Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Mol Pharm ; 20(6): 2899-2910, 2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-37116080

RESUMEN

Oxidative stress, reactive oxygen species generation, and overexpression of VEGF are signatory events in diabetic retinopathy. The downregulation of VEGF and anti-inflammatory action pave the way for diabetic retinopathy (DR) therapy. In that, lower absorption kinetics of melatonin limits its immense therapeutic potential. Hence, we have demonstrated a reverse microemulsion method to synthesize melatonin-loaded polydopamine nanoparticles to replenish both at a single platform with an improved melatonin delivery profile. The study has evaluated in vitro and in vivo protection efficiency of biocompatible melatonin-loaded polydopamine nanoparticles (MPDANPs). The protection mechanism was explained by downregulation of VEGF, CASPASE3, and PKCδ against high-glucose/streptozotocin (STZ)-induced insults, in vitro and in vivo. The anti-inflammatory and antiangiogenic effect and potential of MPDANPs to enhance melatonin in vivo stability with prolonged circulation time have proved MPDANPs as a potential therapeutic candidate in DR management. The DR therapeutic potential of MPDANPs has been arbitrated by improving the bioavailability of melatonin and inhibition of VEGF-PKCδ crosstalk in vivo.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Melatonina , Humanos , Retinopatía Diabética/tratamiento farmacológico , Melatonina/farmacología , Melatonina/uso terapéutico , Retina , Factor A de Crecimiento Endotelial Vascular
2.
Mol Pharm ; 17(2): 604-621, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31904978

RESUMEN

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.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2/genética , Epigénesis Genética/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Nanopartículas/administración & dosificación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myb/genética , Animales , Supervivencia Celular/efectos de los fármacos , Composición de Medicamentos/métodos , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Células HL-60 , Humanos , Leucemia Mieloide Aguda/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/química , Albúmina Sérica Humana/química , Distribución Tisular , Transfección , Células U937 , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Nanomedicine ; 24: 102088, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31476446

RESUMEN

Posttranslational modification and agglomeration of α-synuclein (α-Syn), mitochondrial dysfunction, oxidative stress and loss of dopaminergic neurons are hallmark of Parkinson's disease (PD). This paper evaluates neuroprotection efficacy of nature inspired biocompatible polydopamine nanocarrier for metformin delivery (Met encapsulated PDANPs) by crossing blood brain barrier in in vitro, 3D and in vivo experimental PD models. The neuroprotective potential was arbitrated by downregulation of phospho-serine 129 (pSer129) α-Syn, with reduction in oxidative stress, prevention of apoptosis and anti-inflammatory activities. The neuroprotective mechanism proved novel interaction of epigenetic regulator EZH2 mediated ubiquitination and proteasomal degradation of aggregated pSer129 α-Syn. In summary, this study divulges the neuroprotective role of Met loaded PDANPs by reversing the neurochemical deficits by confirming an epigenetic mediated nanotherapeutic approach for the PD prevention.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Metformina , Modelos Biológicos , Nanoestructuras , Enfermedad de Parkinson/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , alfa-Sinucleína/metabolismo , Línea Celular Tumoral , Humanos , Indoles/química , Indoles/farmacología , Metformina/química , Metformina/farmacología , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Polímeros/química , Polímeros/farmacología
4.
Nanotechnology ; 28(36): 365102, 2017 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-28820142

RESUMEN

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.

5.
Small ; 10(24): 5068-76, 2014 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-25060135

RESUMEN

A rationally designed two-step synthesis of silica vesicles is developed with the formation of vesicular structure in the first step and fine control over the entrance size by tuning the temperature in the second step. The silica vesicles have a uniform size of ≈50 nm with excellent cellular uptake performance. When the entrance size is equal to the wall thickness, silica vesicles after hydrophobic modification show the highest loading amount (563 mg/g) towards Ribonuclease A with a sustained release behavior. Consequently, the silica vesicles are excellent nano-carriers for cellular delivery applications of therapeutical biomolecules.


Asunto(s)
Preparaciones de Acción Retardada , Portadores de Fármacos , Ribonucleasa Pancreática/administración & dosificación , Dióxido de Silicio/química , Línea Celular , Microscopía Electrónica de Rastreo
6.
Mol Pharm ; 11(10): 3642-55, 2014 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-25166282

RESUMEN

Mesoporous silica nanoparticles (MCM-41) with different surface chemistry were used as carrier system to study its influence on drug delivery and anticancer activity of curcumin (CUR). CUR was encapsulated in pristine MCM-41 (hydrophilic and negatively charged), amino functionalized MCM-41 (MCM-41-NH2 which is hydrophilic and positively charged), and methyl functionalized MCM-41 (MCM-41-CH3 which is hydrophobic and negatively charged) and evaluated for in vitro release and cell cytotoxicity in human squamous cell carcinoma cell line (SCC25). Various techniques were employed to evaluate the performance of these materials on cellular uptake and anticancer activity in the SCC25 cell line. Both positively and negatively charged surfaces demonstrated enhanced drug release and anticancer activity compared to pure CUR. Positively charged nanoparticles showed higher cell uptake compared to negatively charged nanoparticles owing to its electrostatic interaction with cells. However, hydrophobic surface modified nanoparticles (MCM-41-CH3) showed no improvement in drug release and anticancer activity due to its poor wetting effect. Cell cycle analysis and cell apoptosis studies revealed different pathway mechanisms followed by the positively and negatively charged nanoparticles but exhibiting similar anticancer activity in SCC25 cells.


Asunto(s)
Nanopartículas/química , Dióxido de Silicio/química , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Curcumina/química , Curcumina/farmacocinética , Curcumina/farmacología , Humanos
7.
ACS Appl Mater Interfaces ; 16(7): 8417-8429, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38344952

RESUMEN

The molecular pathways that melatonin follows as a Parkinson's disease (PD) antagonist remain poorly elucidated, despite it being a safe and a potential neurotherapeutic drug with a few limitations such as less bioavailability, premature oxidation, brain delivery, etc. Here, we used a biocompatible protein (HSA) nanocarrier for the delivery of melatonin to the brain. This nanomelatonin showed better antioxidative and neuroprotective properties, and it not only improves mitophagy to remove unhealthy mitochondria but also improves mitochondrial biogenesis to counteract rotenone-induced toxicity in an in vitro PD model. We also showed BMI1, a member of the PRC1 complex that regulates mitophagy, whose protein expression was enhanced after nanomelatonin dosage. These effects were translated to a rodent model, where nanomelatonin improves the TH+ve neuron population in SNPC and protects against rotenone-mediated toxicity. Our findings highlight the significantly better in vitro and in vivo neuroprotective effect of nanomelatonin as well as the molecular/cellular dynamics it influences to regulate mitophagy as a measure of the potential therapeutic candidate for PD.


Asunto(s)
Melatonina , Nanopartículas , Fármacos Neuroprotectores , Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/tratamiento farmacológico , Melatonina/farmacología , Melatonina/uso terapéutico , Mitofagia , Rotenona/farmacología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico
8.
Nanoscale ; 16(34): 16075-16088, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39087878

RESUMEN

Neuroblastoma is the most prevalent and aggressive solid tumor that develops extracranially in children between the ages of 0-14 years, which accounts for 8-10% of all childhood malignancies and ∼15% of pediatric cancer-related mortality. The polycomb repressive complex 2 (PRC2) protein, EZH2, is overexpressed in neuroblastoma and mediates histone H3 methylation at lysine 27 (K27) positions through its methyl transferase activity and is a potential epigenetic silencer of many tumor suppressor genes in cancer. Phosphorylation of EZH2 decreases its stability and leads to proteasomal degradation. The 4-oxo-N-(4-hydroxyphenyl) retinamide (4O4HPR) promotes EZH2 degradation via activation of PKC-δ, but its limited solubility and physiological instability limit its application. In the current study, the encapsulation of 4O4HPR in Human Serum Albumin Nanoparticles (HSANPs) enhanced the solubility and physiological stability of the nanoformulation, leading to improved therapeutic efficacy through G2-M cell cycle arrest, depolarization of mitochondrial membrane potential, generation of reactive oxygen species and caspase 3 mediated apoptosis activation. The molecular mechanistic approach of 4O4HPR loaded HSANPs has activated caspase 3, which further cleaves PKC-δ into two fragments wherein the cleaved fragment of PKC-δ possesses the kinase activity that phosphorylates EZH2 and decreases the protein stability leading to its further ubiquitination in SH-SY5Y cells. Co-immunoprecipitation experiments revealed the direct interaction between PKC-δ and EZH2 phosphorylation, followed by ubiquitination. Moreover, 4O4HPR loaded HSANPs demonstrated improved in vivo biodistribution, greater dispersibility, and biocompatibility and exhibited enhanced protein instability and degradation of EZH2 in the neuroblastoma xenograft mouse model.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2 , Nanopartículas , Neuroblastoma , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/química , Humanos , Neuroblastoma/patología , Neuroblastoma/metabolismo , Neuroblastoma/tratamiento farmacológico , Animales , Línea Celular Tumoral , Nanopartículas/química , Ratones , Albúmina Sérica Humana/química , Albúmina Sérica Humana/metabolismo , Apoptosis/efectos de los fármacos , Fenretinida/química , Fenretinida/farmacología , Especies Reactivas de Oxígeno/metabolismo , Ratones Desnudos , Proteolisis/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Antineoplásicos/química , Antineoplásicos/farmacología , Caspasa 3/metabolismo
9.
ACS Chem Neurosci ; 15(1): 71-77, 2024 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-38109795

RESUMEN

The post-translational modification and aggregation of alpha-synuclein are one of the major causes of Parkinson's disease (PD) regulation. In that, the phosphorylation and nitration of synuclein elevate the aggregation, while O-GlcNacylation prevents the aggregation of synuclein. The inhibition of synuclein aggregation directs the development of PD therapy. The endowed O-GlcNacylation of synuclein could be a promising strategy to inhibit synucleinopathy. Therefore, the neuroprotective chitosan-based FTY720 nanoformulation, PP2A (Protein phosphatase 2) activator has been employed to evaluate the PP2A role in the O-GlcNacylation of synuclein in an in vivo PD model. The neuroprotective effect of our nanoformulation is attributed to the upregulation of tyrosine hydroxylase (TH), the PD therapeutic target, with behavioral improvement in animals against rotenone-induced PD deficits. The neuroprotective molecular insights revealed the camouflaged role of PP2A by endowing the OGT activity that induces O-GlcNacylation of synuclein in the reduction of synucleinopathy.


Asunto(s)
Enfermedad de Parkinson , Sinucleinopatías , Animales , Sinucleinopatías/tratamiento farmacológico , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , alfa-Sinucleína/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Fosforilación , Procesamiento Proteico-Postraduccional
10.
Carbohydr Polym ; 346: 122575, 2024 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-39245478

RESUMEN

Many neurodegenerative and psychiatric malignancies like Parkinson' disease (PD) originate from an imbalance of 17ß-Estradiol (E2) in the human brain. However, the peripheral side effects of the usage of E2 for PD therapy and less understanding of the molecular mechanism hinder establishing its neurotherapeutic potential. In the present work, systemic side effects were overcome by targeted delivery using Dopamine receptor D3 (DRD3) conjugated E2-loaded chitosan nanoparticles (Ab-ECSnps) that showed a promising delivery to the brain. E2 is a specific calpain inhibitor that fosters neurodegeneration by disrupting mitochondrial function, while B-cell-specific Moloney murine leukemia virus integration region 1 (BMI1), an epigenetic regulator, is crucial in preserving mitochondrial homeostasis. We showed the administration of Ab-ECSnps inhibits calpain's translocation into mitochondria while promoting the translocation of BMI1 to mitochondria, thereby conferring neurotherapeutic benefits by enhancing cell viability, increasing mitochondrial DNA copy number, and preserving mitochondrial membrane potential. Further, we showed a novel molecular mechanism of BMI1 regulation by calpain that might contribute to maintaining mitochondrial homeostasis for attenuating PD. Concomitantly, Ab-ECSnps showed neurotherapeutic potential in the in vivo PD model. We showed for the first time that our brain-specific targeted delivery might regulate calpain-mediated BMI1 expression, thereby preserving mitochondrial homeostasis to alleviate PD.


Asunto(s)
Calpaína , Quitosano , Mitocondrias , Nanopartículas , Enfermedad de Parkinson , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Calpaína/metabolismo , Calpaína/genética , Animales , Enfermedad de Parkinson/tratamiento farmacológico , Nanopartículas/química , Quitosano/química , Humanos , Ratones , Epigénesis Genética/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Supervivencia Celular/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL
11.
ACS Appl Mater Interfaces ; 15(10): 12708-12718, 2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36857164

RESUMEN

Transferrin is an iron transporting protein consisting of bilobal protein shells (apotransferrin) with dual domains in each lobe, holding an interdomain iron binding cleft. This cleft is useful in synthesizing an iron oxide core inside the transferrin shell. In vitro reconstitution chemistry provides a nano-dimensional synthesis of the mineral core inside the protein shell. The present study demonstrates the synthesis of magnetotransferrin with reconstitution of apotransferrin to form iron oxide nanoparticles within the transferrin. Transmission electron microscopy investigations along with analysis of electronic diffraction patterns and magnetometry studies indicate entrapment of superparamagnetic iron (III) oxide nanoparticles. In vivo/ex vivo imaging of the brain and immunogold staining of brain sections further validate the brain targeting potential of "magnetotransferrin". The in vivo therapeutic potential of magneto transferrin has been demonstrated by induction of TRPV1 magnetic stimuli protein, having an important regulatory role in Parkinsonism management. In an exploration of neuroprotective mechanisms, deacetylation of H3K27 of synuclein has been revealed through the TRPV1-mediated HDAC3 activation in the treatment of Parkinsonism. Thus, this magnetic protein could be a potent candidate for brain targeting, bio-imaging, and therapy of neurological infirmities.


Asunto(s)
Hierro , Transferrina , Transferrina/química , Hierro/metabolismo , Encéfalo/metabolismo , Magnetismo
12.
Biomater Sci ; 10(18): 5301-5317, 2022 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-35917200

RESUMEN

Glioblastoma (GBM) is a complex brain cancer with frequent relapses and high mortality and still awaits effective treatment. Mitochondria dysfunction is a pathogenic condition in GBM and could be a prime therapeutic target for ceasing GBM progression. Strategies to overcome brain solid tumor barriers and selectively target mitochondria within specific cell types may improve GBM treatment. Here, we present hypericin-conjugated gold nanoparticles (PEG-AuNPs@Hyp) where hypericin is a mitochondrion-targeting agent exhibiting multimodal therapy by critically impacting the IDH2 gene (Isocitrate dehydrogenase) and its interaction with polycomb methyltransferase EZH1/2 for GBM therapy. It significantly localizes in mitochondria by enhanced cellular uptake in the human GBM cell lines/three-dimensional (3D) culture model under red-light exposure. It triggers oxidative stress and changes the mitochondrial potential, with increased Bax/Bcl2 ratio enhancing GBM cell death. The suppressed expression of mutated IDH2 and polycomb group of proteins upon PEG-AuNPs@Hyp/light exposure regulates mitochondria-targeting-mediated GBM metabolism with epigenetic repression of complex machinery function. Polyubiquitination and proteasomal degradation of EZH1 indicate the implication of these polycomb proteins in GBM progression. Chromatin immunoprecipitation reveals the IDH2 and EZH1/EZH2 direct interaction, confirming the role played by IDH2 in modulating the expression of EZH1 and EZH2. In vivo studies further displayed better tumor ablation in a GBM tumor-bearing nude mouse model. The present multimodal nanoformulation compromised the functional dependency of polycomb on mitochondrial IDH2 and established the mechanism of GBM inhibition.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Isocitrato Deshidrogenasa/metabolismo , Nanopartículas del Metal , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Epigénesis Genética , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Oro/metabolismo , Humanos , Ratones , Ratones Desnudos , Mitocondrias/metabolismo , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo
13.
Biomater Adv ; 133: 112602, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35527145

RESUMEN

Parkinson's disease (PD) is the second most neurodegenerative disease caused due to synucleinopathy leads to the death of dopaminergic and serotonergic neurons. The approach to reduce synucleinopathy paves the therapeutic way in PD management. Recent studies highlight anti-Parkinsonism effect of Hytrin that regulates energy homeostasis via activation of mitochondrial redox regulator; IDH2 leading to attenuation of synucleinopathy. However, the burst release kinetics of Hytrin restricts its therapeutic potential. Therefore, we aimed to improve Hytrin release kinetics through nanocarrier mediated delivery, replenish dopamine and serotonin by formulating Hytrin loaded polydopamine serotonin nanohybrid for PD protection. Present study also explores IDH2 mediated neuroprotective action in retardation of synucleinopathy for PD prevention. Nanoformulation has shown effective neurotherapeutic potential by improving Hytrin release profile in the reduction of PD symptoms in vitro and ex vivo. The neuroprotective effect has been attributed to IDH2 induction and alpha-synuclein reduction against rotenone insults. The direct physical interaction of IDH2 and alpha-synuclein, PD hallmark has been uncovered. The study divulges that the restorative effect of our nanoformulation significantly retards the PD deficits byinducing IDH2 mediated alpha-synuclein ubiquitination and proteasomal degradation pathway.


Asunto(s)
Enfermedades Neurodegenerativas , Fármacos Neuroprotectores , Enfermedad de Parkinson , Sinucleinopatías , Dopamina/metabolismo , Humanos , Indoles , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Polímeros , Prazosina/análogos & derivados , Serotonina/uso terapéutico , alfa-Sinucleína/metabolismo
14.
ACS Med Chem Lett ; 13(7): 1109-1117, 2022 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-35859882

RESUMEN

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.

15.
ACS Biomater Sci Eng ; 8(9): 3810-3818, 2022 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-36005299

RESUMEN

The imbalance in the bone remodeling process with more bone resorption by osteoclasts compared to bone formation by osteoblasts results in a metabolic bone disorder known as osteoporosis. This condition reduces the bone mineral density and increases the risk of fractures due to low bone mass and disrupted bone microarchitecture. Osteoclastogenesis increases when the receptor activator NFκB ligand (RANKL) on the osteoblast surface binds to the receptor activator NFκB (RANK) on the osteoclast surface and the function of the decoy receptor of RANKL, osteoprotegrin, is compromised due to external stimuli such as heparin and lipopolysaccharides. The RANK/RANKL axis promotes the nuclear factor kappa B (NFκB) expression, which in turn increases the histone methyltransferase activity of EzH2 and EzH1 for the epigenetic regulation of osteoclastogenesis-related genes. Genistein counteracts NFκB-induced osteoclastogenesis and downstream signaling through the direct regulation of histone methyltransferase, EzH2 and EzH1, transcription. However, genistein possesses limitations like low bioavailability, low water solubility, high estrogen activity, and thyroid side effects, which obstruct its therapeutic usage. Here, the nanoemulsified formulation of genistein with vitamin D was utilized to circumvent the limitations of genistein so that it can be utilized for therapeutic purposes in osteoporosis management. The nanoemulsification of genistein and vitamin D was performed through the spontaneous emulsification using Tween 80 and medium chain triglyceride oil as an organic phase. The physiologically stable and biocompatible combination of the genistein and vitamin D nanoemulsion (GVNE) exhibited the controlled release pattern of genistein with Korsmeyer-Peppas and Higuchi models under different pH conditions (7.4, 6.5, and 1.2). The GVNE potentially enhanced the therapeutic efficacy under in vitro osteoporosis models and helped restore disease parameters like alkaline phosphatase activity, tartrate-resistant acid phosphatase activity, and the formation of multinuclear giant cells. Molecularly, the GVNE overturned the LPS-induced osteoclastogenesis by downregulation of NFκB expression along with its binding on EzH2 and EzH1 promoters. GVNE effects on the osteoporosis model established it as an efficient antiosteoporotic therapy. This nanonutraceutical-based formulation provides an epigenetic regulation of osteoporosis management and opens new avenues for alternate epigenetic therapies for osteoporosis.


Asunto(s)
Genisteína , Osteoporosis , Epigénesis Genética , Genisteína/uso terapéutico , Histona Metiltransferasas/genética , Histona Metiltransferasas/metabolismo , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , FN-kappa B/uso terapéutico , Osteoporosis/tratamiento farmacológico , Osteoporosis/genética , Osteoporosis/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , Ligando RANK/uso terapéutico , Vitamina D/uso terapéutico
16.
J Colloid Interface Sci ; 606(Pt 2): 2024-2037, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-34749449

RESUMEN

The remediation of non-reactive phosphate pollutants in the aquatic system is essential for protecting the ecological niche. In this work, a highly robust protein nanoparticles networked rare-earth metal carbonate-grafted bio-composite membrane (abbreviated as REMC) was fabricated via chemical crosslinking of three-dimensional (3D) hierarchical lanthanum carbonate (mREM) and casein nanoparticles (CsNPs) for selective rejection of non-reactive phosphates. The main components of the REMC membrane are mREM and CsNPs, which were prepared via SDS/CTAB templated homogeneous precipitation and the coacervation/desolvation hybrid method, respectively. The active lanthanum ion (La3+) on the 3D spherulitic surface of mREM exhibited excellent phosphate adsorption capacity (maximum adsorption capacity was 358 mg.g-1) across a wide pH range and in a multi-ionic environment. A series of batch testing and characterizations revealed that the active La3+ and dominating phosphate centers in the REMC membrane framework enable non-enzymatic phosphatase-like activity, cleaving the phosphate ester bond of organic phosphates and releasing free phosphate anions. These released phosphate ions are retained in the REMC membrane via an ion exchange mechanism, where they contribute to improved phosphate removal capacities. Furthermore, CsNPs have a dual function in the membrane, acting as a matrix in the REMC membrane framework and contributing to phosphate ion sequestrations in a synergistic manner. The catalysis of para-nitrophenyl phosphates (pNPP) to paranitrophenol (pNP) in a sequential dephosphorylation by REMC offers an estimate of reaction kinetics and elucidates the underlying mechanism of improved phosphate selectivity in a multi-ionic environment. Furthermore, phosphate specificity, homogeneous binding capacity, reusability, and visual observation of REMC membrane saturation binding direct it's useful economic, industrial applications in aqueous phosphate contaminant removal, which could be beneficial for the active recovery of the aquatic ecosystem.


Asunto(s)
Lantano , Contaminantes Químicos del Agua , Adsorción , Catálisis , Ecosistema , Concentración de Iones de Hidrógeno , Intercambio Iónico , Cinética , Organofosfatos , Fosfatos
17.
Life Sci ; 302: 120655, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35598656

RESUMEN

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.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Melatonina , Animales , Ratones , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/genética , Sulfato de Dextran/toxicidad , Dextranos/metabolismo , Modelos Animales de Enfermedad , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Epigénesis Genética , Enfermedades Inflamatorias del Intestino/patología , Melatonina/farmacología , Melatonina/uso terapéutico , Óxido Nítrico Sintasa de Tipo II/metabolismo
18.
Biomater Adv ; 136: 212796, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35929295

RESUMEN

Inflammatory Bowel (IBD) is an umbrella term which includes Crohn's Disease (CD) and Ulcerative Colitis (UC). At present, therapies available for management of the UC includes, corticosteroid, immuno-suppressants and antibiotics are used for mild to moderate UC conditions which can cause nephrotoxicity, hepatotoxicity and cardiotoxicity. Hence, a novel therapeutic candidate having potent anti-inflammatory effect is urgently warranted for the management of UC. Melatonin has emerged as a potent anti-inflammatory agent. However, poor solubility limits its therapeutic potential. Therefore, colon targeted Eudragit-S-100 coated chitosan nanoparticles have been demonstrated to improve melatonin therapeutic efficacy. It was found that melatonin loaded chitosan and colon targeted chitosan nanoparticles had promising anti-inflammatory efficacy in terms of NO scavenging activity in an in-vitro LPS challenged macrophages. Also, colon targeted oral chitosan nano-formulation exhibited remarkable protection in an in vivo UC mice model by improving gross pathological parameters, histo-architectural protection, goblet cell depletion, and immune cells infiltration which can be extrapolated to clinical studies.


Asunto(s)
Quitosano , Colitis Ulcerosa , Enfermedades Inflamatorias del Intestino , Melatonina , Animales , Antiinflamatorios/farmacología , Quitosano/uso terapéutico , Colitis Ulcerosa/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/inducido químicamente , Melatonina/farmacología , Ratones
19.
Neurochem Res ; 36(8): 1383-96, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21472456

RESUMEN

Malignant neuroblastomas are solid tumors in children. Available therapeutic agents are not highly effective for treatment of malignant neuroblastomas. Therefore, new treatment strategies are urgently needed. We tested the efficacy of combination of SU5416 (SU), an inhibitor of the vascular endothelial growth factor receptor-2 (VEGFR-2), and (-)-epigallocatechin-3-gallate (EGCG), a polyphenolic compound from green tea, for controlling growth of human malignant neuroblastoma SH-SY5Y and SK-N-BE2 cells. Combination of 20 µM SU and 50 µM EGCG synergistically inhibited cell survival, suppressed expression of VEGFR-2, inhibited cell migration, caused cell cycle arrest, and induced apoptosis. Combination of SU and EGCG effectively blocked angiogenic and survival pathways and modulated expression of cell cycle regulators. Apoptosis was induced by down regulation of Bcl-2, activation of caspase-3, and cleavage of the DNA repair enzyme poly(ADP-ribose) polymerase (PARP). Taken together, this combination of drugs can be a promising therapeutic strategy for controlling the growth of human malignant neuroblastoma cells.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticarcinógenos/farmacología , Apoptosis/efectos de los fármacos , Catequina/análogos & derivados , Ciclo Celular/efectos de los fármacos , Indoles/farmacología , Neuroblastoma/patología , Pirroles/farmacología , Catequina/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Factor de Transcripción E2F1/metabolismo , Humanos , Etiquetado Corte-Fin in Situ , Neovascularización Patológica , Neuroblastoma/fisiopatología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína de Retinoblastoma/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
20.
Neurochem Res ; 36(12): 2292-305, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21786169

RESUMEN

Glioblastoma shows poor response to current therapies and warrants new therapeutic strategies. We examined the efficacy of combination of valproic acid (VPA) and taxol (TX) or nanotaxol (NTX) in human glioblastoma LN18 and T98G cell lines. Cell differentiation was manifested in changes in morphological features and biochemical markers. Cell growth was controlled with down regulation of vascular endothelial growth factor (VEGF), epidermal growth factor receptor (EGFR), nuclear factor-kappa B (NF-κB), phospho-Akt (p-Akt), and multi-drug resistance (MDR) marker, indicating suppression of angiogenic, survival, and multi-drug resistance pathways. Cell cycle analysis showed that combination therapy (VPA and TX or NTX) increased the apoptotic sub G1 population and apoptosis was further confirmed by Annexin V-FITC/PI binding assay and scanning electron microscopy. Combination therapy caused activation of caspase-8 and cleavage of Bid to tBid and increased Bax:Bcl-2 ratio and mitochondrial release of cytochrome c and apoptosis-inducing factor (AIF). Upregulation of calpain and caspases (caspase-9 and caspase-3) and substrate degradation were also detected in course of apoptosis. The combination of VPA and NTX most effectively controlled the growth of LN18 and T98G cells. Therefore, this combination of drugs can be used as an effective treatment for controlling growth of human glioblastoma cells.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Paclitaxel/uso terapéutico , Ácido Valproico/uso terapéutico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Paclitaxel Unido a Albúmina , Albúminas/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Factor Inductor de la Apoptosis/metabolismo , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citocromos c/metabolismo , Regulación hacia Abajo , Quimioterapia Combinada , Humanos , Paclitaxel/administración & dosificación , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA