Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem Pharmacol ; : 116326, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38815626

RESUMO

Hepatic urea cycle, previously known as ornithine cycle, is the chief biochemical pathway that deals with the disposal of excessive nitrogen in form of urea, resulted from protein breakdown and concomitant condensation of ammonia. Enzymes involved in urea cycle are expressed differentially outside hepatic tissue and are mostly involved in production of arginine from citrulline in arginine-depleted condition. Inline, cancer cells frequently adapt metabolic rewiring to support sufficient biomass production in order to sustain tumor cell survival, multiplication and subsequent growth. For the accomplishment of this aim, metabolic reprogramming in cancer cells is set in way so that cellular nitrogen and carbon repertoire can be utilized and channelized maximally towards anabolic reactions. A strategy to meet such outcome is to cut down unnecessary catabolic reactions and nitrogen elimination. Thus, transfigured urea cycle is a hallmark of neoplasia. During oncogenesis, altered expression and regulation of enzymes involved in urea cycle is a revolutionary approach meet to maximum incorporation of nitrogen for sustaining tumor specific biogenesis. Currently, we have reviewed neoplasm-specific deregulations of urea cycle-enzymes in different types and stages of cancers suggesting its context-oriented dynamic nature. Considering such insight to be valuable in terms of prospective cancer diagnosis and therapeutics adaptive evolution of deregulated urea cycle has been enlightened.

2.
ACS Appl Bio Mater ; 7(3): 1656-1670, 2024 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-38364267

RESUMO

Triple-negative breast cancer (TNBC) is considered to be one of the most difficult subtypes of breast cancer (BC) to treat. The sheer absence of certain receptors makes it very tough to target, leaving high-dose chemotherapy as probably the sole therapeutic option at the cost of nonspecific toxic effects. Carnosic acid (CA) has been established as a potential chemotherapeutic agent against a range of cancer cells. However, its in vivo chemotherapeutic potential is significantly challenged due to its poor pharmacokinetic attributes. In this study, poly(lactic-co-glycolic) acid (PLGA) nanoparticles (NPs) were formulated to circumvent the biopharmaceutical limitations of CA. CA-loaded polymeric NPs (CA-PLGA NPs) have been evaluated as a potential therapeutic option in the treatment of TNBC. Different in vitro studies exhibited that CA-PLGA NPs significantly provoked oxidative-stress-mediated apoptotic death in MDA-MB-231 cells. The improved anticancer potential of CA-PLGA NPs over CA was found to be associated with improved cellular uptake of the nanoformulation by TNBC cells. In vivo studies also established the improvement in the chemotherapeutic efficacy of CA-nanoformulation over that of free CA without showing any sign of systemic toxicity. Thus, CA-PLGA NPs emerge as a promising candidate to fix two bugs with a single code, resolving biopharmaceutical attributes of CA as well as introducing a treatment option for TNBC.


Assuntos
Abietanos , Produtos Biológicos , Nanopartículas , Neoplasias de Mama Triplo Negativas , Humanos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Disponibilidade Biológica , Polímeros , Produtos Biológicos/uso terapêutico
3.
Cell Signal ; 117: 111089, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38331012

RESUMO

A bunch of complexes harboring vanadium as metal centers have been reported to exhibit a wide array of antineoplastic properties that come under non­platinum metallodrug series and emerge to offer alternative therapeutic strategies from the mechanistic behaviors of platinum-drugs. Though antineoplastic activities of vanado-complexes have been documented against several animal and xenografted human cancers, the definite mechanism of action is yet to unveil. In present study, a novel water soluble 1-methylimidazole substituted mononuclear dipicolinic acid based oxidovanadium (IV) complex (OVMI) has been evaluated for its antineoplastic properties in breast carcinoma both in vitro and in vivo. OVMI has been reported to generate cytotoxicity in human triple negative breast carcinoma cells, MDA-MB-231 as well as in mouse 4T1 cells by priming them for apoptosis. ROS-mediated, mitochondria-dependent as well as ER-stress-evoked apoptotic death seemed to be main operational hub guiding the cytotoxicity of OVMI in vitro. Moreover, OVMI has been noticed to elicit antimetastatic effect in vitro. Therapeutic application of OVMI has been extended on 4T1-based mammary tumor of female Balb/c mice, where it has been found to reduce tumor size, volume and restore general tissue architecture successfully to a great extent. Apart from that, OVMI has been documented to limit 4T1-based secondary pulmonary metastasis along with being non-toxic and biocompatible in vivo.


Assuntos
Antineoplásicos , Carcinoma , Neoplasias de Mama Triplo Negativas , Feminino , Animais , Camundongos , Humanos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Apoptose , Carcinoma/tratamento farmacológico , Água/farmacologia , Linhagem Celular Tumoral , Camundongos Endogâmicos BALB C , Proliferação de Células
7.
Int J Pharm ; 631: 122555, 2023 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-36586636

RESUMO

Cancer is a disease of global importance. In order to mitigate conventional chemotherapy-related side effects, phytochemicals with inherent anticancer efficacy have been opted. However, the use of nanotechnology is essential to enhance the bioavailability and therapeutic efficacy of these phytochemicals. Herein, we have formulated folic acid conjugated polyacrylic acid capped mesoporous silica nanoparticles (∼47.6 nm in diameter) for pH-dependent targeted delivery of chrysin to breast cancer (MCF-7) cells. Chrysin loaded mesoporous silica nanoparticles (Chr- mSiO2@PAA/FA) have been noted to induce apoptosis in MCF-7 cells through oxidative insult and mitochondrial dysfunction with subsequent G1 arrest. Further, in tumor bearing mice, intravenous incorporation of Chr-mSiO2@PAA/FA has been noticed to enhance the anti-neoplastic effects of chrysin via tumor site-specific accumulation. Enhanced cytotoxicity of chrysin contributed towards in vivo tumor regression, restoration of normalized tissue architecture and maintenance of healthy body weight. Besides, no serious systemic toxicity was manifested in response to Chr-mSiO2@PAA/FA administration in vivo. Thus, the study evokes about the anticancer potentiality of chrysin and its increased therapeutic activity via incorporation into folic acid conjugated mesoporous silica nanoparticles, which may hold greater impact in field of future biomedical research.


Assuntos
Nanopartículas , Neoplasias , Camundongos , Animais , Sistemas de Liberação de Medicamentos , Dióxido de Silício , Ácido Fólico , Concentração de Íons de Hidrogênio , Portadores de Fármacos , Porosidade
8.
Biochem Pharmacol ; 207: 115367, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36481348

RESUMO

Often the outer leaflets of living cells bear a coat of glycosylated proteins, which primarily regulates cellular processes. Glycosylation of such proteins occurs as part of their post-translational modification. Within the endoplasmic reticulum, glycosylation enables the attachment of specific oligosaccharide moieties such as, 'glycan' to the transmembrane receptor proteins which confers precise biological information for governing the cell fate. The nature and degree of glycosylation of cell surface receptors are regulated by a bunch of glycosyl transferases and glycosidases which fine-tune attachment or detachment of glycan moieties. In classical death receptors, upregulation of glycosylation by glycosyl transferases is capable of inducing cell death in T cells, tumor cells, etc. Thus, any deregulated alternation at surface glycosylation of these death receptors can result in life-threatening disorder like cancer. In addition, transmembrane glycoproteins and lectin receptors can transduce intracellular signals for cell death execution. Exogenous interaction of lectins with glycan containing death receptors signals for cell death initiation by modulating downstream signalings. Subsequently, endogenous glycan-lectin interplay aids in the customization and implementation of the cell death program. Lastly, the glycan-lectin recognition system dictates the removal of apoptotic cells by sending accurate signals to the extracellular milieu. Since glycosylation has proven to be a biomarker of cellular death and disease progression; glycans serve as specific therapeutic targets of cancers. In this context, we are reviewing the molecular mechanisms of the glycan-lectin regulatory network as an integral part of cell death machinery in cancer to target them for successful therapeutic and clinical approaches.


Assuntos
Lectinas , Neoplasias , Humanos , Glicômica , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Polissacarídeos/metabolismo , Transferases , Receptores de Morte Celular
9.
Life Sci ; 307: 120876, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-35961595

RESUMO

Asiatic acid (AA), an aglycone of pentacyclic triterpene glycoside, obtained from the leaves of Centella asiatica exerts anticancer effects by inhibiting cellular proliferation and inducing apoptosis in a wide range of carcinogenic distresses. However, its chemotherapeutic efficacy is dampened by its low bioavailability. Polymeric nanoparticles (NPs) exhibit therapeutic efficacy and compliance by improving tissue penetration and lowering toxicity. Thus, to increase the therapeutic effectiveness of AA in the treatment of breast cancer, AA-loaded poly lactic-co-glycolic acid (PLGA) NPs (AA-PLGA NPs) have been formulated. The AA-PLGA NPs were characterized on the basis of their average particle size, zeta potential, electron microscopic imaging, drug loading, and entrapment efficiency. The NPs exhibited sustained drug release profile in vitro. Developed NPs exerted dose-dependent cytotoxicity to MCF-7 and MDA-MB-231 cells without damaging normal cells. The pro-oxidant and pro-apoptotic properties of AA-PLGA NPs were determined by the study of the cellular levels of SOD, CAT, GSH-GSSG, MDA, protein carbonylation, ROS, mitochondrial membrane potential, and FACS analyses on MCF-7 cells. Immunoblotting showed that AA-PLGA NPs elicited an intrinsic pathway of apoptosis in MCF-7 cells. In vivo studies on female BALB/c mice exhibited reduced volume of mammary pad tumor tissues and augmented expression of caspase-3 when administered with AA-PLGA NPs. No systemic adverse effect of AA-PLGA NPs was observed in our studies. Thus, AA-PLGA NPs can act as an efficient drug delivery system against breast cancer.


Assuntos
Antineoplásicos/farmacologia , Nanopartículas , Neoplasias , Animais , Caspase 3 , Linhagem Celular Tumoral , Portadores de Fármacos , Feminino , Dissulfeto de Glutationa , Glicolatos , Glicosídeos , Camundongos , Tamanho da Partícula , Triterpenos Pentacíclicos/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Espécies Reativas de Oxigênio , Superóxido Dismutase
11.
Life Sci ; 301: 120606, 2022 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-35508254

RESUMO

Vanadium is a transitional metal having several therapeutic aspects that can be exploited for its anticancer activity. Herein, we have verified anticancer effectivity of synthesized novel water soluble mononuclear dipicolinic acid-1-allyl imidazole-based oxidovanadium (IV) complex [VOL(1-allylimz)2] with respect to anticancer effectivity of known standard platinum-based anticancer agent cisplatin. In current work, we have verified VOL(1-allylimz)2 as highly potential anticancer agent selectively against human breast cancer cells. VOL(1-allylimz)2 has been noticed to elicit dose dependent cytotoxicity in MCF-7 cell line through induction of intracellular oxidative stress and mitochondrial membrane potential. Apart from in vitro validation, in vivo studies in male Swiss Albino mice also have seen to portray dose-dependent anticancer effect of [VOL(1-allylimz)2], where indications of oxidative stress induction became prominent too. Besides, both mitochondrial as well as extra-mitochondrial apoptosis in tumor cells have been shown to be induced by [VOL(1-allylimz)2] treatment, together enforcing its anticancer potency. In contrast to cisplatin, which shows high chances of nephrotoxicity in cancer patients, [VOL(1-allylimz)2] has been found to be comparatively safe for in vivo studies.


Assuntos
Antineoplásicos , Cisplatino , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose , Linhagem Celular Tumoral , Cisplatino/farmacologia , Humanos , Imidazóis/farmacologia , Masculino , Camundongos , Estresse Oxidativo , Vanádio/farmacologia
12.
Photodiagnosis Photodyn Ther ; 39: 102861, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35421600

RESUMO

Here we have synthesized water soluble and biocompatible carbon dots (CDs) from taurine via thermal decomposition method. The CDs showed nearly spherical shape with diameter less than 10 nm. The CDs exhibited excitation dependent fluorescence emission and could be used for mammalian cell imaging. The CDs showed excellent DPPH and hydrogen peroxide radical scavenging activity in cell free system. Besides, the CDs also displayed significant intracellular radical scavenging activity in human normal kidney epithelial (NKE) cells. Furthermore, nanohybrids consisting of both CDs and nanoceria (CeO2) were prepared and tested for their biomedical applications. The nanohybrids showed significant antioxidant activities in both cell free and intracellular conditions. The CDs and nanohybrids possessed very little toxicity upto the concentration of 100 µg/mL when treated for 24 hours in human NKE cells. The CDs as well as nanohybrids further displayed significant bacterial growth inhibition against both gram-positive and gram-negative bacteria under dark as well as light illumination condition via the bacterial membrane damage. However, under the light illumination, the bacterial growth inhibition of CDs and nanohybrids was further enhanced due to the generation of reactive oxygen radicals and subsequent DNA degradation. A higher dose-dependent intracellular antioxidant and antibacterial activities of the nanohybrid is attributed to the synergistic effect of nanoceria and CDs. All these results clearly reflected that our synthesized CDs and their nanohybrids can be used for several biomedical applications.


Assuntos
Carbono , Fotoquimioterapia , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Antioxidantes/química , Antioxidantes/farmacologia , Bactérias Gram-Negativas , Bactérias Gram-Positivas , Humanos , Mamíferos , Fotoquimioterapia/métodos , Taurina
13.
Life Sci ; 298: 120525, 2022 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-35378139

RESUMO

AIMS: Synthesis of novel drug delivery system for targeted delivery of cuminaldehyde to breast cancer cells and the subsequent analyses of anti-neoplastic potential of the drug. MAIN METHODS: 3-carboxy-phenyl boronic acid (PBA) conjugated and polyacrylic acid (PAA) gated mesoporous silica nanoparticles (MSNs) were synthesized for the targeted delivery of cuminaldehyde (CUM) to breast cancer cells. Enhancement of anti-neoplastic effects of cuminaldehyde (4-isopropylbenzaldehyde) by the nanoconjugates was assessed. KEY FINDINGS: The anti-cancer effects of non-targeted and targeted drug-nanoconjugates were examined in vitro and in vivo. The targeted drug-nanoconjugates caused cell cycle arrest and induced the intrinsic pathway of apoptosis in MCF-7 cells through mitochondrial damage. In vivo intravenous injection of the targeted drug-nanoconjugates led to effective reduction in growth of 4 T1 induced mammary pad tumor in female BALB/c mice via augmented accumulation of cuminaldehyde. The drug-nanoconjugates did not exhibit any systemic toxicity. SIGNIFICANCE: Therefore, MSN-PBA-CUM-PAA represents a potent therapeutic model for breast cancer treatment.


Assuntos
Antineoplásicos , Neoplasias da Mama , Nanopartículas , Animais , Antineoplásicos/uso terapêutico , Benzaldeídos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Cimenos , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Camundongos , Nanoconjugados/uso terapêutico , Porosidade , Dióxido de Silício/uso terapêutico
14.
Adv Colloid Interface Sci ; 295: 102495, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34375877

RESUMO

Recently, zinc oxide nanoparticles (ZnONPs) are gaining much interest of nanobiotechnologists due to their profound biomedical applications. ZnONPs are used as antibacterial agents, which cause both gram-positive and negative bacterial cell death through the generation of reactive free radicals as well as membrane rupture. ZnONPs show excellent antioxidant properties in normal mammalian cells via the scavenging of reactive free radicals and up-regulation of antioxidant enzyme activities. Besides, it also shows hypoglycaemic effect in diabetic animals via pancreatic ß-cells mediated increased insulin secretion and glucose uptake by liver, skeletal muscles and adipose tissues. Among the other potential applications, ZnONPs-induced bone and soft-tissue regeneration open a new horizon in the field of tissue engineering. Here, first we reviewed the complete synthesis routes of ZnONPs by physical, chemical, and biological pathways as well as outlined the advantages and disadvantages of the techniques. Further, we discussed the several important aspects of physicochemical analysis of ZnONPs. Additionally, we extensively reviewed the important biomedical applications of ZnONPs as antibacterial, antioxidant, and antidiabetic agents, and in the field of tissue engineering with special emphasis on their mechanisms of actions. Furthermore, the future perspectives of the ZnONPs are also discussed.


Assuntos
Nanopartículas , Óxido de Zinco , Animais , Antibacterianos/farmacologia , Antioxidantes/farmacologia , Hipoglicemiantes/farmacologia , Óxido de Zinco/farmacologia
15.
Mater Sci Eng C Mater Biol Appl ; 126: 112142, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34082953

RESUMO

Herein, we have evaluated the in vivo therapeutic efficacy and systemic toxicity profile of a synthetic anticancer compound [3,3'-((4-(trifluoromethyl)phenyl)methylene)bis(2-hydroxynaphthalene-1,4-dione)]. A multifunctional mesoporous silica nanoparticle (MSN) based drug delivery network was also fabricated which specifically showed targeting nature towards the cancer cell. The mesopores of silica nanoparticles were tagged with phenyl boronic acid (PBA) for targeted drug delivery into tumor tissue. 1j was then loaded inside the nanocarriers followed by pore blocking with gold nanoparticles (GN) to attain a redox-responsive controlled drug delivery pattern. The synthesized nanocarriers (1j@-MSN-PBA-GN) having mean diameter of ~86 nm exhibited a moderate 1j loading content of 13.68% with overall negative surface charge. Both the targeted and non-targeted nanoformulations were tested for their anticancer activities both in vitro and in vivo models, and found more effective as compared with free 1j treatment. However, the targeted nanoformulations showed higher therapeutic effect due to increased cellular internalization and caused mitochondria-dependent apoptosis in MCF-7 cells via oxidative stress. Besides, the targeted nanoformulation significantly inhibited in the development of solid tumor in comparison to non-targeted nanoformulations and free 1j as a consequence of increased internalization of the drug-candidate in tumor tissue. Therefore, this study proposes that 1j can be considered as a potent anti-carcinogenic compound in vivo and its therapeutic potential is further increased by using PBA functionalized and GN gated MSN-based controlled drug delivery system without showing any significant systemic toxicity.


Assuntos
Nanopartículas Metálicas , Nanopartículas , Doxorrubicina , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Ouro , Humanos , Naftoquinonas , Oxirredução , Porosidade , Dióxido de Silício
16.
Mater Sci Eng C Mater Biol Appl ; 116: 111239, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32806268

RESUMO

Herein, a mesoporous silica nanoparticle (MSN) based biocompatible, targeted and controlled drug delivery system has been synthesized for tumor tissue-specific drug delivery. Umbelliferone, a natural coumarin derivative was loaded into the pores of MSN and capped with pH-sensitive poly acrylic acid (PAA). For targeted delivery of umbelliferone in tumor tissue, folic acid (FA) was grafted onto the surface of drug-loaded and PAA-coated MSN. The successful construction of the nanohybrid (Umbe@MSN-PAA-FA) was confirmed by performing a series of characterization. The synthesized pH-responsive nanohybrid showed diameter of around 50 nm with overall negative surface charge and drug loading content of 12.56%. In vitro study showed that the nanohybrid caused significant cytotoxicity through the induction of both oxidative stress as well as mitochondrial damage in folate receptor over-expressed in human breast cancer cell, MCF-7 compared with free umbelliferone. In vivo study also exhibited that the nanohybrid effectively reduced tumor growth in tumor-bearing mice compared with free umbelliferone due to the enlarged bioavailability of the drug in tumor tissue. Besides, the nanohybrid did not exhibit any significant sign of systemic toxicity in other vital organs. Together, the study denoted that PAA and FA functionalized MSN controlled-drug delivery system could assist to increase the anticancer potential of umbelliferone.


Assuntos
Nanopartículas , Neoplasias , Animais , Preparações de Ação Retardada , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Concentração de Íons de Hidrogênio , Camundongos , Porosidade , Dióxido de Silício , Umbeliferonas/farmacologia
17.
Arch Toxicol ; 94(7): 2293-2317, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32524152

RESUMO

The immune system plays a pivotal role in maintaining the defense mechanism against external agents and also internal danger signals. Metabolic programming of immune cells is required for functioning of different subsets of immune cells under different physiological conditions. The field of immunometabolism has gained ground because of its immense importance in coordination and balance of immune responses. Metabolism is very much related with production of energy and certain by-products. Reactive oxygen species (ROS) are generated as one of the by-products of various metabolic pathways. The amount, localization of ROS and redox status determine transcription of genes, and also influences the metabolism of immune cells. This review discusses ROS, metabolism of immune cells at different cellular conditions and sheds some light on how ROS might regulate immunometabolism.


Assuntos
Metabolismo Energético , Sistema Imunitário/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Animais , Humanos , Sistema Imunitário/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Doenças do Sistema Nervoso/imunologia , Doenças do Sistema Nervoso/metabolismo , Neuroimunomodulação , Oxirredução , Espécies Reativas de Oxigênio/imunologia
18.
Biochim Biophys Acta Gen Subj ; 1864(3): 129503, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31816347

RESUMO

BACKGROUND: In recent times, coordination complexes of iron in various oxidation states along with variety of ligand systems have been designed and developed for effective treatment of cancer cells without adversely affecting the normal cell and tissues of various organs. METHODS: In this study, we have evaluated the mechanism of action of a Fe(II) Schiff base complex in the crop plant Trigonella foenum-graecum L. (Fenugreek) as the screening system by using morphological, cytological, biochemical and molecular approaches. Further functional characterization was performed using MCF-7 cell line and solid tumour model for the assessment of anti-tumour activity of the complex. RESULTS: Our results indicate efficiency of the Fe(II) Schiff base complex in the induction of double strand breaks in DNA. Complex treatment clearly induced cytotoxic and genotoxic damage in Trigonella seedlings. The Fe-complex treatment caused cell cycle arrest via the activation of ATM-ATR kinase mediated DNA damage response pathway with the compromised expression of CDK1, CDK2 and CyclinB1 protein in Trigonella seedlings. In cultured MCF-7 cells, the complex induces cytotoxicity and DNA fragmentation through intracellular ROS generation. Fe-complex treatment inhibited tumour growth in solid tumour model with no additional side effects. CONCLUSION: The growth inhibitory and cytotoxic effects of the complex result from activation of DNA damage response along with oxidative stress and cell cycle arrest. GENERAL SIGNIFICANCE: Overall, our results have provided comprehensive information on the mechanism of action and efficacy of a Fe(II) Schiff base complex in higher eukaryotic genomes and indicated its future implications as potential therapeutic agent.


Assuntos
Ferro/metabolismo , Trigonella/metabolismo , Proteína Quinase CDC2/efeitos dos fármacos , Ciclina B1/efeitos dos fármacos , Quinase 2 Dependente de Ciclina/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Compostos Ferrosos/metabolismo , Humanos , Células MCF-7/metabolismo , Oxirredução , Estresse Oxidativo , Bases de Schiff/metabolismo , Trigonella/química
19.
Mater Sci Eng C Mater Biol Appl ; 100: 129-140, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30948047

RESUMO

Naturally occurring bioactive compounds are gaining much importance as anti-tumor agents in recent times due to their high therapeutic potential and less systemic toxicity. However, different preclinical and clinical studies have noted significant shortcomings, such as nonspecific tumor targeting and low bioavailability which limit their usage in therapeutics. Therefore, a safe and compatible nanoparticle mediated controlled drug delivery system is in high demand to enable effective transport of the drug candidates in the tumor tissue. Herein, we have synthesized phenylboronic acid (PBA) conjugated Zinc oxide nanoparticles (PBA-ZnO), loaded with quercetin (a bioflavonoid widely found in plants), with zeta potential around -10.2 mV and diameter below 40 nm. Presence of PBA moieties over the nanoparticle surface facilitates targeted delivery of quercetin to the sialic acid over-expressed cancer cells. Moreover, Quercetin loaded PBA-ZnO nanoparticles (denoted as PBA-ZnO-Q) showed pH responsive drug release behavior. Results suggested that PBA-ZnO-Q induced apoptotic cell death in human breast cancer cells (MCF-7) via enhanced oxidative stress and mitochondrial damage. In line with the in vitro results, PBA-ZnO-Q was found to be effective in reducing tumor growth in EAC tumor bearing mice. Most interestingly, PBA-ZnO-Q is found to reduce tumor associated toxicity in liver, kidney and spleen. The cytotoxic potential of the nanohybrid is attributed to the combinatorial cytotoxic effects of quercetin and ZnO in the cancer cells. Overall, the presented data highlighted the chemotherapeutic potential of the novel nanohybrid, PBA-ZnO-Q which can be considered for clinical cancer treatment.


Assuntos
Portadores de Fármacos/química , Nanopartículas Metálicas/química , Quercetina/química , Óxido de Zinco/química , Animais , Apoptose/efeitos dos fármacos , Ácidos Borônicos/química , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Liberação Controlada de Fármacos , Feminino , Humanos , Concentração de Íons de Hidrogênio , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Quercetina/farmacologia , Quercetina/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Transplante Heterólogo
20.
Ann N Y Acad Sci ; 1443(1): 54-74, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31017675

RESUMO

The role of the evolutionarily conserved Wnt signaling pathway is well documented in several cellular processes, such as cell proliferation, differentiation, cell motility, and maintenance of the stem cell niche. The very first indication that aberrant Wnt signaling can cause carcinogenesis came from a finding that the mutation of the adenomatous polyposis coli gene (APC) predisposes a person to colorectal carcinoma. Later, with progressing research it became clear that abnormal activation or mutation of the genes related to this pathway could drive tumorigenesis. Here, we review recent advances in research regarding Wnt signaling regulation and its role in several cancer subtypes. Additionally, the utility of Wnt pathway-targeted cancer therapy intervention is also highlighted, with an overview of current approaches to target the Wnt pathway in oncogenesis and the future scopes and challenges associated with them.


Assuntos
Neoplasias/terapia , Via de Sinalização Wnt , Animais , Carcinogênese , Humanos , Neoplasias/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA