Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
NPJ Regen Med ; 9(1): 6, 2024 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-38245543

RESUMO

Mesenchymal stem cells (MSCs) are novel therapeutics for the treatment of Crohn's disease. However, their mechanism of action is unclear, especially in disease-relevant chronic models of inflammation. Thus, we used SAMP-1/YitFc (SAMP), a chronic and spontaneous murine model of small intestinal inflammation, to study the therapeutic effects and mechanism of action of human bone marrow-derived MSCs (hMSC). hMSC dose-dependently inhibited naïve T lymphocyte proliferation via prostaglandin E2 (PGE2) secretion and reprogrammed macrophages to an anti-inflammatory phenotype. We found that the hMSCs promoted mucosal healing and immunologic response early after administration in SAMP when live hMSCs are present (until day 9) and resulted in a complete response characterized by mucosal, histological, immunologic, and radiological healing by day 28 when no live hMSCs are present. hMSCs mediate their effect via modulation of T cells and macrophages in the mesentery and mesenteric lymph nodes (mLN). Sc-RNAseq confirmed the anti-inflammatory phenotype of macrophages and identified macrophage efferocytosis of apoptotic hMSCs as a mechanism that explains their long-term efficacy. Taken together, our findings show that hMSCs result in healing and tissue regeneration in a chronic model of small intestinal inflammation and despite being short-lived, exert long-term effects via sustained anti-inflammatory programming of macrophages via efferocytosis.

2.
bioRxiv ; 2023 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-37292753

RESUMO

Objective: Mesenchymal stem cells (MSCs) are novel therapeutics for treatment of Crohn's disease. However, their mechanism of action is unclear, especially in disease-relevant chronic models of inflammation. Thus, we used SAMP-1/YitFc, a chronic and spontaneous murine model of small intestinal inflammation, to study the therapeutic effect and mechanism of human bone marrow-derived MSCs (hMSC). Design: hMSC immunosuppressive potential was evaluated through in vitro mixed lymphocyte reaction, ELISA, macrophage co-culture, and RT-qPCR. Therapeutic efficacy and mechanism in SAMP were studied by stereomicroscopy, histopathology, MRI radiomics, flow cytometry, RT-qPCR, small animal imaging, and single-cell RNA sequencing (Sc-RNAseq). Results: hMSC dose-dependently inhibited naïve T lymphocyte proliferation in MLR via PGE 2 secretion and reprogrammed macrophages to an anti-inflammatory phenotype. hMSC promoted mucosal healing and immunologic response early after administration in SAMP model of chronic small intestinal inflammation when live hMSCs are present (until day 9) and resulted in complete response characterized by mucosal, histological, immunologic, and radiological healing by day 28 when no live hMSCs are present. hMSC mediate their effect via modulation of T cells and macrophages in the mesentery and mesenteric lymph nodes (mLN). Sc-RNAseq confirmed the anti-inflammatory phenotype of macrophages and identified macrophage efferocytosis of apoptotic hMSCs as a mechanism of action that explains their long-term efficacy. Conclusion: hMSCs result in healing and tissue regeneration in a chronic model of small intestinal inflammation. Despite being short-lived, exert long-term effects via macrophage reprogramming to an anti-inflammatory phenotype. Data Transparency Statement: Single-cell RNA transcriptome datasets are deposited in an online open access repository 'Figshare' (DOI: https://doi.org/10.6084/m9.figshare.21453936.v1 ).

3.
Int J Biol Macromol ; 232: 123283, 2023 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-36657541

RESUMO

Hydrogels have emerged as a versatile platform for a numerous biomedical application due to their ability to absorb a huge quantity of biofluids. In order to design hydrogels, natural polymers are an attractive option owing to their biocompatibility and biodegradability. Due to abundance in occurrence, cost effectiveness, and facile crosslinking approaches, alginate has been extensively investigated to fabricate hydrogel matrix. Management of cancer and chronic wounds have always been a challenge for pharmaceutical and healthcare sector. In both cases, curcumin have been shown significant improvement and effectiveness. However, the innate restraints like poor bioavailability, hydrophobicity, and rapid systemic clearance associated with curcumin have restricted its clinical translations. The current review explores the cascade of research around curcumin encapsulated alginate hydrogel matrix for wound healing and cancer therapy. The focus of the review is to emphasize the mechanistic effects of curcumin with its fate inside the cells. Further, the review discusses different approaches to designed curcumin loaded alginate hydrogels along with the parameters that regulates their release behavior. Finally, the review is concluded with emphasize on some key aspect on increasing the efficacy of these hydrogels along with novel strategies to further develop curcumin loaded alginate hydrogel matrix with multifacet applications.


Assuntos
Curcumina , Neoplasias , Hidrogéis/farmacologia , Curcumina/farmacologia , Curcumina/uso terapêutico , Alginatos/farmacologia , Cicatrização , Polímeros/farmacologia
4.
Small ; 19(12): e2206401, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36585372

RESUMO

Stimulation of cells with electrical cues is an imperative approach to interact with biological systems and has been exploited in clinical practices over a wide range of pathological ailments. This bioelectric interface has been extensively explored with the help of piezoelectric materials, leading to remarkable advancement in the past two decades. Among other members of this fraternity, colloidal perovskite barium titanate (BaTiO3 ) has gained substantial interest due to its noteworthy properties which includes high dielectric constant and excellent ferroelectric properties along with acceptable biocompatibility. Significant progression is witnessed for BaTiO3 nanoparticles (BaTiO3 NPs) as potent candidates for biomedical applications and in wearable bioelectronics, making them a promising personal healthcare platform. The current review highlights the nanostructured piezoelectric bio interface of BaTiO3 NPs in applications comprising drug delivery, tissue engineering, bioimaging, bioelectronics, and wearable devices. Particular attention has been dedicated toward the fabrication routes of BaTiO3 NPs along with different approaches for its surface modifications. This review offers a comprehensive discussion on the utility of BaTiO3 NPs as active devices rather than passive structural unit behaving as carriers for biomolecules. The employment of BaTiO3 NPs presents new scenarios and opportunity in the vast field of nanomedicines for biomedical applications.


Assuntos
Nanopartículas , Nanoestruturas , Bário , Compostos de Bário/química
5.
ACS Med Chem Lett ; 13(7): 1109-1117, 2022 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-35859882

RESUMO

Neuroblastoma (NB) is an extracranial pediatric tumor with highly invasive growth of cancer biomass and frequent metastases. During the differentiation process in embryonic development, altered epigenetic modifications lead to dysregulated expression of pluripotency markers, resulting in epithelial-mesenchymal transition (EMT) progression. Currently, available chemotherapies have provided a limited solution to this problem due to systemic toxicities and drug resistance. Epigenetic therapeutic molecules like histone deacetylase inhibitors are still in the initial stages of development. We have developed a retinoid (N-(4-hydroxyphenyl) retinamide, 4HPR) loaded acetylated human serum albumin (HSA) nanoformulation to address the epigenetic imbalance and chemoresistance in NB. The idea was conceived to deliver an acetyl pool along with a chemotherapeutic drug, 4HPR, to restrict the invasiveness of NB by maintaining the balance between histone acetylation and trimethylation. The therapeutic efficacy of the formulation was successfully evaluated in the in vitro and in vivo xenograft mouse model system of neuroblastoma. The synthesized nanoparticles show high biocompatibility and therapeutic efficacy in treating neuroblastoma subcutaneous xenografts in nude mice.

6.
Pharmaceutics ; 14(5)2022 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-35631579

RESUMO

The intrinsic architecture and complexity of the brain restricts the capacity of therapeutic molecules to reach their potential targets, thereby limiting therapeutic possibilities concerning neurological ailments and brain malignancy. As conventional models fail to recapitulate the complexity of the brain, progress in the field of microfluidics has facilitated the development of advanced in vitro platforms that could imitate the in vivo microenvironments and pathological features of the blood-brain barrier (BBB). It is highly desirous that developed in vitro BBB-on-chip models serve as a platform to investigate cancer metastasis of the brain along with the possibility of efficiently screening chemotherapeutic agents against brain malignancies. In order to improve the proficiency of BBB-on-chip models, hydrogels have been widely explored due to their unique physical and chemical properties, which mimic the three-dimensional (3D) micro architecture of tissues. Hydrogel-based BBB-on-chip models serves as a stage which is conducive for cell growth and allows the exchange of gases and nutrients and the removal of metabolic wastes between cells and the cell/extra cellular matrix (ECM) interface. Here, we present recent advancements in BBB-on-chip models targeting brain malignancies and examine the utility of hydrogel-based BBB models that could further strengthen the future application of microfluidic devices in oncology research.

7.
Life Sci ; 302: 120655, 2022 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-35598656

RESUMO

AIMS: Inflammatory Bowel Disease is characterised by abdominal pain, diarrhoea, rectal bleeding and weight loss. Sometimes it may leads to severe health complications resulting in death of an individual. Current research efforts to highlight the role of melatonin in regulating EZH2, a master epigenetic regulator and its beneficiary effect in case of IBD management. MATERIAL METHODS: Murine macrophages (RAW 264.7) were treated with lipopolysaccharides (LPS) to activate them for generating inflammatory response to investigate efficacy of melatonin in-vitro models. Similarly, for developing in vivo models, Dextran sodium sulphate (36-50 kDa) was used. Evaluations of anti-inflammatory activities were carried out by nitrite assay, western blotting, q-PCR, immunofluorescence, and histological studies. KEY FINDINGS: Reduction of epigenetic target, EZH2 by melatonin significantly improves the clinical symptoms of dextran sodium sulphate induced colitis and may be implicated as a potential therapeutic target in IBD management. The present study evaluates the efficacy of melatonin by epigenetic regulation in IBD models. Down regulation of EZH2 by melatonin reduced the chemical induced inflammatory insults in in vitro and in vivo models. Exploration of molecular pathways has revealed interlink of EZH2 and NOS2, a hallmark of inflammation. Molecular mechanistic action of melatonin is attributed to inhibition of the expression and physical interaction of EZH2 and NOS2. SIGNIFICANCE: Our study highlights melatonin therapeutic effect via attenuating interaction between EZH2 and NOS2 which is beneficial in managing IBD treatment.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Melatonina , Animais , Camundongos , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/genética , Sulfato de Dextrana/toxicidade , Dextranos/metabolismo , Modelos Animais de Doenças , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epigênese Genética , Doenças Inflamatórias Intestinais/patologia , Melatonina/farmacologia , Melatonina/uso terapêutico , Óxido Nítrico Sintase Tipo II/metabolismo
9.
Mater Sci Eng C Mater Biol Appl ; 129: 112394, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34579913

RESUMO

The ability of some tumours to impart radioresistance serves as a barrier in the cancer therapeutics. Mitochondrial metabolism significantly persuades this cancer cell survival, incursion and plays a crucial role in conferring radioresistance. It would be of great importance to target the active mitochondria to overcome this resistance and achieve tumoricidal efficacy. The current report investigates the improved radiosensitization effect (under Gamma irradiation) in hepatocellular carcinoma through active mitochondrial targeting of alpha-ketoglutarate decorated iron oxide-gold core-shell nanoparticles (GNP). The loading of a chemotherapeutic drug N-(4-hydroxyphenyl)retinamide in GNP allows adjuvant chemotherapy, which further sensitizes cancerous cells for radiotherapy. The GNP shows a drug loading efficiency of 8.5 wt% with a sustained drug release kinetics. The X-Ray diffraction (XRD) pattern and High-Resolution Transmission Electron microscopy (HRTEM) indicates the synthesis of core iron oxide nanoparticles with indications of a thin layer of gold shell on the surface with 1:7 ratios of Fe: Au. The GNP application significantly reduced per cent cell viability in Hepatocellular carcinoma cells through improved radiosensitization at 5 Gy gamma radiation dose. The molecular mechanism revealed a sharp increment in reactive oxygen species (ROS) generation and DNA fragmentation. The mitochondrial targeting probes confirm the presence of GNP in the mitochondria, which could be the possible reason for such improved cellular damage. In addition to the active mitochondrial targeting, the currently fabricated nanoparticles work as a potent Magnetic Resonance Imaging (MRI)/Computed Tomography (CT) contrast agent. This multifunctional therapeutic potential makes GNP as one of the most promising theragnostic molecules in cancer therapeutics.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Nanopartículas Metálicas , Carcinoma Hepatocelular/tratamento farmacológico , Compostos Férricos , Ouro , Humanos , Ácidos Cetoglutáricos , Neoplasias Hepáticas/tratamento farmacológico , Mitocôndrias
10.
Sci Total Environ ; 772: 144797, 2021 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-33578167

RESUMO

Surface functionalization on silver nanoparticles greatly affects the dynamics of protein corona formation. In the present study, the implications of protein pre-coating on corona formation and nanoparticle's physiological stability, cellular uptake and toxicity were studied on similar sized alkaline protease coated nanoparticles of biological and chemical origin along with the uncoated nanoparticle as compared to the albumin coated nanoparticles. All four nanoparticle types invited serum protein adsorption on their surface. However, the presence of protein pre-coating on nanoparticle surface significantly reduced the extent of further protein binding. Moreover, corona formation on pristine nanoparticles significantly improved their stability in the biological medium. The effect was found to be diluted in protein pre-coated nanoparticles with due exception. Results obtained in the cell-based experiment suggested that the nanoparticles binding to the cell, its uptake, and toxicity in different cell lines can be directly linked to their physiological stability owing to corona formation.


Assuntos
Nanopartículas Metálicas , Nanopartículas , Coroa de Proteína , Adsorção , Nanopartículas Metálicas/toxicidade , Proteínas , Prata/toxicidade
11.
Phytomedicine ; 80: 153386, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33113500

RESUMO

BACKGROUND: Overexpression of polycomb protein contributes to epigenetic repression in oral squamous cell carcinoma (OSCC) ensuing in poor prognosis and aggressive phenotype. Several plant-based compounds could help prevent epigenome alteration and cancer progression, but their low bioavailability limits their therapeutic activity. HYPOTHESIS: In this study, we have synthesized genistein nanoformulation (GLNPs) and evaluated its epigenetic regulation mechanism for selective apoptosis induction in OSCC. METHODS: Lactalbumin was used to prepare nanoformulation of Genistein. The mechanism of epigenetic regulation and selective apoptosis by Genistein loaded nanoparticles was studied in OSCC cell line JHU011 and fibroblast cell line L929 using immunofluorescence, Western blotting and ChIP-qPCR assay. RESULTS: We have found that GLNPs treatment selectively induced apoptosis in OSCC compared to the normal fibroblast cells. This selective effect in OSCC is achieved through enhanced reactive oxygen species (ROS) generation followed by Bax mitochondrial translocation and caspase 3 activation. Further, GLNPs induced withdrawal of epigenetic transcription repression through concurrent downregulation of the polycomb group proteins (PcG) Bmi 1 and EZH2 along with their successive targets, UbH2AK119 and H3K27me3, which have immense therapeutic implications in the treatment of OSCC. Last, we have established that GLNPs regulate EZH2expression through proteasomal mediated degradation and 3PK inhibition; 3PK protein was found physically linked with EZH2 protein and its promoter region (-1107 to -1002). This event indicates that 3PK might play some crucial role in EZH2 expression and epigenetic control of OSCC. Moreover, the formulation showed improved biodistribution, aqueous dispersibility and enhanced biocompatibility In-vivo. CONCLUSIONS: These results provide evidence that GLNPs may withdraw epigenetic transcriptional repression and selectively induce apoptosis in human oral squamous cell carcinoma.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Genisteína/farmacologia , Neoplasias Bucais/tratamento farmacológico , Animais , Antineoplásicos Fitogênicos/farmacocinética , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Epigênese Genética/efeitos dos fármacos , Genisteína/administração & dosagem , Genisteína/farmacocinética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Neoplasias Bucais/genética , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Nanopartículas/administração & dosagem , Nanopartículas/química , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Acta Biomater ; 109: 121-131, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32335311

RESUMO

Wound healing remains a healthcare challenge in patients suffering from grave tissue damage due to burn injuries and severe medical conditions like diabetes and ischemia. A repeated wound dressing in such cases leads to tissue damage, which could further inflate the wound healing. It is also challenging to analyze the depth of wound bed in these conditions, which could affect the recovery period. To address this need, we have developed an injectable hydrogel from natural polysaccharide κ-carrageenan and a pigmented protein C-phycocyanin. C-phycocyanin has wound healing, antimicrobial, antioxidant and anti-inflammatory properties along with the In-vivo fluorescence imaging ability. Gelling property of κ-carrageenan could be utilized along with C-phycocyanin as an injectable and regenerative wound dressings matrix to monitor wound healing in real-time without upsetting the healing process. The hydrogel presented herein was built from ionic crosslinking of κ-carrageenan monomers along with C-phycocyanin, which provides an interconnected network of porous material with hydrophilic surface and mechanical stiffness. This porosity allows nutrients transportation and gaseous exchange across the wound healing site for the proliferation of various cells. Hydrogel material enhances the proliferation of dermal fibroblasts in vitro without inducing inflammation along with reducing the blood clotting time with no haemolysis. We have found that κ-carrageenan-C-phycocyanin (κ-CRG-C-Pc) hydrogel not only exhibit superior haemostatic capabilities in traumatic injury condition but also provide support for rapid wound healing. Overall, these findings demonstrate the potential of κ-carrageenan-C-phycocyanin hydrogels as a wound-healing and imaging platform towards accelerating tissue repair and real-time monitoring. STATEMENT OF SIGNIFICANCE: Blood clotting and inflammation are the most crucial stages of wound healing along with appropriate monitoring of the healing process. Thus, there is a need of system that could provide point-to-point care and monitoring in this multistage process. Here, we have introduced a self healing, injectable hydrogel system with in vivo imaging abilities from κ-carragenan and C-phycocyanin. C-phycocyanin improves the stability of κ-carragenan matrix and provide support to cellular adhesion, proliferation, and migration. Its anti-inflammatory response and rapid blood clotting ability further empower its applicability in critical medical conditions and wound recovery.


Assuntos
Anti-Inflamatórios/uso terapêutico , Carragenina/química , Hemostáticos/uso terapêutico , Hidrogéis/química , Ficocianina/uso terapêutico , Cicatrização/efeitos dos fármacos , Animais , Anti-Inflamatórios/química , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Corantes Fluorescentes/química , Corantes Fluorescentes/uso terapêutico , Cabras , Hemólise/efeitos dos fármacos , Hemostáticos/química , Hidrogéis/síntese química , Inflamação/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Imagem Óptica , Ficocianina/química , Porosidade , Células RAW 264.7 , Ratos Wistar
13.
Mol Pharm ; 17(2): 604-621, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31904978

RESUMO

Acute myeloid leukemia (AML) is a malignant disorder of hematopoietic progenitor cells with a poor prognosis of 26% of patients surviving 5 years after diagnosis. Poor bioavailability and solubility are significant factors limiting the efficacy of chemopreventive agents. In AML, the epigenetic regulator polycomb group of protein member EZH2 is highly expressed and is essential for the survival of leukemic cells. An EZH2-specific inhibitor, EPZ011989, encapsulated in human serum albumin nanoparticles (HSANPs) was synthesized for the first time via the desolvation method. The noncovalent interactions between EPZ011989 and HSANPs in nanocomposites facilitating the efficient loading and sustainable release of the drug showed enhanced cellular uptake and nuclear localization of EPZ011989-loaded HSANPs in human AML cell lines. The reduction of cell viability, colony formation inhibition, cell cycle arrest at the G2/M phase, and cell proliferation assay promoting apoptosis through the loss of mitochondrial homeostasis exerting antileukemic activity were evident. The real-time polymerase chain reaction (PCR) and western blot-based studies showed that the present nanoformulation reduces the level of PcG proteins, including EZH2, BMI-1, etc. This downregulation is associated with reduced H3K27me3 and H2AK119ub modifications conferring chromatin compaction. The immunoprecipitation study showed the physical interaction of EZH2 and c-Myb can be linked to the regulation of leukemogenesis. Further investigation revealed the mechanism of EZH2 and c-Myb downregulation via ubiquitination and proteasomal degradation pathway, confirmed by using proteasome inhibitor, suggesting the key role of proteasomal degradation machinery. Moreover, c-Myb interacted with the EZH2 promoter, which is evident by the chromatin immunoprecipitation assay and siRNA silencing. Furthermore, the formulation of EPZ011989 in HSANPs improved its biodistribution in vivo and showed excellent aqueous dispersibility and biocompatibility. In vivo studies further showed that EPZ011989-loaded HSANPs reduce the expression of CD11b+ and CD45+ markers in immunophenotyping from peripheral blood and bone marrow in engrafted nude mice. Targeted depletion of EZH2 alleviated the disease progression in nude mice and prolonged their survival. The findings provide valuable experimental evidence for the targeted epigenetic therapy of AML. The present results demonstrate an epigenetic regulation-based superior antileukemic therapy.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Epigênese Genética/efeitos dos fármacos , Leucemia Mieloide Aguda/tratamento farmacológico , Nanopartículas/administração & dosagem , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myb/genética , Animais , Sobrevivência Celular/efeitos dos fármacos , Composição de Medicamentos/métodos , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Células HL-60 , Humanos , Leucemia Mieloide Aguda/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/química , Albumina Sérica Humana/química , Distribuição Tecidual , Transfecção , Células U937 , Ensaios Antitumorais Modelo de Xenoenxerto
14.
ACS Biomater Sci Eng ; 6(5): 3139-3153, 2020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33463265

RESUMO

Epigenetically regulated therapeutic intervention of cancer is an emerging era of research in the development of a promising therapy. Epigenetic changes are intrinsically reversible and providing the driving force to drug resistance in colorectal cancer (CRC). The regulation of polycomb group (PcG) proteins, BMI1 and EZH2, and the associated CRC progression hold promises for a novel treatment regime. The present study enlightens targeted photodynamic therapy (PDT) with potential photosensitizer hypericin nanocomposite in the development of epigenetic-based CRC therapy. We have synthesized hypericin-loaded transferrin nanoformulations (HTfNPs) overcoming the compromised hydrophobicity and poor bioavailability of the placebo drug. Targeted PDT with hypericin nanocomposite-induced BMI1 degradation assisted CRC retardation. In the present study, transferrin nanoparticles were reported to control the premature release of hypericin and improve its availability with better targeting at the disease site. Targeted intracellular internalization to colon cancer cells having a differential expression of transferrin receptors, in vivo biodistribution, stability, and pharmacokinetics provide promising applications in the nanodelivery system. Indeed, in vitro anticancer efficiency, cell cycle arrest at the G0/G1 phase, and elevated reactive oxygen species (ROS) generation confirm the anticancer effect of nanoformulation. In the exploration of mechanism, nanotherapeutic intervention by activation of PP2A, Caspase3 and inhibition of BMI1, EZH2, 3Pk, NFκB was evident. An exciting outcome of this study uncovered the camouflaged role of PP2A in the regulation of BMI1. PP2A mediates the ubiquitination/degradation of BMI1, which is revealed by changes in the physical interaction of PP2A and BMI1. Our study confirms the anticancer effect of HTfNP-assisted PDT by inducing PP2A-mediated BMI1 ubiquitination/degradation demonstrating an epigenetic-driven nanotherapeutic approach in CRC treatment.


Assuntos
Neoplasias Colorretais , Nanopartículas , Fotoquimioterapia , Antracenos , Neoplasias Colorretais/tratamento farmacológico , Humanos , Perileno/análogos & derivados , Distribuição Tecidual , Transferrina
15.
Mater Sci Eng C Mater Biol Appl ; 107: 110315, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31761231

RESUMO

We have prepared a water dispersible Nanocrystalline hierarchical structure of paclitaxel using self-assembly approach. Its unique structure provides the advantage of increased payload and higher retention of a chemotherapeutic drug inside the tumor site for a prolonged period. Micron scale structure, sustained release behaviour, multichannel fluorescence property and devoid of any vehicle molecule makes it promising in treating solid tumors through localized drug delivery approach like TACE. This method is based on one pot synthesis through co-precipitation reaction followed by crosslinking. Therapeutic efficacy of the molecules is successfully verified through the significant reduction in the volume of the 3D spheroid model of hepatocellular carcinoma (HCC).


Assuntos
Carcinoma Hepatocelular/terapia , Quimioembolização Terapêutica/métodos , Neoplasias Hepáticas/terapia , Paclitaxel/química , Paclitaxel/farmacologia , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacocinética , Antineoplásicos Fitogênicos/farmacologia , Liberação Controlada de Fármacos , Células Hep G2 , Humanos , Nanopartículas/química , Paclitaxel/farmacocinética , Espectroscopia de Infravermelho com Transformada de Fourier , Esferoides Celulares/efeitos dos fármacos , Difração de Raios X
16.
Carbohydr Polym ; 180: 365-375, 2018 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-29103516

RESUMO

Recurrence of glioblastoma is one of the major concerns due to its heterogeneous nature and association of Glioma Initiating stem-like Cells (GICs). Nanoparticles mediated delivery of chemotherapeutic agent targeting both cancer and glioma stem cells could provide a solution to recurrent malignancies of the glioblastoma tumor. The approach described here provides enhanced chemotherapeutic potency utilizing 1,3ß-Glucan as an outer shell to the chitosan nanoparticles (Cs-NPs) loaded with paclitaxel to prevent hemolysis with, the core-shell nano-structure (Cs-PTX-NP) enabling effective chemotherapy against malignant glioblastoma. The prepared nanoparticles (1,3ß-Cs-PTX-NPs) with sustained release of the paclitaxel provide a targeted therapeutic approach that overcome systemic toxicities with the 1,3ß-Glucan shell and improve drug bioavailability. Hemolysis investigation indicated that 1,3ß-Cs-PTX-NP was significantly less hemolytic than paclitaxel enabling intravenous delivery. Also, 1,3ß-Cs-PTX-NPs were considerably more cytotoxic (IC50) against glioma cancer LN18 cells and C6 stem-like cells compared with the PTX. In conclusion, this study found that 1,3ß-Cs-PTX-NP addressed serious limitation with systemic delivery of paclitaxel by preventing hemolysis and providing chemotherapeutic delivery with significant anti-cancer efficacy against recurrent glioblastoma.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Quitosana/análogos & derivados , Glucanos/química , Hemólise/efeitos dos fármacos , Nanopartículas/química , Paclitaxel/administração & dosagem , Antineoplásicos Fitogênicos/efeitos adversos , Linhagem Celular Tumoral , Liberação Controlada de Fármacos , Humanos , Nanopartículas/efeitos adversos , Paclitaxel/efeitos adversos
17.
Nanotechnology ; 28(36): 365102, 2017 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-28820142

RESUMO

Melatonin (MEL) has promising medicinal value as an anticancer agent in a variety of malignancies, but there are difficulties in achieving a therapeutic dose due to its short half-life, low bioavailability, poor solubility and extensive first-pass metabolism. In this study chitosan/tripolyphosphate (TPP) nanoparticles were prepared by an ionic gelation method to overcome the therapeutic challenges of melatonin and to improve its anticancer efficacy. Characterization of the melatonin-loaded chitosan (MEL-CS) nanoformulation was performed using transmission and scanning electron microscopies, dynamic light scattering, Fourier transform infrared spectroscopy, Raman spectroscopy and x-ray diffraction. In vitro release, cellular uptake and efficacy studies were tested for their enhanced anticancer potential in human U87MG glioblastoma cells. Confocal studies revealed higher cellular uptake of MEL-CS nanoparticles and enhanced anticancer efficacy in human malignant glioblastoma cancer cells than in healthy non-malignant human HEK293T cells in mono- and co-culture models. Our study has shown for the first time that MEL-CS nanocomposites are therapeutically more effective as compared to free MEL at inducing functional anticancer efficacy in the human brain tumour U87MG cell line.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...