Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Biol Macromol ; 240: 124441, 2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-37060978

RESUMO

In the biological systems, exposure to nanoparticles (NPs) can cause complicated interactions with proteins, the formation of protein corona and structural changes to proteins. These changes depend not only on NP physicochemical properties, but also on the intrinsic stability of protein molecules. Although, the formation of protein corona on the surface of NPs and the underlying mechanisms have been fully explored in various studies, no comprehensive review has discussed the direct biochemical and biophysical interactions between NPs and blood proteins, particularly transferrin. In this review, we first discussed the interaction of NPs with proteins to comprehend the effects of physicochemical properties of NPs on protein structure. We then overviewed the transferrin structure and its direct interaction with NPs to explore transferrin stability and its iron ion (Fe3+) release behavior. Afterwards, we surveyed the various biological functions of transferrin, such as Fe3+ binding, receptor binding, antibacterial activity, growth, differentiation, and coagulation, followed by the application of transferrin-modified NPs in the development of drug delivery systems for cancer therapy. We believe that this study can provide useful insight into the design and development of bioconjugates containing NP-transferrin for potential biomedical applications.


Assuntos
Nanopartículas , Coroa de Proteína , Transferrina/química , Coroa de Proteína/química , Nanopartículas/química , Ferro/metabolismo , Ligação Proteica
2.
J Control Release ; 348: 127-147, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35660636

RESUMO

Nanoparticles (NPs) have been used in numerous applications as anticancer, antibacterial and antioxidant agents. Artificial engineering of protein interactions with NPs in biological systems is crucial to develop potential NPs for drug delivery and cancer nanotherapy. The protein corona (PC) on the NP surface, displays an interface between biomacromolecules and NPs, governing their pharmacokinetics and pharmacodynamics. Upon interaction of proteins with the NPs, their surface features are modified and they can easily be removed from the circulation by the mononuclear phagocytic system (MPS). PC properties heavily depend on the biological microenvironment and NP physicochemical parameters. Based on this context, we have surveyed different approaches that have been used for artificial engineering of the PC composition on NP surfaces. We discussed the effects of NP size, shape, surface modifications (PEGylation, self-peptide, other polymers), and protein pre-coating on the PC properties. Additionally, other factors including protein source and structure, intravenous injection and the subsequent shear flow, plasma protein gradients, temperature and local heat transfer, and washing media were considered in the context of their effects on the PC properties and overall target cellular effects. Moreover, the effects of NP-PC complexes on cancer cells based on cellular interactions, organization of intracellular PC (IPC), targeted drug delivery (TDD) and regulation of burst drug release profile of nanoplatforms, enhanced biocompatibility, and clinical applications were discussed followed by challenges and future perspective of the field. In conclusion, this paper can provide useful information to manipulate PC properties on the NP surface, thus trying to provide a literature survey to shorten their shipping from preclinical to clinical trials and to lay the basis for a personalized PC.


Assuntos
Nanopartículas , Neoplasias , Coroa de Proteína , Liberação Controlada de Fármacos , Humanos , Nanopartículas/química , Neoplasias/tratamento farmacológico , Polímeros/metabolismo , Coroa de Proteína/metabolismo , Proteínas/metabolismo , Microambiente Tumoral
3.
J Adv Res ; 33: 227-239, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34603792

RESUMO

Background: Because enzymes can control several metabolic pathways and regulate the production of free radicals, their simultaneous use with nanoplatforms showing protective and combinational properties is of great interest in the development of therapeutic nano-based platforms. However, enzyme immobilization on nanomaterials is not straightforward due to the toxic and unpredictable properties of nanoparticles in medical practice. Aim of review: In fact, because of the ability to load enzymes on nano-based supports and increase their renewability, scientific groups have been tempted to create potential therapeutic enzymes in this field. Therefore, this study not only pays attention to the therapeutic and diagnostic applications of diseases by enzyme-nanoparticle (NP) bio-conjugate (abbreviated as: ENB), but also considers the importance of nanoplatforms used based on their toxicity, ease of application and lack of significant adverse effects on loaded enzymes. In the following, based on the published reports, we explained that the immobilization of enzymes on polymers, inorganic metal oxide and hybrid compounds provide hopes for potential use of ENBs in medical activities. Then, the use of ENBs in bioassay activities such as paper-based or wearing biosensors and lab-on-chip/microfluidic biosensors were evaluated. Finally, this review addresses the current challenges and future perspective of ENBs in biomedical applications. Key scientific concepts of review: This literature may provide useful information regarding the application of ENBs in biosensing and therapeutic platforms.


Assuntos
Nanopartículas Metálicas , Nanoestruturas , Enzimas Imobilizadas , Nanopartículas Metálicas/toxicidade , Óxidos , Polímeros
4.
J Control Release ; 338: 341-357, 2021 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-34428480

RESUMO

Microneedle arrays have recently received much attention as cancer detection and treatment platforms, because invasive injections and detection of the biopsy are not needed, and drug metabolism by the liver, as well as adverse effects of systemic drug administration, are diminished. Microneedles have been used for diagnosis, vaccination, and in targeted drug delivery of breast cancer. In this review, we summarize the recent progress in diagnosis and targeted drug delivery for breast cancer treatment, using microneedle arrays to deliver active molecules through the skin. The results not only suggest that health and well-being of patients are improved, but also that microneedle arrays can deliver anticancer compounds in a relatively noninvasive manner, based on body weight, breast tumor size, and circulation time of the drug. Moreover, microneedles could allow simultaneous loading of multiple drugs and enable controlled release, thus effectively optimizing or preventing drug-drug interactions. This review is designed to encourage the use of microneedles for diagnosis and treatment of breast cancer, by describing general properties of microneedles, materials used for construction, mechanism of action, and principal benefits. Ongoing challenges and future perspectives for the application of microneedle array systems in breast cancer detection and treatment are highlighted.


Assuntos
Neoplasias da Mama , Administração Cutânea , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Feminino , Humanos , Microinjeções , Agulhas , Pele/metabolismo
5.
J Control Release ; 333: 91-106, 2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-33774120

RESUMO

The bioprinting technique with specialized tissue production allows the study of biological, physiological, and behavioral changes of cancerous and non-cancerous tissues in response to pharmacological compounds in personalized medicine. To this end, to evaluate the efficacy of anticancer drugs before entering the clinical setting, tissue engineered 3D scaffolds containing breast cancer and derived from the especially patient, similar to the original tissue architecture, can potentially be used. Despite recent advances in the manufacturing of 3D bioprinted breast cancer tissue (BCT), many studies still suffer from reproducibility primarily because of the uncertainty of the materials used in the scaffolds and lack of printing methods. In this review, we present an overview of the breast cancer environment to optimize personalized treatment by examining and identifying the physiological and biological factors that mimic BCT. We also surveyed the materials and techniques related to 3D bioprinting, i.e, 3D bioprinting systems, current strategies for fabrication of 3D bioprinting tissues, cell adhesion and migration in 3D bioprinted BCT, and 3D bioprinted breast cancer metastasis models. Finally, we emphasized on the prospective future applications of 3D bioprinted cancer models for rapid and accurate drug screening in breast cancer.


Assuntos
Bioimpressão , Neoplasias da Mama , Neoplasias da Mama/tratamento farmacológico , Feminino , Humanos , Impressão Tridimensional , Estudos Prospectivos , Reprodutibilidade dos Testes , Engenharia Tecidual , Alicerces Teciduais
6.
J Biomol Struct Dyn ; 39(10): 3771-3779, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-32397906

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative representative of a severe respiratory illness resulted in widespread human infections and deaths in nearly all of the countries since late 2019. There is no therapeutic FDA-approved drug against SARS-CoV-2 infection, although a combination of anti-viral drugs is directly being practiced in some countries. A broad-spectrum of antiviral agents are being currently evaluated in clinical trials, and in this review, we specifically focus on the application of Remdesivir (RVD) as a potential anti-viral compound against Middle East respiratory syndrome (MERS) -CoV, SARS-CoV and SARS-CoV-2. First, we overview the general information about SARS-CoV-2, followed by application of RDV as a nucleotide analogue which can potentially inhibits RNA-dependent RNA polymerase of COVs. Afterwards, we discussed the kinetics of SARS- or MERS-CoV proliferation in animal models which is significantly different compared to that in humans. Finally, some ongoing challenges and future perspective on the application of RDV either alone or in combination with other anti-viral agents against CoVs infection were surveyed to determine the efficiency of RDV in preclinical trials. As a result, this paper provides crucial evidence of the potency of RDV to prevent SARS-CoV-2 infections.Communicated by Ramaswamy H. Sarma.


Assuntos
Antivirais , Tratamento Farmacológico da COVID-19 , RNA Polimerase Dependente de RNA , Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Animais , Antivirais/farmacologia , Reposicionamento de Medicamentos , Humanos , RNA Polimerase Dependente de RNA/antagonistas & inibidores , SARS-CoV-2/efeitos dos fármacos
7.
J Biomol Struct Dyn ; 39(10): 3780-3786, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-32397951

RESUMO

Researchers have reported some useful information about the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) leading to CoV disease 2019 (COVID-19). Several studies have been performed in order to develop antiviral drugs, from which a few have been prescribed to patients. Also, several diagnostic tests have been designed to accelerate the process of identifying and treating COVID-19. It has been well-documented that the surface of host cells is covered by some receptors, known as angiotensin-converting enzyme 2 (ACE2), which mediates the binding and entry of CoV. After entering, the viral RNA interrupts the cell proliferation system to activate self-proliferation. However, having all the information about the outbreakof the SARS-COV-2, it is not still clear which factors determine the severity of lung and heart function impairment induced by COVID-19. A major step in exploring SARS-COV-2 pathogenesis is to determine the distribution of ACE2 in different tissues . In this review, the structure and origin of CoV, the role of ACE2 as a receptor of SARS-COV-2 on the surface of host cells, and the ACE2 distribution in different tissues with a focus on lung and cardiovascular system have been discussed. It was also revealed that acute and chronic cardiovascular diseases (CVDs) may result in the clinical severity of COVID-19. In conclusion, this review may provide useful information in developing some promising strategies to end up with a worldwide COVID-19 pandemic.Communicated by Ramaswamy H. Sarma.


Assuntos
Enzima de Conversão de Angiotensina 2/genética , COVID-19 , COVID-19/diagnóstico , Coração , Humanos , Pulmão , Pandemias , SARS-CoV-2 , Índice de Gravidade de Doença
8.
Mater Sci Eng C Mater Biol Appl ; 119: 111649, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33321685

RESUMO

The interaction of nanoparticles with protein and cells may provide important information regarding their biomedical implementations. Herein, after synthesis of tin oxide (SnO2) nanoparticles by hydrothermal method, their interaction with human serum albumin (HSA) was evaluated by multispectroscopic and molecular docking (MD) approaches. Furthermore, the selective antiproliferative impact of SnO2 nanoparticles against leukemia K562 cells was assessed by different cellular assays, whereas lymphocytes were used as control cells. TEM, DLS, zeta potential and XRD techniques showed that crystalline SnO2 nanoparticles have a size of less than 50 nm with a good colloidal stability. Fluorescence and CD spectroscopy analysis indicated that the HSA undergoes some slight conformational changes after interaction with SnO2 nanoparticles, whereas the secondary structure of HSA remains intact. Moreover, MD outcomes revealed that the charged residues of HSA preferentially bind to SnO2 nanoclusters in the binding pocket. Antiproliferative examinations displayed that SnO2 nanoparticles can selectively cause the mortality of K562 cells through induction of cell membrane leakage, activation of caspase-9, -8, -3, down regulation of Bcl-2 mRNA, the elevation of ROS level, S phase arrest, and apoptosis. In conclusion, this data may indicate that SnO2 nanoparticles can be used as promising particles to be integrated into therapeutic platforms.


Assuntos
Nanopartículas , Compostos de Estanho , Humanos , Células K562 , Simulação de Acoplamento Molecular
9.
Int J Nanomedicine ; 13: 6871-6884, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30498348

RESUMO

BACKGROUND: Implementations of nanoparticles have been receiving great interest in medicine and technology due to their unique characteristics. However, their toxic impacts on the biological system are not well explored. AIM: This study aims to investigate the influence of fabricated nano graphene oxide (NGO) sheets on the secondary and quaternary structural alterations of human hemoglobin (Hb) and cytotoxicity against lymphocyte cells. MATERIALS AND METHODS: Different spectroscopic methods, such as extrinsic and synchronous fluorescence spectroscopy and far circular dichroism (CD) spectroscopy, molecular docking investigation, cellular assays (trypan blue exclusion, cellular uptake, ROS, cell cycle, and apoptosis), and molecular assay (fold changes in anti/proapoptotic genes [B-cell lymphoma-2 {BCL2}/BAX] expression levels) were used in this study. RESULTS: Transmission electron microscopy, X-ray diffraction, Fourier transform infrared spectroscopy, and zeta potential investigations revealed the nano-sized nature of NGOs with good colloidal stability. Extrinsic fluorescence spectroscopy by using 8-anilinonaphthalene-1 -sulfonic acid and synchronous fluorescence spectroscopy showed that NGOs can unfold the quaternary structure of Hb in the vicinity of Tyr residues. The CD investigation demonstrated that the α-helicity of Hb experienced substantial alteration upon interaction with increasing concentrations of NGOs. The molecular docking study showed that NGOs interacted with polar residues of Hb. Cellular and molecular assays revealed that NGOs lead to ROS formation, cell cycle arrest, and apoptosis through the BAX and BCL2 pathway. CONCLUSION: These data reveal that NGOs can induce some protein structural changes and stimulate cytotoxicity against normal cell targets. Therefore, their applications in healthy systems should be limited.


Assuntos
Apoptose/efeitos dos fármacos , Grafite/química , Hemoglobinas/química , Linfócitos/patologia , Nanopartículas/toxicidade , Óxidos/química , Fenômenos Biofísicos , Células Cultivadas , Hemoglobinas/metabolismo , Humanos , Linfócitos/efeitos dos fármacos , Simulação de Acoplamento Molecular , Nanopartículas/química , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA