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












Base de dados
Intervalo de ano de publicação
1.
Langmuir ; 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38967248

RESUMO

Colloidal crystals and their two-dimensional (2D) monolayers, which have been commonly applied in nanosphere lithography, have the potential to revolutionize many engineering disciplines; however, current production techniques are hampered by a restricted preparation area, laborious procedures, and the need for advanced equipment. We propose a self-assembly-driven, simple, and low-cost method to prepare 2D colloidal nanosphere monolayers with excellent repeatability across wide regions. The self-assembly capability of colloidal polystyrene (PS) nanospheres at the air/water interface was utilized to transfer the assembled monolayers onto a substrate. This innovative method combines the advantages of methods that permit deposition at the air/water interface, such as Langmuir and drop coating, in order to deliver defect-free, simple-to-install, and simple-to-apply deposition across vast regions. Using field emission scanning electron microscopy and atomic force microscopy, the resultant coatings were characterized. The size of the nanospheres was reduced using an oxygen plasma etch process in an inductively coupled plasma reactive ion etching system, and the reflectance properties of the substrates for various nanosphere sizes were investigated. By evaporation of a thin gold capping layer on the templates, their optical properties were compared using surface-enhanced Raman scattering spectroscopy. This work has the potential to expand the use of nanosphere lithography by offering a simple and reproducible method that eliminates the need for complicated experimental setups and reduces the amount of material required for monolayer coating, thus lowering the cost.

2.
Mil Med Res ; 11(1): 35, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38835066

RESUMO

Neuroendocrine neoplasms (NENs) are highly heterogeneous and potentially malignant tumors arising from secretory cells of the neuroendocrine system. Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) are the most common subtype of NENs. Historically, GEP-NENs have been regarded as infrequent and slow-growing malignancies; however, recent data have demonstrated that the worldwide prevalence and incidence of GEP-NENs have increased exponentially over the last three decades. In addition, an increasing number of studies have proven that GEP-NENs result in a limited life expectancy. These findings suggested that the natural biology of GEP-NENs is more aggressive than commonly assumed. Therefore, there is an urgent need for advanced researches focusing on the diagnosis and management of patients with GEP-NENs. In this review, we have summarized the limitations and recent advancements in our comprehension of the epidemiology, clinical presentations, pathology, molecular biology, diagnosis, and treatment of GEP-NETs to identify factors contributing to delays in diagnosis and timely treatment of these patients.


Assuntos
Tumores Neuroendócrinos , Neoplasias Pancreáticas , Neoplasias Gástricas , Humanos , Tumores Neuroendócrinos/terapia , Tumores Neuroendócrinos/epidemiologia , Tumores Neuroendócrinos/diagnóstico , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/epidemiologia , Neoplasias Pancreáticas/diagnóstico , Neoplasias Gástricas/epidemiologia , Neoplasias Gástricas/terapia , Neoplasias Gástricas/diagnóstico , Neoplasias Intestinais/terapia , Neoplasias Intestinais/epidemiologia , Neoplasias Intestinais/diagnóstico
3.
MedComm (2020) ; 5(7): e583, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38919334

RESUMO

Nonviral vectors, such as liposomes, offer potential for targeted gene delivery in cancer therapy. Liposomes, composed of phospholipid vesicles, have demonstrated efficacy as nanocarriers for genetic tools, addressing the limitations of off-targeting and degradation commonly associated with traditional gene therapy approaches. Due to their biocompatibility, stability, and tunable physicochemical properties, they offer potential in overcoming the challenges associated with gene therapy, such as low transfection efficiency and poor stability in biological fluids. Despite these advancements, there remains a gap in understanding the optimal utilization of nanoliposomes for enhanced gene delivery in cancer treatment. This review delves into the present state of nanoliposomes as carriers for genetic tools in cancer therapy, sheds light on their potential to safeguard genetic payloads and facilitate cell internalization alongside the evolution of smart nanocarriers for targeted delivery. The challenges linked to their biocompatibility and the factors that restrict their effectiveness in gene delivery are also discussed along with exploring the potential of nanoliposomes in cancer gene therapy strategies by analyzing recent advancements and offering future directions.

4.
ACS Omega ; 9(17): 19627-19636, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38708264

RESUMO

Metal-organic frameworks (MOFs) are utilized as nanocarriers to enhance the efficiency of chemotherapy drugs, including cisplatin, which exhibit limitations such as side effects and resistance mechanisms. To evaluate the role of MOFs, we employed a molecular dynamics simulation, which, unlike other experiments, is cost-effective, less dangerous, and provides accurate results. Furthermore, we conducted molecular docking simulations to understand the interaction between cisplatin and MOF, as well as their internal interactions and how they bind to each other. Cisplatin and MOF molecules were parametrized using the Avogadro software and x2top command in GROMACS 5.1.2 and optimized by CP2K software; the Charmm-GUI site parametrized the cell cancer membrane. Three molecular dynamics simulations were conducted in four stages at various pHs, followed by simulated umbrella sampling. The simulations analyzed the pH responsiveness, total energy, Gibbs free energy, gyration radius, radial distribution function (RDF), solvent accessible surface area, and nanoparticles' toxicity. Results demonstrated that a neutral pH level (7.4) has greater adsorption and interaction compared to acidic pH values (6.4 and 5.4) because it displays the highest total energy (-17.1 kJ/mol), the highest RDF value (6.66), and the shortest distance (0.51 nm). Furthermore, the combination of cisplatin and MOFs displayed increased penetration compared to that of their individual forms. This study highlights the suitability of MOFs as nanocarriers and identifies the optimal pH values for desirable outcomes. Thus, it provides future studies with appropriate data to conduct their experiments in assessing MOFs.

5.
Int J Biol Macromol ; 273(Pt 1): 132579, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38795895

RESUMO

Cancer phototherapy has been introduced as a new potential modality for tumor suppression. However, the efficacy of phototherapy has been limited due to a lack of targeted delivery of photosensitizers. Therefore, the application of biocompatible and multifunctional nanoparticles in phototherapy is appreciated. Chitosan (CS) as a cationic polymer and hyaluronic acid (HA) as a CD44-targeting agent are two widely utilized polymers in nanoparticle synthesis and functionalization. The current review focuses on the application of HA and CS nanostructures in cancer phototherapy. These nanocarriers can be used in phototherapy to induce hyperthermia and singlet oxygen generation for tumor ablation. CS and HA can be used for the synthesis of nanostructures, or they can functionalize other kinds of nanostructures used for phototherapy, such as gold nanorods. The HA and CS nanostructures can combine chemotherapy or immunotherapy with phototherapy to augment tumor suppression. Moreover, the CS nanostructures can be functionalized with HA for specific cancer phototherapy. The CS and HA nanostructures promote the cellular uptake of genes and photosensitizers to facilitate gene therapy and phototherapy. Such nanostructures specifically stimulate phototherapy at the tumor site, with particle toxic impacts on normal cells. Moreover, CS and HA nanostructures demonstrate high biocompatibility for further clinical applications.


Assuntos
Quitosana , Terapia Genética , Ácido Hialurônico , Imunoterapia , Neoplasias , Fototerapia , Ácido Hialurônico/química , Humanos , Quitosana/química , Neoplasias/terapia , Imunoterapia/métodos , Terapia Genética/métodos , Fototerapia/métodos , Animais , Nanoestruturas/química , Nanoestruturas/uso terapêutico , Terapia Combinada , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Nanopartículas/química
6.
J Nutr Biochem ; 130: 109647, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38604457

RESUMO

Macrophages are phagocytic cells with important physiological functions, including the digestion of cellular debris, foreign substances, and microbes, as well as tissue development and homeostasis. The tumor microenvironment (TME) shapes the aggressiveness of cancer, and the biological and cellular interactions in this complicated space can determine carcinogenesis. TME can determine the progression, biological behavior, and therapy resistance of human cancers. The macrophages are among the most abundant cells in the TME, and their functions and secretions can determine tumor progression. The education of macrophages to M2 polarization can accelerate cancer progression, and therefore, the re-education and reprogramming of these cells is promising. Moreover, macrophages can cause inflammation in aggravating pathological events, including cardiovascular diseases, diabetes, and neurological disorders. The natural products are pleiotropic and broad-spectrum functional compounds that have been deployed as ideal alternatives to conventional drugs in the treatment of cancer. The biological and cellular interactions in the TME can be regulated by natural products, and for this purpose, they enhance the M1 polarization of macrophages, and in addition to inhibiting proliferation and invasion, they impair the chemoresistance. Moreover, since macrophages and changes in the molecular pathways in these cells can cause inflammation, the natural products impair the pro-inflammatory function of macrophages to prevent the pathogenesis and progression of diseases. Even a reduction in macrophage-mediated inflammation can prevent organ fibrosis. Therefore, natural product-mediated macrophage targeting can alleviate both cancerous and non-cancerous diseases.


Assuntos
Produtos Biológicos , Inflamação , Macrófagos , Neoplasias , Microambiente Tumoral , Humanos , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Produtos Biológicos/farmacologia , Produtos Biológicos/uso terapêutico , Animais , Dieta
7.
ACS Appl Bio Mater ; 7(5): 2637-2659, 2024 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-38687958

RESUMO

Extensive research has been conducted on the application of nanoparticles in the treatment of cancer and infectious diseases. Due to their exceptional characteristics and flexible structure, they are classified as highly efficient drug delivery systems, ensuring both safety and targeted delivery. Nevertheless, nanoparticles still encounter obstacles, such as biological instability, absence of selectivity, recognition as unfamiliar elements, and quick elimination, which restrict their remedial capacity. To surmount these drawbacks, biomimetic nanotechnology has been developed that utilizes T cell and natural killer (NK) cell membrane-encased nanoparticles as sophisticated methods of administering drugs. These nanoparticles can extend the duration of drug circulation and avoid immune system clearance. During the membrane extraction and coating procedure, the surface proteins of immunological cells are transferred to the biomimetic nanoparticles. Such proteins present on the surface of cells confer several benefits to nanoparticles, including prolonged circulation, enhanced targeting, controlled release, specific cellular contact, and reduced in vivo toxicity. This review focuses on biomimetic nanosystems that are derived from the membranes of T cells and NK cells and their comprehensive extraction procedure, manufacture, and applications in cancer treatment and viral infections. Furthermore, potential applications, prospects, and existing challenges in their medical implementation are highlighted.


Assuntos
Membrana Celular , Células Matadoras Naturais , Nanopartículas , Neoplasias , Linfócitos T , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Nanopartículas/química , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Linfócitos T/imunologia , Linfócitos T/efeitos dos fármacos , Membrana Celular/química , Viroses/tratamento farmacológico , Antineoplásicos/química , Antineoplásicos/farmacologia , Animais , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Tamanho da Partícula , Teste de Materiais , Antivirais/química , Antivirais/farmacologia , Antivirais/uso terapêutico
8.
Drug Discov Today ; 29(7): 103981, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38614161

RESUMO

The combination of peptides and nanoparticles in cancer therapy has shown synergistic results. Nanoparticle functionalization with peptides can increase their targeting ability towards tumor cells. In some cases, the peptides can develop self-assembled nanoparticles, in combination with drugs, for targeted cancer therapy. The peptides can be loaded into nanoparticles and can be delivered by other drugs for synergistic cancer removal. Multifunctional types of peptide-based nanoparticles, including pH- and redox-sensitive classes, have been introduced in cancer therapy. The tumor microenvironment remolds, and the acceleration of immunotherapy and vaccines can be provided by peptide nanoparticles. Moreover, the bioimaging and labeling of cancers can be mediated by peptide nanoparticles. Therefore, peptides can functionalize nanoparticles in targeted cancer therapy.


Assuntos
Nanopartículas , Neoplasias , Peptídeos , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Peptídeos/química , Peptídeos/administração & dosagem , Peptídeos/uso terapêutico , Animais , Microambiente Tumoral , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Imunoterapia/métodos
9.
Cancer Lett ; 591: 216867, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38593919

RESUMO

Autophagy, a self-digestion mechanism, has emerged as a promising target in the realm of cancer therapy, particularly in bladder cancer (BCa), a urological malignancy characterized by dysregulated biological processes contributing to its progression. This highly conserved catabolic mechanism exhibits aberrant activation in pathological events, prominently featured in human cancers. The nuanced role of autophagy in cancer has been unveiled as a double-edged sword, capable of functioning as both a pro-survival and pro-death mechanism in a context-dependent manner. In BCa, dysregulation of autophagy intertwines with cell death mechanisms, wherein pro-survival autophagy impedes apoptosis and ferroptosis, while pro-death autophagy diminishes tumor cell survival. The impact of autophagy on BCa progression is multifaceted, influencing metastasis rates and engaging with the epithelial-mesenchymal transition (EMT) mechanism. Pharmacological modulation of autophagy emerges as a viable strategy to impede BCa progression and augment cell death. Notably, the introduction of nanoparticles for targeted autophagy regulation holds promise as an innovative approach in BCa suppression. This review underscores the intricate interplay of autophagy with cell death pathways and its therapeutic implications in the nuanced landscape of bladder cancer.


Assuntos
Autofagia , Neoplasias da Bexiga Urinária , Humanos , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/metabolismo , Autofagia/efeitos dos fármacos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Nanopartículas , Apoptose/efeitos dos fármacos , Animais , Ferroptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos
10.
Cancer Lett ; 591: 216857, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38583648

RESUMO

The considerable death rates and lack of symptoms in early stages of gastric cancer (GC) make it a major health problem worldwide. One of the most prominent risk factors is infection with Helicobacter pylori. Many biological processes, including those linked with cell death, are disrupted in GC. The cellular "self-digestion" mechanism necessary for regular balance maintenance, autophagy, is at the center of this disturbance. Misregulation of autophagy, however, plays a role in the development of GC. In this review, we will examine how autophagy interacts with other cell death processes, such as apoptosis and ferroptosis, and how it affects the progression of GC. In addition to wonderful its role in the epithelial-mesenchymal transition, it is engaged in GC metastasis. The role of autophagy in GC in promoting drug resistance stands out. There is growing interest in modulating autophagy for GC treatment, with research focusing on natural compounds, small-molecule inhibitors, and nanoparticles. These approaches could lead to breakthroughs in GC therapy, offering new hope in the fight against this challenging disease.


Assuntos
Autofagia , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal , Neoplasias Gástricas , Neoplasias Gástricas/patologia , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo , Humanos , Autofagia/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Ferroptose/efeitos dos fármacos , Animais , Helicobacter pylori
11.
Cancer Lett ; 591: 216860, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38583650

RESUMO

Cancer is the result of genetic abnormalities that cause normal cells to grow into neoplastic cells. Cancer is characterized by several distinct features, such as uncontrolled cell growth, extensive spreading to other parts of the body, and the ability to resist treatment. The scientists have stressed the development of nanostructures as novel therapeutic options in suppressing cancer, in response to the emergence of resistance to standard medicines. One of the specific mechanisms with dysregulation during cancer is autophagy. Nanomaterials have the ability to specifically carry medications and genes, and they can also enhance the responsiveness of tumor cells to standard therapy while promoting drug sensitivity. The primary mechanism in this process relies on autophagosomes and their fusion with lysosomes to break down the components of the cytoplasm. While autophagy was initially described as a form of cellular demise, it has been demonstrated to play a crucial role in controlling metastasis, proliferation, and treatment resistance in human malignancies. The pharmacokinetic profile of autophagy modulators is poor, despite their development for use in cancer therapy. Consequently, nanoparticles have been developed for the purpose of delivering medications and autophagy modulators selectively and specifically to the cancer process. Furthermore, several categories of nanoparticles have demonstrated the ability to regulate autophagy, which plays a crucial role in defining the biological characteristics and response to therapy of tumor cells.


Assuntos
Autofagia , Nanoestruturas , Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neoplasias/genética , Neoplasias/metabolismo , Autofagia/efeitos dos fármacos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Nanopartículas , Resistencia a Medicamentos Antineoplásicos , Animais
12.
ACS Appl Mater Interfaces ; 16(13): 15718-15729, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38506616

RESUMO

Surgical removal of tumor tissue remains the primary clinical approach for addressing breast cancer; however, complete tumor excision is challenging, and the remaining tumor cells can lead to tumor recurrence and metastasis over time, which substantially deteriorates the life quality of the patients. With the aim to improve local cancer radiotherapy, this work reports the fabrication of alginate (Alg) scaffolds containing bovine serum albumin (BSA)-coated bismuth sulfide (Bi2S3@BSA) nanoradiosensitizers using three-dimensional (3D) printing. Under single-dose X-ray irradiation in vitro, Alg-Bi2S3@BSA scaffolds significantly increase the formation of reactive oxygen species, enhance the inhibition of breast cancer cells, and suppress their colony formation capacity. In addition, scaffolds implanted under tumor tissue in murine model show high therapeutic efficacy by reducing the tumor volume growth rate under single-dose X-ray irradiation, while histological observation of main organs reveals no cytotoxicity or side effects. 3D-printed Alg-Bi2S3@BSA scaffolds produced with biocompatible and biodegradable materials may potentially lower the recurrence and metastasis rates in breast cancer patients by inhibiting residual tumor cells following postsurgery as well as exhibit anticancer properties in other solid tumors.


Assuntos
Bismuto , Neoplasias da Mama , Nanopartículas , Sulfetos , Humanos , Animais , Camundongos , Feminino , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/radioterapia , Alginatos , Alicerces Teciduais , Impressão Tridimensional , Engenharia Tecidual
13.
Biomed Pharmacother ; 173: 116465, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38507955

RESUMO

This study introduces an innovative co-delivery approach using the MCM-co-polymerized nanosystem, integrating chitosan and polyethylene glycol, and targeted by the MUC-1 aptamer (MCM@CS@PEG-APT). This system enables simultaneous delivery of the GFP plasmid and doxorubicin (DOX). The synthesis of the nanosystem was thoroughly characterized at each step, including FTIR, XRD, BET, DLS, FE-SEM, and HRTEM analyses. The impact of individual polymers (chitosan and PEG) on payload retardation was compared to the co-polymerized MCM@CS@PEG conjugation. Furthermore, the DOX release mechanism was investigated using various kinetic models. The nanosystem's potential for delivering GFP plasmid and DOX separately and simultaneously was assessed through fluorescence microscopy and flow cytometry. The co-polymerized nanosystem exhibited superior payload entrapment (1:100 ratio of Plasmid:NPs) compared to separately polymer-coated counterparts (1:640 ratio of Plasmid:NPs). Besides, the presence of pH-sensitive chitosan creates a smart nanosystem for efficient DOX and GFP plasmid delivery into tumor cells, along with a Higuchi model pattern for drug release. Toxicity assessments against breast tumor cells also indicated reduced off-target effects compared to pure DOX, introducing it as a promising candidate for targeted cancer therapy. Cellular uptake findings demonstrated the nanosystem's ability to deliver GFP plasmid and DOX separately into MCF-7 cells, with rates of 32% and 98%, respectively. Flow cytometry results confirmed efficient co-delivery, with 42.7% of cells showing the presence of both GFP-plasmid and DOX, while 52.2% exclusively contained DOX. Overall, our study explores the co-delivery potential of the MCM@CS@PEG-APT nanosystem in breast cancer therapy. This system's ability to co-deliver multiple agents preciselyopens new avenues for targeted therapeutic strategies.


Assuntos
Neoplasias da Mama , Quitosana , Nanopartículas , Humanos , Feminino , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Polimerização , Doxorrubicina/farmacologia , Oligonucleotídeos , Plasmídeos , DNA , Sistemas de Liberação de Medicamentos/métodos , Portadores de Fármacos
14.
Small ; 20(26): e2308479, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38385813

RESUMO

Microneedles (MNs) have maintained their popularity in therapeutic and diagnostic medical applications throughout the past decade. MNs are originally designed to gently puncture the stratum corneum layer of the skin and have lately evolved into intelligent devices with functions including bodily fluid extraction, biosensing, and drug administration. MNs offer limited invasiveness, ease of application, and minimal discomfort. Initially manufactured solely from metals, MNs are now available in polymer-based varieties. MNs can be used to create systems that deliver drugs and chemicals uniformly, collect bodily fluids, and are stimulus-sensitive. Although these advancements are favorable in terms of biocompatibility and production costs, they are insufficient for the therapeutic use of MNs. This is the first comprehensive review that discusses individual MN functions toward the evolution and development of smart and multifunctional MNs for a variety of novel and impactful future applications. The study examines fabrication techniques, application purposes, and experimental details of MN constructs that perform multiple functions concurrently, including sensing, drug-molecule release, sampling, and remote communication capabilities. It is highly likely that in the near future, MN-based smart devices will be a useful and important component of standard medical practice for different applications.


Assuntos
Agulhas , Humanos , Sistemas de Liberação de Medicamentos , Animais , Nanomedicina Teranóstica , Microinjeções/instrumentação , Microinjeções/métodos
15.
Cancer Metastasis Rev ; 43(1): 457-479, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38227149

RESUMO

Epithelial-mesenchymal transition (EMT) is a complicated molecular process that governs cellular shape and function changes throughout tissue development and embryogenesis. In addition, EMT contributes to the development and spread of tumors. Expanding and degrading the surrounding microenvironment, cells undergoing EMT move away from the main location. On the basis of the expression of fibroblast-specific protein-1 (FSP1), fibroblast growth factor (FGF), collagen, and smooth muscle actin (-SMA), the mesenchymal phenotype exhibited in fibroblasts is crucial for promoting EMT. While EMT is not entirely reliant on its regulators like ZEB1/2, Twist, and Snail proteins, investigation of upstream signaling (like EGF, TGF-ß, Wnt) is required to get a more thorough understanding of tumor EMT. Throughout numerous cancers, connections between tumor epithelial and fibroblast cells that influence tumor growth have been found. The significance of cellular crosstalk stems from the fact that these events affect therapeutic response and disease prognosis. This study examines how classical EMT signals emanating from various cancer cells interfere to tumor metastasis, treatment resistance, and tumor recurrence.


Assuntos
Transição Epitelial-Mesenquimal , Neoplasias , Humanos , Transição Epitelial-Mesenquimal/fisiologia , Neoplasias/metabolismo , Transdução de Sinais , Fenótipo , Resistência a Medicamentos , Linhagem Celular Tumoral , Microambiente Tumoral
16.
Transl Oncol ; 39: 101838, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38016356

RESUMO

As a clinically approved treatment strategy, chemotherapy-mediated tumor suppression has been compromised, and in spite of introducing various kinds of anticancer drugs, cancer eradication with chemotherapy is still impossible. Chemotherapy drugs have been beneficial in improving the prognosis of cancer patients, but after resistance emerged, their potential disappeared. Oxaliplatin (OXA) efficacy in tumor suppression has been compromised by resistance. Due to the dysregulation of pathways and mechanisms in OXA resistance, it is suggested to develop novel strategies for overcoming drug resistance. The targeted delivery of OXA by nanostructures is described here. The targeted delivery of OXA in cancer can be mediated by polymeric, metal, lipid and carbon nanostructures. The advantageous of these nanocarriers is that they enhance the accumulation of OXA in tumor and promote its cytotoxicity. Moreover, (nano)platforms mediate the co-delivery of OXA with drugs and genes in synergistic cancer therapy, overcoming OXA resistance and improving insights in cancer patient treatment in the future. Moreover, smart nanostructures, including pH-, redox-, light-, and thermo-sensitive nanostructures, have been designed for OXA delivery and cancer therapy. The application of nanoparticle-mediated phototherapy can increase OXA's potential in cancer suppression. All of these subjects and their clinical implications are discussed in the current review.

17.
Sci Rep ; 13(1): 15131, 2023 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-37704633

RESUMO

To solve the traditional radiotherapy obstacles, and also to enhance the radiation therapy efficacy various radiosensitizers have been developed. Radiosensitizers are promising agents that under X-ray irradiation enhance injury to tumor tissue by accelerating DNA damage. In this report, silver-silver sulfide nanoparticles (Ag-Ag2S NPs) were synthesized via a facile, one-pot and environmentally friendly biomineralization method. Ag-Ag2S was coated with bovine serum albumin (BSA) in situ and applied as an X-ray sensitizer to enhance the efficiency of radiotherapy. Also, folic acid (FA) was conjugated to Ag-Ag2S@BSA to impart active targeting capability to the final formulation (Ag-Ag2S@BSA-FA). Prepared NPs were characterized by transmission electron microscopes (TEM), scanning electron microscope (SEM), dynamic light scattering (DLS), ultraviolet-visible spectroscopy (UV-Vis), X-ray diffraction analysis (XRD), and X-ray photoelectron spectroscopy (XPS) techniques. Results show that most of the NPs have well-defined uniform Janus structures. The biocompatibility of the NPs was then evaluated both in vitro and in vivo. A series of in vitro assays were performed on 4T1 cancer cells to evaluate the therapeutic efficacy of the designed NPs. In addition, the radio-enhancing ability of the NPs was tested on the 4T1 breast cancer murine model. MTT, live and dead cell staining, apoptosis, ROS generation, and clonogenic in vitro assays demonstrated the efficacy of NPs as radiosensitizers in radiotherapy. In vivo results as well as H&E staining tumor tissues confirmed tumor destruction in the group that received Ag-Ag2S@BSA-FA NPs and exposed to X-ray. The results showed that prepared tumor-targeted Ag-Ag2S@BSA-FA NPs could be potential candidates as radiosensitizers for enhanced radiotherapy.


Assuntos
Neoplasias , Radioterapia (Especialidade) , Radiossensibilizantes , Animais , Camundongos , Prata/farmacologia , Biomineralização , Radiossensibilizantes/farmacologia , Projetos de Pesquisa , Ácido Fólico
18.
Talanta ; 265: 124846, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37379752

RESUMO

Doxorubicin (DOX) is a highly effective anticancer drug with a narrow therapeutic window; thus, sensitive and timely detection of DOX is crucial. Using electrodeposition of silver nanoparticles (AgNPs) and electropolymerization of alginate (Alg) layers on the surface of a glassy carbon electrode, a novel electrochemical probe was constructed (GCE). The fabricated AgNPs/poly-Alg-modified GCE probe was utilized for the quantification of DOX in unprocessed human plasma samples. For the electrodeposition of AgNPs and electropolymerization of alginate (Alg) layers on the surface of GCE, cyclic voltammetry (CV) was used in the potential ranges of -2.0 to 2.0 V and -0.6 to 0.2 V, respectively. The electrochemical activity of DOX exhibited two oxidation processes at the optimum pH value of 5.5 on the surface of the modified GCE. The DPV spectra of poly(Alg)/AgNPs modified GCE probe toward consecutive concentrations of DOX in plasma samples demonstrated wide dynamic ranges of 15 ng/mL-0.1 µg/mL and 0.1-5.0 µg/mL, with a low limit of quantification (LLOQ) of 15 ng/mL. The validation results indicated that the fabricated electrochemical probe might serve as a highly sensitive and selective assay for the quantification of DOX in patient samples. As an outstanding feature, the developed probe could detect DOX in unprocessed plasma samples and cell lysates without the requirement for pretreatment.


Assuntos
Incrustação Biológica , Nanopartículas Metálicas , Humanos , Carbono , Doxorrubicina/análise , Prata , Incrustação Biológica/prevenção & controle , Eletrodos , Alginatos , Técnicas Eletroquímicas/métodos , Limite de Detecção
19.
ACS Biomater Sci Eng ; 9(7): 4138-4148, 2023 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-37311018

RESUMO

Polyethylene glycol (PEG) was utilized to functionalize the surface of zinc ferrite nanoparticles (NPs) synthesized by the hydrothermal process in order to prevent aggregation and improve the biocompatibility of the NPs for the proposed magnetic resonance imaging (MRI) agent. Various spectroscopy techniques were used to examine the NPs' structure, size, morphology, and magnetic properties. The NPs had a cubic spinel structure with an average size of 8 nm. The formations of the spinel ferrite and the PEG coating band at the ranges of 300-600 and 800-2000 cm-1, respectively, were validated by Fourier-transform infrared spectroscopy. The NPs were spherical in shape, and energy-dispersive X-ray spectroscopy with mapping confirmed the presence of zinc, iron, and oxygen in the samples. The results of high-resolution transmission electron microscopy revealed an average size of 14 nm and increased stability after PEG coating. The decrease in zeta potential from -24.5 to -36.5 mV confirmed the PEG coating on the surface of the NPs. A high saturation magnetization of ∼50 emu/g, measured by vibration sample magnetometer, indicated the magnetic potential of NPs for biomedical applications. An MTT assay was used to examine the cytotoxicity and viability of human normal skin cells (HSF 1184) exposed to zinc ferrite and PEG@Zn ferrite NPs at various concentrations. After 24 h of treatment, negligible cytotoxicity of PEG-coated NPs was observed at high concentrations. Magnetic resonance imaging (MRI) suggested that PEG@Zn ferrite NPs are a unique and perfectly suited contrast agent for T2-weighted MRI and can successfully enhance the image contrast.


Assuntos
Meios de Contraste , Nanopartículas , Humanos , Meios de Contraste/farmacologia , Meios de Contraste/química , Zinco , Polietilenoglicóis/química , Imageamento por Ressonância Magnética/métodos , Nanopartículas/química
20.
ACS Appl Bio Mater ; 6(6): 2266-2276, 2023 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-37273221

RESUMO

Fruits are safe, toxin-free, and biomolecule-rich raw materials that may be utilized to decrease metal ions and stabilize nanoparticles. Here, we demonstrate the green synthesis of magnetite nanoparticles which were first capped with a layer of silica, followed by the decoration of silver nanoparticles, termed Ag@SiO2@Fe3O4, by using lemon fruit extract as the reducing agent in a size range of ∼90 nm. The effect of the green stabilizer on the characteristics of nanoparticles was examined via different spectroscopy techniques, and the elemental composition of the multilayer-coated structures was verified. The saturation magnetization of bare Fe3O4 nanoparticles at room temperature was recorded as 78.5 emu/g, whereas it decreased to 56.4 and 43.8 emu/g for silica coating and subsequent decoration with silver nanoparticles. All nanoparticles displayed superparamagnetic behavior with almost zero coercivity. While magnetization decreased with further coating processes, the specific surface area increased with silica coating from 67 to 180 m2 g-1 and decreased after the addition of silver and reached 98 m2 g-1, which can be explained by the organization of silver nanoparticles in an island-like model. Zeta potential values also decreased from -18 to -34 mV with coating, indicating an enhanced stabilization effect of the addition of silica and silver. The antibacterial tests against Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus) revealed that the bare Fe3O4 and SiO2@Fe3O4 did not show sufficient effect, while Ag@SiO2@Fe3O4, even at low concentrations (≤ 200 µg/mL), displayed high antibacterial activity due to the existence of silver atoms on the surface of nanoparticles. Furthermore, the in vitro cytotoxicity assay revealed that Ag@SiO2@Fe3O4 nanoparticles were not toxic to HSF-1184 cells at 200 µg/mL concentration. Antibacterial activity during consecutive magnetic separation and recycling steps was also investigated, and nanoparticles offered a high antibacterial effect for more than 10 cycles of recycling, making them potentially useful in biomedical fields.


Assuntos
Nanopartículas de Magnetita , Staphylococcus aureus , Prata/farmacologia , Prata/química , Dióxido de Silício/farmacologia , Dióxido de Silício/química , Nanopartículas de Magnetita/química , Escherichia coli , Antibacterianos/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...