxide (Y2O3) nanoparticles, can lead to innovative discoveries in the field of chronic wound healing by offering cues that promote cell proliferation in the scaffolds. To achieve this, Y2O3 nanoparticles were synthesized and incorporated within poly(vinyl alcohol) (PVA) nanofibrous scaffolds. Moreover, lysine was infused in the nanofibrous scaffolds to tune its cell adhesion and antimicrobial property. The structure and morphology of the synthesized nanofibers were confirmed through various physicochemical characterizations. Notably, all the fabricated scaffolds have remarkably tuned WVTR values within the range of 2000-2500 g/m2/day, favorable for removing the wound exudate, which facilitate the healing process. The scaffolds exhibited substantial antimicrobial property of approximately 68% and 72.2% against both E. coli and S. aureus at optimized Y2O3 loading. They further prevented the formation of biofilm by 68.6% for S. aureus and 51.2% for P. aeruginosa, suggesting the inhibition of recurrent wound infection. The scaffolds illustrated good blood biocompatibility, cytocompatibility, and cell adhesion capabilities. In vitro ROS inhibition study also corroborated the antioxidant property of the scaffold. Similarly, the wound scratching experiment showed high proliferative capability of a yttria-loaded PVA/lysine (S3) sample through the development of an extracellular matrix support. Molecular insight of wound healing was also validated through flow cytometry analysis and immunocytochemistry imaging studies. The findings revealed increased collagen I (Col-I) expression of approximately 19.48% in cultured fibrocytes. The findings are validated from immunocytochemistry imaging. In summary, the results furnish a captivating paradigm for the use of these scaffolds as a therapeutic biomaterial and to foster their potential efficacy toward wound care management.

2.
Biochim Biophys Acta Rev Cancer ; 1879(5): 189168, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39121913

RESUMEN

Autophagy is a normal physiological process that aids the recycling of cellular nutrients, assisting the cells to cope with stressed conditions. However, autophagy's effect on cancer, including glioma, is uncertain and involves complicated molecular mechanisms. Several contradictory reports indicate that autophagy may promote or suppress glioma growth and progression. Autophagy inhibitors potentiate the efficacy of chemotherapy or radiation therapy in glioma. Numerous compounds stimulate autophagy to cause glioma cell death. Autophagy is also involved in the therapeutic resistance of glioma. This review article aims to detangle the complicated molecular mechanism of autophagy to provide a better perception of the two-sided role of autophagy in glioma and its therapeutic implications. The protein and epigenetic modulators of the cytoprotective and cytotoxic role of autophagy are described in this article. Moreover, several signaling pathways are associated with autophagy and its effects on glioma. We have reviewed the molecular pathways and highlighted the signaling axis involved in cytoprotective and cytotoxic autophagy. Additionally, this article discusses the role of autophagy in therapeutic resistance, including glioma stem cell maintenance and tumor microenvironment regulation. It also summarizes several investigations on the anti-glioma effects of autophagy modulators to understand the associated mechanisms and provide insights regarding its therapeutic implications.


Asunto(s)
Autofagia , Glioma , Transducción de Señal , Microambiente Tumoral , Humanos , Glioma/patología , Glioma/tratamiento farmacológico , Glioma/metabolismo , Autofagia/efectos de los fármacos , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/tratamiento farmacológico , Animales , Resistencia a Antineoplásicos , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo
3.
ACS Omega ; 9(30): 33204-33223, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39100348

RESUMEN

Dynamic metal-coordinated adhesive and self-healable hydrogel materials have garnered significant attention in recent years due to their potential applications in various fields. These hydrogels can form reversible metal-ligand bonds, resulting in a network structure that can be easily broken and reformed, leading to self-healing capabilities. In addition, these hydrogels possess excellent mechanical strength and flexibility, making them suitable for strain-sensing applications. In this work, we have developed a mechanically robust, highly stretchable, self-healing, and adhesive hydrogel by incorporating Ca2+-dicarboxylate dynamic metal-ligand cross-links in combination with low density chemical cross-links into a poly(acrylamide-co-maleic acid) copolymer structure. Utilizing the reversible nature of the Ca2+-dicarboxylate bond, the hydrogel exhibited a tensile strength of up to ∼250 kPa and was able to stretch to 15-16 times its original length. The hydrogel exhibited a high fracture energy of ∼1500 J m-2, similar to that of cartilage. Furthermore, the hydrogel showed good recovery, fatigue resistance, and fast self-healing properties due to the reversible Ca2+-dicarboxylate cross-links. The presence of Ca2+ resulted in a highly conductive hydrogel, which was utilized to design a flexible resistive strain sensor. This hydrogel can strongly adhere to different substrates, making it advantageous for applications in flexible electronic devices. When adhered to human body parts, the hydrogel can efficiently detect limb movements. The hydrogel also exhibited excellent performance as a solid electrolyte for flexible supercapacitors, with a capacitance of ∼260 F/g at 0.5 A/g current density. Due to its antifreezing and antidehydration properties, this hydrogel retains its flexibility at subzero temperatures for an extended period. Additionally, the porous network and high water content of the hydrogel impart remarkable electromagnetic attenuation properties, with a value of ∼38 dB in the 14.5-20.5 GHz frequency range, which is higher than any other hydrogel without conducting fillers. Overall, the hydrogel reported in this study exhibits diverse applications as a strain sensor, solid electrolyte for flexible supercapacitors, and efficient material for electromagnetic attenuation. Its multifunctional properties make it a promising candidate for use in various fields as a state-of-the-art material.

4.
ACS Appl Bio Mater ; 7(9): 5810-5822, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39186444

RESUMEN

Amphiphilic self-indicating and responsive polymer-based prodrugs have generated much interest as potential stimuli-responsive intelligent drug delivery systems (DDS) due to their ability to selectively deliver drugs to the cancer cells and to monitor real-time cellular uptake of the drug by imaging technique(s). In this direction, we have synthesized a new pH-responsive N-vinyl-2-pyrrolidone and coumarin-based fluorescent self-indicating polymeric prodrug (SIPD), poly(NVP)-b-poly(FPA.DOX-r-FPA-r-CA). This block copolymer prodrug self-assembled into stable micellar nanoparticles under physiological conditions that reduced undesirable drug leakage to normal cells but resulted in the release of the anticancer drug doxorubicin (DOX) in cancer cells because of acidic pH-induced cleavage of imine bonds between DOX and the copolymer. While the polymer was found to be highly biocompatible with both normal (HEK-293) cells and cancer (MCF-7) cells even at high concentrations by MTT assay, the polymer prodrug nanoparticles showed toxicity even higher than that of free DOX in cancer cells. Phase contrast microscopy also depicted the cytotoxic effects of the nanoparticles on cancer cells. The coumarin units present in the polymer served as a fluorescence resonance energy transfer (FRET) pair with the covalently attached DOX molecules, which was established by steady-state and time-resolved fluorescence spectroscopy. Furthermore, confocal microscopy results confirmed the FRET phenomenon, as the fluorescence intensity of coumarin in the micellar nanoparticles remained quenched initially in MCF-7 cells but recovered with time as the DOX molecules were released and gradually shifted toward the targeted nucleus. All of these studies implied that the synthesized prodrug nanoparticles may provide another viable option for delivering chemotherapeutic drugs into cancer cells with a capability of real-time monitoring of drug release.


Asunto(s)
Doxorrubicina , Nanopartículas , Polímeros , Profármacos , Humanos , Profármacos/química , Profármacos/farmacología , Doxorrubicina/farmacología , Doxorrubicina/química , Concentración de Iones de Hidrógeno , Nanopartículas/química , Polímeros/química , Células MCF-7 , Supervivencia Celular/efectos de los fármacos , Tamaño de la Partícula , Ensayo de Materiales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Liberación de Fármacos , Células HEK293 , Sistemas de Liberación de Medicamentos , Estructura Molecular
5.
J Chem Inf Model ; 64(14): 5580-5589, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-38982947

RESUMEN

Targeted treatments for breast cancer that minimize harm to healthy cells are highly sought after. Our study explores the potentiality of riboflavin as a targeted anticancer compound that can be activated by light irradiation. Here, we integrated time-dependent density functional theory (TD-DFT) calculations and an in vitro study under visible light. The TD-DFT calculations revealed that the electronic charge transferred from the DNA base to riboflavin, with the most significant excitation peak occurring within the visible light range. Guided by these insights, an in vitro study was conducted on the breast cancer cell lines MCF-7 and MDA-MB-231. The results revealed substantial growth inhibition in these cell lines when exposed to riboflavin under visible light, with no such impact observed in the absence of light exposure. Interestingly, riboflavin exhibited no/minimal growth-inhibitory effects on the normal cell line L929, irrespective of light conditions. Moreover, through EtBr displacement (DNA-EtBr) and the TUNEL assay, it has been illustrated that, upon exposure to visible light, riboflavin can intercalate within DNA and induce DNA damage. In conclusion, under visible light conditions, riboflavin emerges as a promising candidate with a selective and effective potent anticancer agent against breast cancer while exerting a minimal influence on regular cellular activity.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Daño del ADN , Teoría Funcional de la Densidad , Luz , Riboflavina , Riboflavina/farmacología , Riboflavina/química , Humanos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Antineoplásicos/farmacología , Antineoplásicos/química , Línea Celular Tumoral , Femenino , Proliferación Celular/efectos de los fármacos , ADN , Modelos Moleculares
6.
Biochim Biophys Acta Rev Cancer ; 1879(5): 189158, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39032537

RESUMEN

Tumor microenvironment is formed by various cellular and non-cellular components which interact with one another and form a complex network of interactions. Some of these cellular components also attain a secretory phenotype and release growth factors, cytokines, chemokines etc. in the surroundings which are capable of inducing even greater number of signalling networks. All these interactions play a decisive role in determining the course of tumorigenesis. The treatment strategies against cancer also exert their impact on the local microenvironment. Such interactions and anticancer therapies have been found to induce more deleterious outcomes like immunosuppression and chemoresistance in the process of tumor progression. Hence, understanding the tumor microenvironment is crucial for dealing with cancer and chemoresistance. This review is an attempt to develop some understanding about the tumor microenvironment and different factors which modulate it, thereby contributing to tumorigenesis. Along with summarising the major components of tumor microenvironment and various interactions taking place between them, it also throws some light on how the existing and potential therapies exert their impact on these dynamics.


Asunto(s)
Neoplasias , Microambiente Tumoral , Humanos , Neoplasias/patología , Neoplasias/genética , Transducción de Señal , Animales , Resistencia a Antineoplásicos , Carcinogénesis/patología , Comunicación Celular
7.
Int J Pharm ; 662: 124490, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39032873

RESUMEN

A sodium alginate (Alg) based REDOX (reduction and oxidation)-responsive and fluorescent active microgel was prepared via water in oil (w/o) mini-emulsion polymerization technique. Here, we initially synthesized sodium alginate-based disulfide cross linked microgels and after that those microgels were tagged with rhodamine amine derivative (RhB-NH2) by ionic interaction to get the pH-responsive fluorescent property. Functionalized microgels were characterized using 1H NMR, FTIR, DLS, HRTEM, FESEM, UV-vis, and fluorescence spectroscopy analyses. Presence of the REDOX-responsive disulfide-containing crosslinkers in the microgels enhances the release of doxorubicin (DOX), an anti-cancer drug in the reducing environment of the cancer-cells (simulated). Existence of the rhodamine-amine derivative in the microgels triggers the pH-dependent fluorescence property by showing fluorescence emission at 560-580 nm at pH 5.5 (cancer cell pH). The cytotoxicity of the biopolymer based microgel was assessed over both cancerous HeLa (IC50 100 µg/mL) and non-cancerous MDCK (IC50 200 µg/mL) cells by MTT assay which showed the synthesized microgel is non-toxic whereas DOX-loaded microgels showed significant toxicity. FACS and cell uptake (in vitro) analyses were conducted to understand the cell apoptosis cycle and behavior of the cancer cells in presence of the DOX-loaded microgels. This pH-responsive fluorescent active alginate-based biomaterial could be a promising material for the anti-cancer drug delivery and other medical fields.


Asunto(s)
Alginatos , Doxorrubicina , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Microgeles , Oxidación-Reducción , Alginatos/química , Concentración de Iones de Hidrógeno , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacología , Doxorrubicina/química , Humanos , Células HeLa , Animales , Microgeles/química , Sistemas de Liberación de Medicamentos/métodos , Perros , Antineoplásicos/química , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Rodaminas/química , Supervivencia Celular/efectos de los fármacos , Portadores de Fármacos/química , Colorantes Fluorescentes/química
8.
J Med Chem ; 67(12): 10321-10335, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38836562

RESUMEN

Breast cancer remains a global health challenge, and innovative strategies are required to target estrogen receptor α (ERα), a key player in its development. This study investigates the potential of campesterol, a natural phytosterol, as an ERα inhibitor for breast cancer. Our approach integrates in silico, in vitro, and ex vivo experiments to assess the therapeutic potential of campesterol. In silico analyses highlight campesterol as a promising ERα ligand with favorable binding affinities and dynamic properties. Structural analysis reveals conformational changes in ERα upon campesterol binding. In vitro studies confirm the selective growth inhibition of campesterol against ERα-positive breast cancer cells. This study extends to ER+ breast cancer patient-derived organoids (PDOs), showing the effectiveness of campesterol in ERα-positive breast cancer PDOs. Importantly, it emphasizes the receptor-specific nature of campesterol, providing insights into its context-dependent action. In conclusion, campesterol displays potential as an ERα inhibitor, offering new avenues for ER+ breast cancer treatment.


Asunto(s)
Neoplasias de la Mama , Receptor alfa de Estrógeno , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/metabolismo , Humanos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Femenino , Descubrimiento de Drogas , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Fitosteroles/farmacología , Fitosteroles/química , Proliferación Celular/efectos de los fármacos , Línea Celular Tumoral , Relación Estructura-Actividad , Simulación del Acoplamiento Molecular , Ensayos de Selección de Medicamentos Antitumorales , Colesterol/análogos & derivados
9.
Biomacromolecules ; 25(8): 5181-5197, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-38943659

RESUMEN

Rapid proliferation and a faster rate of glycolysis in cancer cells often result in an elevated local temperature (40-43 °C) at the tumor site. Nanoparticles prepared from polymers with two lower critical solution temperatures (LCSTs) can be utilized to take advantage of this subtle temperature elevation to deliver anticancer drugs preferably to the cancer cells, thereby enhancing the overall therapeutic efficacy and reducing side effects. In this direction, we synthesized N-vinyl-2-pyrrolidone (NVP) and substituted NVP (sub-NVP: C2-NVP, C4-NVP)-based polymers with precisely controlled LCSTs by varying the ratio of NVP and sub-NVP. The first LCST (LCST1) was kept below 37 °C to promote self-assembly, drug loading, and structural stability in physiological conditions and the second LCST (LCST2) was in the range of 40-43 °C to ensure mild hyperthermia-induced drug release. Additionally, covalent attachment of tetraphenylethylene (TPE, AIEgen) resulted in aggregation-induced emission in thermoresponsive micellar nanoparticles in which TPE acted as a Förster Resonance Energy Transfer (FRET) pair with the loaded anticancer drug doxorubicin (DOX). Tracking of FRET-induced fluorescence recovery of TPE molecules was utilized to confirm the real-time thermoresponsive release of DOX from nanoparticles and eventual localization of TPE in the cytoplasm and DOX in the nucleus. In vitro cellular studies such as cytotoxicity, cellular uptake, and thermoresponsive drug release showed that the DOX-loaded polymeric nanoparticles were nontoxic to normal cells (HEK-293) but significantly more effective in cancer cells (MCF-7) at 40 °C. To our knowledge, this is the first report of preferential delivery of anticancer drugs only by exploiting the slightly elevated temperature of cancer cells.


Asunto(s)
Doxorrubicina , Liberación de Fármacos , Nanopartículas , Polímeros , Humanos , Nanopartículas/química , Doxorrubicina/farmacología , Doxorrubicina/química , Doxorrubicina/administración & dosificación , Polímeros/química , Pirrolidinonas/química , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/administración & dosificación , Micelas , Temperatura , Sistemas de Liberación de Medicamentos/métodos , Células MCF-7 , Portadores de Fármacos/química , Transferencia Resonante de Energía de Fluorescencia , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Estilbenos
10.
Biochim Biophys Acta Rev Cancer ; 1879(3): 189105, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38701938

RESUMEN

The present study explores the complex roles of High Mobility Group Box 1 (HMGB1) in the context of cancer development, emphasizing glioblastoma (GBM) and other central nervous system (CNS) cancers. HMGB1, primarily known for its involvement in inflammation and angiogenesis, emerges as a multifaceted player in the tumorigenesis of GBM. The overexpression of HMGB1 correlates with glioma malignancy, influencing key pathways like RAGE/MEK/ERK and RAGE/Rac1. Additionally, HMGB1 secretion is linked to the maintenance of glioma stem cells (GSCs) and contributes to the tumor microenvironment's (TME) vascular leakiness. Henceforth, our review discusses the bidirectional impact of HMGB1, acting as both a promoter of tumor progression and a mediator of anti-tumor immune responses. Notably, HMGB1 exhibits tumor-suppressive roles by inducing apoptosis, limiting cellular proliferation, and enhancing the sensitivity of GBM to therapeutic interventions. This dualistic nature of HMGB1 calls for a nuanced understanding of its implications in GBM pathogenesis, offering potential avenues for more effective and personalized treatment strategies. The findings underscore the need to explore HMGB1 as a prognostic marker, therapeutic target, and a promising tool for stimulating anti-tumor immunity in GBM.


Asunto(s)
Neoplasias del Sistema Nervioso Central , Glioblastoma , Proteína HMGB1 , Microambiente Tumoral , Humanos , Proteína HMGB1/metabolismo , Glioblastoma/patología , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/inmunología , Neoplasias del Sistema Nervioso Central/metabolismo , Neoplasias del Sistema Nervioso Central/patología , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Animales , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/metabolismo , Transducción de Señal , Proliferación Celular
11.
Biochim Biophys Acta Mol Cell Res ; 1871(6): 119752, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38776987

RESUMEN

Preserving a functional mitochondrial network is crucial for cellular well-being, considering the pivotal role of mitochondria in ensuring cellular survival, especially under stressful conditions. Mitophagy, the selective removal of damaged mitochondria through autophagy, plays a pivotal role in preserving cellular homeostasis by preventing the production of harmful reactive oxygen species from dysfunctional mitochondria. While the involvement of mitophagy in neurodegenerative diseases has been thoroughly investigated, it is becoming increasingly evident that mitophagy plays a significant role in cancer biology. Perturbations in mitophagy pathways lead to suboptimal mitochondrial quality control, catalyzing various aspects of carcinogenesis, including establishing metabolic plasticity, stemness, metabolic reconfiguration of cancer-associated fibroblasts, and immunomodulation. While mitophagy performs a delicate balancing act at the intersection of cell survival and cell death, mounting evidence indicates that, particularly in the context of stress responses induced by cancer therapy, it predominantly promotes cell survival. Here, we showcase an overview of the current understanding of the role of mitophagy in cancer biology and its potential as a target for cancer therapy. Gaining a more comprehensive insight into the interaction between cancer therapy and mitophagy has the potential to reveal novel targets and pathways, paving the way for enhanced treatment strategies for therapy-resistant tumors in the near future.


Asunto(s)
Resistencia a Antineoplásicos , Mitocondrias , Mitofagia , Neoplasias , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Mitocondrias/metabolismo , Mitocondrias/patología , Animales , Progresión de la Enfermedad , Carcinogénesis/metabolismo , Especies Reactivas de Oxígeno/metabolismo
12.
ACS Appl Mater Interfaces ; 16(17): 21486-21497, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38640485

RESUMEN

The combined effects of twisted intramolecular charge transfer (TICT) and aggregation-induced emission (AIE) phenomena have demonstrated a significant influence on excited-state chemistry. These combined TICT and AIE features have been extensively utilized to enhance photodynamic and photothermal therapy. Herein, we demonstrated the synergistic capabilities of TICT and AIE phenomena in the design of the photoremovable protecting group (PRPG), namely, NMe2-Napy-BF2. This innovative PRPG incorporates TICT and AIE characteristics, resulting in four remarkable properties: (i) red-shifted absorption wavelength, (ii) strong near-infrared (NIR) emission, (iii) viscosity-sensitive emission property, and (iv) accelerated photorelease rate. Inspired by these intriguing attributes, we developed a nanodrug delivery system (nano-DDS) using our PRPG for cancer treatment. In vitro studies showed that our nano-DDS manifested effective cellular internalization, specific staining of cancer cells, high-resolution confocal imaging of cancerous cells in the NIR region, and controlled release of the anticancer drug chlorambucil upon exposure to light, leading to cancer cell eradication. Most notably, our nano-DDS exhibited a substantially increased two-photon (TP) absorption cross section (435 GM), exhibiting its potential for in vivo applications. This development holds promise for significant advancements in cancer treatment strategies.


Asunto(s)
Naftiridinas , Fotones , Humanos , Naftiridinas/química , Naftiridinas/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Clorambucilo/química , Clorambucilo/farmacología , Fotoquimioterapia , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Nanopartículas/química
13.
Mol Pharm ; 21(4): 1563-1590, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38466810

RESUMEN

Understanding protein sequence and structure is essential for understanding protein-protein interactions (PPIs), which are essential for many biological processes and diseases. Targeting protein binding hot spots, which regulate signaling and growth, with rational drug design is promising. Rational drug design uses structural data and computational tools to study protein binding sites and protein interfaces to design inhibitors that can change these interactions, thereby potentially leading to therapeutic approaches. Artificial intelligence (AI), such as machine learning (ML) and deep learning (DL), has advanced drug discovery and design by providing computational resources and methods. Quantum chemistry is essential for drug reactivity, toxicology, drug screening, and quantitative structure-activity relationship (QSAR) properties. This review discusses the methodologies and challenges of identifying and characterizing hot spots and binding sites. It also explores the strategies and applications of artificial-intelligence-based rational drug design technologies that target proteins and protein-protein interaction (PPI) binding hot spots. It provides valuable insights for drug design with therapeutic implications. We have also demonstrated the pathological conditions of heat shock protein 27 (HSP27) and matrix metallopoproteinases (MMP2 and MMP9) and designed inhibitors of these proteins using the drug discovery paradigm in a case study on the discovery of drug molecules for cancer treatment. Additionally, the implications of benzothiazole derivatives for anticancer drug design and discovery are deliberated.


Asunto(s)
Inteligencia Artificial , Descubrimiento de Drogas , Descubrimiento de Drogas/métodos , Diseño de Fármacos , Aprendizaje Automático , Relación Estructura-Actividad Cuantitativa
14.
J Org Chem ; 89(6): 3747-3768, 2024 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-38394362

RESUMEN

In this study, we designed the 4'-C-acetamidomethyl-2'-O-methoxyethyl (4'-C-ACM-2'-O-MOE) uridine and thymidine modifications, aiming to test them into small interfering RNAs. Thermal melting studies revealed that incorporating a single 4'-C-ACM-2'-O-MOE modification in the DNA duplex reduced thermal stability. In contrast, an increase in thermal stability was observed when the modification was introduced in DNA:RNA hybrid and in siRNAs. Thermal destabilization in DNA duplex was attributed to unfavorable entropy, which was mainly compensated by the enthalpy factor to some extent. A single 4'-C-ACM-2'-O-MOE thymidine modification at the penultimate position of the 3'-end of dT20 oligonucleotides in the presence of 3'-specific exonucleases, snake venom phosphodiesterase (SVPD), demonstrated significant stability as compared to monomer modifications including 2'-O-Me, 2'-O-MOE, and 2'-F. In gene silencing studies, we found that the 4'-C-ACM-2'-O-MOE uridine or thymidine modifications at the 3'-overhang in the passenger strand in combination with two 2'-F modifications exhibited superior RNAi activity. The results suggest that the dual modification is well tolerated at the 3'-end of the passenger strand, which reflects better siRNA stability and silencing activity. Interestingly, 4'-C-ACM-2'-O-MOE-modified siRNAs showed considerable gene silencing even after 96 h posttransfection; it showed that our modification could induce prolonged gene silencing due to improved metabolic stability. Molecular modeling studies revealed that the introduction of the 4'-C-ACM-2'-O-MOE modification at the 3'-end of the siRNA guide strand helps to anchor the strand within the PAZ domain of the hAgo2 protein. The overall results indicate that the 4'-C-ACM-2'-O-MOE uridine and thymidine modifications are promising modifications to improve the stability, potency, and hAgo2 binding of siRNAs.


Asunto(s)
Ácidos Nucleicos , ARN Interferente Pequeño/química , ADN , Timidina , Uridina/química
15.
ACS Biomater Sci Eng ; 10(2): 1112-1127, 2024 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-38163852

RESUMEN

Measurement of pH in living cells is a great and decisive factor for providing an early and accurate diagnosis factor. Along with this, the multimodal transverse and longitudinal relaxivity enhancement potentiality over single modality within a single platform in the magnetic resonance imaging (MRI) field is a very challenging issue for diagnostic purposes in the biomedical field of application. Therefore, this work aims to design a versatile platform by fabricating a novel nanoprobe through holmium- and manganese-ion doping in carbon quantum dots (Ho-Mn-CQDs), which can show nearly neutral intracellular pH sensing and MRI imaging at the same time. These manufactured Ho-Mn-CQDs acted as excellent pH sensors in the near-neutral range (4.01-8.01) with the linearity between 6.01 and 8.01, which could be useful for the intracellular pH-sensing capability. An innumerable number of carboxyl and amino groups are present on the surface of the prepared nanoprobe, making it an excellent candidate for pH sensing through fluorescence intensity quenching phenomena. Cellular uptake and cell viability experiments were also executed to affirm the intracellular accepting ability of Ho-Mn-CQDs. Furthermore, with this pH-sensing quality, these Ho-Mn-CQDs are also capable of acting as T1-T2 dual modal imaging contrast agents in comparison with pristine Ho-doped and Mn-doped CQDs. The Ho-Mn-CQDs showed an increment of r1 and r2 relaxivity values simultaneously compared with only the negative contrast agent, holmium in holmium-doped CQDs, and the positive contrast agent, manganese in manganese-doped CQDs. The above-mentioned observations elucidate that its tiny size, excitation dependence of fluorescence behavior, low cytotoxicity, and dual modal contrast imaging capability make it an ideal candidate for pH monitoring in the near-neutral range and also as a dual modal MRI imaging contrast enhancement nanoprobe at the same time.


Asunto(s)
Medios de Contraste , Manganeso , Carbono , Holmio , Concentración de Iones de Hidrógeno
16.
Int J Biol Macromol ; 256(Pt 1): 128262, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37989431

RESUMEN

This study focused on GSK-3ß, a critical serine/threonine kinase with diverse cellular functions. However, there is limited understanding of the impact of non-synonymous single nucleotide polymorphisms (nsSNPs) on its structure and function. Through an exhaustive in-silico investigation 12 harmful nsSNPs were predicted from a pool of 172 acquired from the NCBI dbSNP database using 12 established tools that detects deleterious SNPs. Consistently, these nsSNPs were discovered in locations with high levels of conservation. Notably, the three harmful nsSNPs F67C, A83T, and T138I were situated in the active/binding site of GSK-3ß, which may affect the protein's capacity to bind to substrates and other proteins. Molecular dynamics simulations revealed that the F67C and T138I mutants had stable structures, indicating rigidness, whereas the A83T mutant was unstable. Analysis of secondary structures revealed different modifications in all mutant forms, which may affect the stability, functioning, and interactions of the protein. These mutations appear to alter the structural dynamics of GSK-3ß, which may have functional ramifications, such as the formation of novel secondary structures and variations in coil-to-helix transitions. In conclusion, this study illuminates the possible structural and functional ramifications of these GSK-3 nsSNPs, revealing how protein compactness, stiffness, and interactions may affect biological activities.


Asunto(s)
Glucógeno Sintasa Quinasa 3 , Polimorfismo de Nucleótido Simple , Glucógeno Sintasa Quinasa 3 beta/genética , Polimorfismo de Nucleótido Simple/genética , Glucógeno Sintasa Quinasa 3/genética , Simulación de Dinámica Molecular , Cicatrización de Heridas , Biología Computacional
17.
J Biomol Struct Dyn ; 42(5): 2270-2281, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37139547

RESUMEN

Glioblastoma, the most severe form of brain tumor and a leading cause of death within a year of diagnosis, is characterized by excessive protein synthesis and folding in the lumen of the endoplasmic reticulum (ER), leading to increased ER stress in the cells of GBM tissues. To mitigate this stress the cancer cells have intelligently adopted a plethora of response mechanisms and Unfolded Protein Response (UPR) is one of those. To bear with this exhaustive situation cells upregulate a strong protein degradation system in form of 26S proteasome and blocking of proteasomal gene synthesis may be a potential therapeutic action against GBM. Proteasomal gene synthesis is exclusively dependent on the transcription factor Nuclear respiratory factor 1 (NRF1) and its activating enzyme DNA damage inducible 1 homolog 2 (DDI2). Here in this study, we performed molecular docking against DDI2 with the 20 FDA-approved drugs and identified Alvimopan and Levocabastine as the top two compounds with the best binding score along with the standard drug Nelfinavir. MD simulation (100 ns) of these protein-ligand docked complexes reveals that the stability and compactness of Alvimopan are high in comparison with Nelfinavir. Our in-silico (Molecular docking and Molecular dynamics simulation) studies pointed out that Alvimopan may be repurposed as a DDI2 inhibitor and can be used as a potential anticancer agent for the treatment of brain tumors.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Antineoplásicos , Proteasas de Ácido Aspártico , Glioblastoma , Humanos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Reposicionamiento de Medicamentos , Glioblastoma/tratamiento farmacológico , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Nelfinavir/farmacología , Proteasas de Ácido Aspártico/antagonistas & inhibidores
18.
Int J Biol Macromol ; 253(Pt 7): 127453, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-37844820

RESUMEN

Brain tumors, particularly gliomas, remain difficult to treat due to their complex and dynamic microenvironment and high mortality rate. The presence of tumor-associated macrophages (TAMs) is considered one of the primary factors contributing to a poor prognosis in Glioma. Previous reports have linked elevated levels of Adenosine deaminase 2 (ADA2) with immunosuppression, tumor progression, and angiogenesis via MAPK, PDGFß signaling pathway in the glioma microenvironment. In contrast, Adenosine deaminase 1 (ADA1), another type of adenosine deaminase, plays a pivotal role in purine metabolism, which is essential for lymphocyte survival. Hence, selectively targeting ADA2 while preserving ADA1 activity could offer a viable approach for regulating macrophage polarization and enhancing the anti-tumor immune response. In pursuit of this objective, our study employed a computational approach, unveiling the remarkable attributes of Daidzin, characterized by its exceptional specificity, and binding affinity towards ADA2 while displaying minimal affinity towards ADA1. Furthermore, Define Secondary Structure of Proteins (DSSP) analysis revealed that Daidzin elicits conspicuous conformational alterations within the dimerization domain of the ADA2 receptor, which could have a crucial impact on its activity. However, the ADA1 structure remained unaltered. Our study offers the potential use of Daidzin as a specific therapeutic agent for modulating the tumor microenvironment and revolutionizing glioma management.


Asunto(s)
Adenosina Desaminasa , Glioma , Humanos , Adenosina Desaminasa/metabolismo , Flavonoides , Transducción de Señal , Glioma/tratamiento farmacológico , Inmunidad , Microambiente Tumoral
19.
Front Bioeng Biotechnol ; 11: 1213932, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37701494

RESUMEN

Targeted delivery of site-specific therapeutic agents is an effective strategy for osteoarthritis treatment. The lack of blood vessels in cartilage makes it difficult to deliver therapeutic agents like peptides to the defect area. Therefore, nucleus-targeting zwitterionic carbon nano-dots (CDs) have immense potential as a delivery vehicle for effective peptide delivery to the cytoplasm as well as nucleus. In the present study, nucleus-targeting zwitterionic CDs have been synthesized as delivery vehicle for peptides while also working as nano-agents towards optical monitoring of cartilage healing. The functional groups of zwitterion CDs were introduced by a single-step microwave assisted oxidation procedure followed by COL II peptide conjugation derived from Capra auricular cartilage through NHS/EDC coupling. The peptide-conjugated CDs (PCDs) allows cytoplasmic uptake within a short period of time (∼30 m) followed by translocation to nucleus after ∼24 h. Moreover, multicolor fluorescence of PCDs improves (blue, green, and read channel) its sensitivity as an optical code providing a compelling solution towards enhanced non-invasive tracking system with multifunctional properties. The PCDs-based delivery system developed in this study has exhibited superior ability to induce ex-vivo chondrogenic differentiation of ADMSCs as compared to bare CDs. For assessment of cartilage regeneration potential, pluronic F-127 based PCDs hydrogel was injected to rabbit auricular cartilage defects and potential healing was observed after 60 days. Therefore, the results confirm that PCDs could be an ideal alternate for multimodal therapeutic agents.

20.
ACS Appl Mater Interfaces ; 15(27): 32099-32109, 2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-37386863

RESUMEN

The regulation of the cell cycle has recently opened up a new research perspective for cancer treatment. So far, no effort has been made for temporal control of cell cycles using a photocleavable linker. Presented herein is the first report of regulation of disrupted cell cycles through the temporal release of a well-known cell cycle regulator α-lipoic acid (ALA), enabled by a newly designed NIR-active quinoxaline-based photoremovable protecting group (PRPG). The suitable quinoxaline-based photocage of ALA (tetraphenylethelene conjugated) has been formulated as fluorescent organic nanoparticles (FONs) and used effectively as a nano-DDS (drug delivery system) for better solubility and cellular internalization. Fascinatingly, the enhanced TP (two-photon) absorption cross section of the nano-DDS (503 GM) signifies its utility for biological applications. Using green light, we have successfully controlled the time span of cell cycles and cell growth of skin melanoma cell lines (B16F10) by the temporal release of ALA. Further, in silico studies and PDH activity assay supported the observed regulatory behavior of our nano-DDS with respect to photoirradiation. Overall, this approach expands the research path toward a futuristic photocontrolled toolbox for cell cycle regulation.


Asunto(s)
Nanopartículas , Profármacos , Ácido Tióctico , Sistema de Administración de Fármacos con Nanopartículas , Quinoxalinas/farmacología , Sistemas de Liberación de Medicamentos/métodos , Ciclo Celular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA