Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell Biochem Funct ; 42(7): e4133, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39390703

RESUMO

The present study is designed to evaluate the nanotherapeutic efficacy of prepared PLGA-loaded Nedaplatin (PLGA-NDP) against 7,12-dimethyl benz(a)anthracene (DMBA)-induced experimental oral carcinogenesis in hamster buccal pouch (HBP) model. The buccal pouch of golden Syrian hamsters was painted with 0.5% DMBA in liquid paraffin three times a week for 14 weeks, ultimately leading to the development of oral squamous cell carcinoma (OSCC). Oral administration of PLGA-NDP (preinitiation) and Cisplatin delivery (5 mg/kg b.wt) started 1 week before the carcinogen exposure and continued on alternative days. Post-administration of PLGA-NDP (5 mg/kg b.wt) started 2 days after carcinogen (DMBA) induction until the end of the experiment. After the 14th week, we observed that DMBA-painted hamsters exhibited tumor formation, morphological alterations, and well-differentiated OSSC in addition to the responsive molecular proteins during oral carcinogenesis. Furthermore, immunoblotting analysis demonstrated that PLGA-NDP inhibits Notch signaling, as evidenced by downregulation of Bcl-Xl, Bcl-2, p21, PGE2, HGF, and CXCL12 proteins, and upregulation of p53 and Bax. This apoptotic response is crucial for PLGA-NDP to induce apoptosis. In addition, RT-PCR results showed that PLGA-NDP nanoparticles play a downregulatory role in the therapeutic action of the notch signaling gene (Notch1, Notch 2, Hes1, Hey1, and Jagged1) at the mRNA transcription level in HBP carcinoma. Taken together, these data indicate that PLGA-NDP is a potent inhibitor of oral carcinogenesis and the expansion of cells that specifically target the Notch signaling pathway indicates that obstructing Notch signaling could potentially serve as a new and innovative therapeutic approach for oral squamous cell carcinoma (OSCC).


Assuntos
9,10-Dimetil-1,2-benzantraceno , Apoptose , Neoplasias Bucais , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Receptores Notch , Transdução de Sinais , Animais , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/patologia , Neoplasias Bucais/induzido quimicamente , Neoplasias Bucais/metabolismo , Apoptose/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Receptores Notch/metabolismo , Masculino , Carcinogênese/efeitos dos fármacos , Carcinogênese/induzido quimicamente , Mesocricetus , Cricetinae , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/metabolismo , Ácido Poliglicólico/química , Modelos Animais de Doenças , Ácido Láctico/química , Ácido Láctico/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos/química , Compostos Organoplatínicos
2.
Crit Rev Microbiol ; 47(2): 224-239, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33476522

RESUMO

An emerging body of research is revealing the microbiota pivotal involvement in determining the health or disease state of several human niches, and that of vitamin D also in extra-skeletal regions. Nevertheless, much of the oral microbiota and vitamin D reciprocal impact in oropharyngeal squamous cell carcinogenesis (OPSCC) is still mostly unknown. On this premise, starting from an in-depth scientific bibliographic analysis, this narrative literature review aims to show a detailed view of the state of the art on their contribution in the pathogenesis of this cancer type. Significant differences in the oral microbiota species quantity and quality have been detected in OPSCC-affected patients; in particular, mainly high-risk human papillomaviruses (HR-HPVs), Fusobacterium nucleatum, Porphyromonas gingivalis, Pseudomonas aeruginosa, and Candida spp. seem to be highly represented. Vitamin D prevents and fights infections promoted by the above identified pathogens, thus confirming its homeostatic function on the microbiota balance. However, its antimicrobial and antitumoral actions, well-described for the gut, have not been fully documented for the oropharynx yet. Deeper investigations of the mechanisms that link vitamin D levels, oral microbial diversity and inflammatory processes will lead to a better definition of OPSCC risk factors for the optimization of specific prevention and treatment strategies.


Assuntos
Bactérias/efeitos dos fármacos , Microbiota , Boca/microbiologia , Neoplasias Orofaríngeas/microbiologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/microbiologia , Vitamina D/farmacologia , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Carcinogênese/efeitos dos fármacos , Humanos , Neoplasias Orofaríngeas/patologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia
3.
Environ Toxicol ; 36(3): 339-351, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33068063

RESUMO

Oral cancer is a multifactorial cancer that affects millions of peoples worldwide. The current exploration aimed to evaluate the mechanisms that thymoquinone nanoencapsulated carrier and its effects on 7,12-Dimethylbenz[a]anthracene (DMBA) stimulated hamster buccal pouch cancer in Syrian hamster model. Nanocarrier was characterized by SEM, TEM, FTIR analysis. The incidence of tumor, and biochemicals makers was studied through standard methods. The mRNA expression level of inflammatory markers NF-κBp50, NF-κBp65, and PI3K/AKT/mTOR markers in the buccal tissues of control and experimental animals were investigated through RT-PCR analysis. In thymoquinone (TQ) loaded calcium alginate and polyvinyl alcohol carrier (TQ/Ca-alg-PVA) no squamous cell carcinogenesis developed and others moderate dysplasia revealed differentiated form of hyperplasia and keratosis. In biochemical analyses with DMBA + TQ/Ca-alg-PVA (20 mg/kg bw) orally administered hamsters showed restored the antioxidants, detoxification, xenobiotic metabolising enzymes in DMBA induced plasma and oral tissues of hamsters. Further, mRNA expression level of NF-κBp50/p65 and PI3K/AKT/mTOR were upregulated in the DMBA alone painted hamster. In contrast, these expressions were down regulated in orally TQ/Ca-alg-PVA treated experimental animals. This ability more eligible to deregulate the inflammatory and PI3K/AKT/mTOR signaling pathway that proved it suppresses anti-invasion/metastasis activity during hamster buccal pouch carcinogenesis. From this study, we recommended that TQ/Ca-alg-PVA has documented as effective chemopreventive agents, in further many molecular machineries need to study.


Assuntos
9,10-Dimetil-1,2-benzantraceno/toxicidade , Benzoquinonas/farmacologia , Álcool de Polivinil/farmacologia , 9,10-Dimetil-1,2-benzantraceno/efeitos adversos , Alginatos/efeitos adversos , Animais , Antracenos/efeitos adversos , Anticarcinógenos/farmacologia , Carcinogênese/efeitos dos fármacos , Carcinógenos , Carcinoma de Células Escamosas/patologia , Bochecha/patologia , Cricetinae , Regulação para Baixo/efeitos dos fármacos , Masculino , Mesocricetus , Neoplasias Bucais/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Álcool de Polivinil/metabolismo , Álcool de Polivinil/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
4.
Molecules ; 26(16)2021 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-34443443

RESUMO

Cytostatic chemotherapeutics provide a classical means to treat cancer, but conventional treatments have not increased in efficacy in the past years, warranting a search for new approaches to therapy. The aim of the study was, therefore, to obtain methacrylic acid (MAA) (co)polymers and to study their immunopharmacological properties. 4-Cyano-4-[(dodecylsulfanylthiocarbonyl)sulfanyl] pentanoic acid (CDSPA) and 2-cyano-2-propyl dodecyl trithiocarbonate (CPDT) were used as reversible chain transfer agents. Experiments were carried out in Wistar rats. The MTT assay was used to evaluate the cytotoxic effect of the polymeric systems on peritoneal macrophages. An experimental tumor model was obtained by grafting RMK-1 breast cancer cells. Serum cytokine levels of tumor-bearing rats were analyzed. The chain transfer agents employed in classical radical polymerization substantially reduced the molecular weight of the resulting polymers, but a narrow molecular weight distribution was achieved only with CDSPA and high CPDT concentrations. Toxicity was not observed when incubating peritoneal macrophages with polymeric systems. In tumor-bearing rats, the IL-10 concentration was 1.7 times higher and the IL-17 concentration was less than half that of intact rats. Polymeric systems decreased the IL-10 concentration and normalized the IL-17 concentration in tumor-bearing rats. The maximum effect was observed for a MAA homopolymer with a high molecular weight. The anion-active polymers proposed as carrier constituents are promising for further studies and designs of carrier constituents of drug derivatives.


Assuntos
Antineoplásicos/imunologia , Antineoplásicos/farmacologia , Portadores de Fármacos/química , Ácidos Polimetacrílicos/farmacologia , Animais , Antineoplásicos/administração & dosagem , Carcinogênese/efeitos dos fármacos , Carcinogênese/patologia , Citocinas/metabolismo , Feminino , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Peso Molecular , Ácidos Polimetacrílicos/administração & dosagem , Ratos Wistar
5.
Mol Pharm ; 17(3): 738-747, 2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-31904241

RESUMO

Our previous study proved that celastrol was a potential candidate for hepatocellular carcinoma (HCC) therapy. However, poor water solubility and toxic side effects may restrict its clinical application. To overcome these shortcomings and optimize its antitumor efficacy, we developed galactosylated liposomes using galactose-modified 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-poly(ethylene glycol) to deliver celastrol (C-GPL). C-GPL improved the water solubility of celastrol and exhibited high encapsulation efficiency, good stability in serum, and slow drug release profile. In vitro studies showed that C-GPL increased the cellular uptake of celastrol through receptor-mediated endocytosis, thereby enhancing celastrol cytotoxicity and cancer cell apoptosis. Particularly, in vivo antitumor activity of C-GPL was assessed in rapid HCC mouse models established via hydrodynamic transfection of the activated forms of AKT and c-Met. Compared to free celastrol, C-GPL significantly prevented liver weight gain, decreased liver damage biomarkers (glutamic-oxalacetic transaminase and alanine aminotransferase) and HCC marker (alpha-fetoprotein), and led to tumor disappearance on the liver surface. The improved therapeutic effect of C-GPL may be attributed to suppression of AKT activation, induction of apoptosis, and retardation of cell proliferation. Importantly, C-GPL exerted low toxicity to normal tissues without causing severe weight loss in mice. Taken together, C-GPL may become a promising drug delivery system for HCC treatment.


Assuntos
Antineoplásicos/administração & dosagem , Carcinogênese/efeitos dos fármacos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Lipossomos/administração & dosagem , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Triterpenos Pentacíclicos/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Carcinogênese/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Modelos Animais de Doenças , Liberação Controlada de Fármacos , Galactose/química , Células Hep G2 , Humanos , Lipossomos/química , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Camundongos , Tamanho da Partícula , Fosfatidiletanolaminas/química , Polietilenoglicóis/química , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-met/genética , Solubilidade , Transfecção , Resultado do Tratamento
6.
Oral Dis ; 25(3): 652-669, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29656422

RESUMO

The oral cavity harbors different taxonomic groups, the evolutionary coexistence of which develops the oral ecosystem. These resident microorganisms can alter the balance between the physiologic and pathologic conditions that affect the host, both locally and systemically. This highly sophisticated nature of the oral cavity poses a significant therapeutic challenge. Numerous human and animal studies have been conducted to potentiate the efficacy and competence of current treatments of pathologic conditions as well as to develop novel therapeutic modalities. One of these studies is the use of the potent antimicrobial agent lactoferrin (LF), which was originally derived from the host immune system. LF is an 80-kDa glycoprotein that has a free iron sequestration mechanism with evident antimicrobial, anti-tumor, and immunomodulatory properties. A wide range of active peptides have been isolated from the N-terminal region of LF, which possess antimicrobial activities. In this review, we discuss the role of LF and LF-derived peptides under a heterogeneous group of oral and maxillofacial conditions, including bacterial, fungal, viral infections; head and neck cancers; xerostomia; and implantology-bone-related manifestations.


Assuntos
Bactérias/efeitos dos fármacos , Lactoferrina/farmacologia , Neoplasias Bucais/prevenção & controle , Peptídeos/farmacologia , Doenças Periodontais/microbiologia , Animais , Candida albicans/efeitos dos fármacos , Carcinogênese/efeitos dos fármacos , Cárie Dentária/microbiologia , Cárie Dentária/prevenção & controle , Humanos , Lactoferrina/genética , Lactoferrina/uso terapêutico , Peptídeos/uso terapêutico , Polimorfismo de Nucleotídeo Único , Streptococcus mutans/efeitos dos fármacos , Fenômenos Fisiológicos Virais/efeitos dos fármacos
7.
Biomacromolecules ; 17(11): 3750-3760, 2016 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-27723964

RESUMO

Toward the goal of establishing physiologically relevant in vitro tumor models, we synthesized and characterized a biomimetic hydrogel using thiolated hyaluronic acid (HA-SH) and an acrylated copolymer carrying multiple copies of cell adhesive peptide (PolyRGD-AC). PolyRGD-AC was derived from a random copolymer of tert-butyl methacrylate (tBMA) and oligomeric (ethylene glycol) methacrylate (OEGMA), synthesized via atom transfer radical polymerization (ATRP). Acid hydrolysis of tert-butyl moieties revealed the carboxylates, through which acrylate groups were installed. Partial modification of the acrylate groups with a cysteine-containing RGD peptide generated PolyRGD-AC. When PolyRGD-AC was mixed with HA-SH under physiological conditions, a macroscopic hydrogel with an average elastic modulus of 630 Pa was produced. LNCaP prostate cancer cells encapsulated in HA-PolyRGD gels as dispersed single cells formed multicellular tumoroids by day 4 and reached an average diameter of ∼95 µm by day 28. Cells in these structures were viable, formed cell-cell contacts through E-cadherin (E-CAD), and displayed cortical organization of F-actin. Compared with the control gels prepared using PolyRDG, multivalent presentation of the RGD signal in the HA matrix increased cellular metabolism, promoted the development of larger tumoroids, and enhanced the expression of E-CAD and integrins. Overall, hydrogels with multivalently immobilized RGD are a promising 3D culture platform for dissecting principles of tumorigenesis and for screening anticancer drugs.


Assuntos
Carcinogênese/efeitos dos fármacos , Hidrogéis/química , Peptídeos/química , Polímeros/química , Biomimética , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Hidrogéis/síntese química , Hidrogéis/farmacologia , Masculino , Metacrilatos/síntese química , Metacrilatos/química , Metacrilatos/farmacologia , Peptídeos/síntese química , Peptídeos/farmacologia , Polímeros/síntese química , Polímeros/farmacologia , Neoplasias da Próstata/induzido quimicamente , Neoplasias da Próstata/patologia
8.
Int J Cancer ; 137(9): 2270-3, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25912754

RESUMO

Polyethylene glycol (PEG) is one of the most powerful agents in reducing chemically induced carcinogenesis in rat colon. However, contrasting results in Min mice dampened the enthusiasm on this potentially strong and virtually safe, cancer chemopreventing agent. Pirc (F344/NTac-Apc (am1137) ) rats carrying a germline heterozygous mutation in the Apc gene, spontaneously develop multiple tumours in the colon thus modelling both familial adenomatous polyposis (FAP) and sporadic colorectal cancer (CRC). Given this similarity, we thought that these rats could be appropriate to test the efficacy of PEG 8000 in reducing carcinogenesis. Pirc male rats aged one month were treated with 5% PEG in drinking water for 2 or 6 months. Precancerous lesions were dramatically reduced after 2 months of PEG treatment (Mucin depleted foci (MDF)/colon were 99 ± 17 and 12 ± 8 in Controls and PEG-treated rats, respectively; p < 0.001; mean ± SD). Similarly, colon tumors were significantly reduced after 6 months of treatment (tumors/rat were 8.1 ± 2.3 and 3.6 ± 2.2 in Controls and PEG-treated rats, respectively; p < 0.05; mean ± SD). Colon proliferation, a parameter correlated to cancer risk, was also significantly lower in PEG-treated rats than in Controls, while apoptosis was not significantly affected. In conclusion, PEG markedly reduces colon carcinogenesis in Pirc rats mutated in Apc; we thus suggest that PEG may be used as chemopreventive agent to reduce cancer risk in FAP and CRC patients.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Anticarcinógenos/farmacologia , Carcinogênese/genética , Neoplasias do Colo/genética , Polietilenoglicóis/farmacologia , Animais , Carcinogênese/efeitos dos fármacos , Laxantes/farmacologia , Masculino , Mutação , Ratos Endogâmicos F344
9.
Int J Cancer ; 137(11): 2545-57, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26076896

RESUMO

Molecular crosstalk between cancer cells and fibroblasts has been an emerging hot issue in understanding carcinogenesis. As oral submucous fibrosis (OSF) is an inflammatory fibrotic disease that can potentially transform into squamous cell carcinoma, OSF has been considered to be an appropriate model for studying the role of fibroblasts during early stage carcinogenesis. In this sense, this study aims at investigating whether areca nut (AN)-exposed fibroblasts cause DNA damage of epithelial cells. For this study, immortalized hNOF (hTERT-hNOF) was used. We found that the levels of GRO-α, IL-6 and IL-8 increased in AN-exposed fibroblasts. Cytokine secretion was reduced by antioxidants in AN-exposed fibroblasts. Increase in DNA double strand breaks (DSB) and 8-oxoG FITC-conjugate was observed in immortalized human oral keratinocytes (IHOK) after the treatment of cytokines or a conditioned medium derived from AN-exposed fibroblasts. Cytokine expression and DNA damage were also detected in OSF tissues. The DNA damage was reduced by neutralizing cytokines or antioxidant treatment. Generation of reactive oxygen species (ROS) and DNA damage response, triggered by cytokines, were abolished when NADPH oxidase (NOX) 1 and 4 were silenced in IHOK, indicating that cytokine-triggered DNA damage was caused by ROS generation through NOX1 and NOX4. Taken together, this study provided strong evidence that blocking ROS generation might be a rewarding approach for cancer prevention and intervention in OSF.


Assuntos
Areca/efeitos adversos , Dano ao DNA/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Gengiva/efeitos dos fármacos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Queratinócitos/efeitos dos fármacos , Antioxidantes/farmacologia , Carcinogênese/efeitos dos fármacos , Carcinogênese/metabolismo , Carcinogênese/patologia , Células Cultivadas , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Gengiva/metabolismo , Gengiva/patologia , Células HEK293 , Humanos , Queratinócitos/metabolismo , Queratinócitos/patologia , Mucosa Bucal/efeitos dos fármacos , Mucosa Bucal/metabolismo , Mucosa Bucal/patologia , NADPH Oxidase 1 , NADPH Oxidase 4 , NADPH Oxidases/metabolismo , Nozes/efeitos adversos , Fibrose Oral Submucosa/metabolismo , Fibrose Oral Submucosa/patologia , Espécies Reativas de Oxigênio/metabolismo
10.
ACS Appl Bio Mater ; 7(5): 3388-3402, 2024 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-38660938

RESUMO

In the present investigation, natural bone-derived hydroxyapatite (HA, 2 wt %) and/or exfoliated graphene (Gr, 0.1 wt %)-embedded polydimethylsiloxane (PDMS) elastomeric films were prepared using a vascular method. The morphology, mechanical properties, crystallinity, and chemical structure of the composite films were evaluated. The in vitro biodegradation kinetics of the films indicates their adequate physiological stability. Most of the results favored PDMS/HA/Gr as a best composite scaffold having more than 703% elongation. A simulation study of the microfluidic vascular channel of the PDMS/HA/Gr scaffold suggests that the pressure drop at the outlet became greater (from 1.19 to 0.067 Pa) unlike velocity output (from 0.071 to 0.089 m/s), suggesting a turbulence-free laminar flow. Our bioactive scaffold material, PDMS/HA/Gr, showed highest cytotoxicity toward the lung cancer and breast cancer cells through Runx3 protein-mediated cytotoxic T lymphocyte (CTL) generation. Our data and predicted mechanism also suggested that the PDMS/HA/Gr-supported peripheral blood mononuclear cells (PBMCs) not only increased the generation of CTL but also upregulated the expression of RUNX3. Since the PDMS/HA/Gr scaffold-supported Runx3 induced CTL generation caused maximum cell cytotoxicity of breast cancer (MCF-7) and lung cancer (A549) cells, PDMS/HA/Gr can be treated as an excellent potential candidate for CTL-mediated cancer therapy.


Assuntos
Materiais Biocompatíveis , Dimetilpolisiloxanos , Durapatita , Grafite , Teste de Materiais , Nanocompostos , Alicerces Teciduais , Durapatita/química , Durapatita/farmacologia , Grafite/química , Grafite/farmacologia , Humanos , Dimetilpolisiloxanos/química , Nanocompostos/química , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Alicerces Teciduais/química , Tamanho da Partícula , Carcinogênese/efeitos dos fármacos , Antineoplásicos/farmacologia , Antineoplásicos/química , Sobrevivência Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos
11.
Sci Rep ; 11(1): 20672, 2021 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-34667240

RESUMO

Peri-implantitis may result in the loss of dental implants. Cold atmospheric pressure plasma (CAP) was suggested to promote re-osseointegration, decrease antimicrobial burden, and support wound healing. However, the long-term risk assessment of CAP treatment in the oral cavity has not been addressed. Treatment with two different CAP devices was compared against UV radiation, carcinogen administration, and untreated conditions over 12 months. Histological analysis of 406 animals revealed that repeated CAP exposure did not foster non-invasive lesions or squamous cell carcinoma (SCCs). Carcinogen administration promoted non-invasive lesions and SCCs. Molecular analysis by a qPCR screening of 144 transcripts revealed distinct inflammatory profiles associated with each treatment regimen. Interestingly, CAP treatment of carcinogen-challenged mucosa did not promote but instead left unchanged or reduced the proportion of non-invasive lesions and SCC formation. In conclusion, repeated CAP exposure of murine oral mucosa was well tolerated, and carcinogenic effects did not occur, motivating CAP applications in patients for dental and implant treatments in the future.


Assuntos
Carcinogênese/efeitos dos fármacos , Carcinógenos/administração & dosagem , Mucosa Bucal/efeitos dos fármacos , Gases em Plasma/administração & dosagem , Animais , Antibacterianos/farmacologia , Pressão Atmosférica , Implantes Dentários/efeitos adversos , Inflamação/induzido quimicamente , Masculino , Camundongos , Osseointegração/efeitos dos fármacos , Peri-Implantite/induzido quimicamente , Propriedades de Superfície/efeitos dos fármacos , Cicatrização/efeitos dos fármacos
12.
Laryngoscope ; 130(11): E593-E597, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-31808958

RESUMO

OBJECTIVE: Glucosylceramide (Glu-Cer), a glycosylated form of ceramide, has been reported to have cytotoxic effects in the cells of various cancers. We previously reported that dietary Glu-Cer from rice bran had inhibitory effects on human head and neck squamous cell carcinoma (HNSCC) in nonobese diabetes (NOD)/severe combined immunodeficiency (SCID) mice. In HNSCC, preventing recurrence and second primary cancer is required to improve prognosis. The purpose of the present study was to determine whether dietary Glu-Cer had anticarcinogenic and antitumorigenic effects in a mouse model of HNSCC. METHODS: A total of 40 CB6F1-Tg rasH2@Jcl mice were divided into two groups: control and Glu-Cer. All mice were given 4-nitroquinoline 1-oxide for 24 weeks. Control group mice were fed the normal diet without Glu-Cer. The Glu-Cer group mice were given a mixture of the normal diet plus 0.25% Glu-Cer for 24 weeks. Microscopic examination was performed to identify grossly visible preneoplasms and neoplasms in the mouth, pharynx, and esophagus. Epithelial regions were classified as normal tissue, carcinoma in situ (CIS), or SCC; and the number of each type of region was counted. RESULTS: Compared with the Glu-Cer group mice, control group mice more frequently developed individual and multiple tumors of each type, including CIS and SCC, in the mouth, pharynx, or esophagus. CONCLUSION: Tumor development was effectively inhibited by dietary Glu-Cer derived from rice bran, indicating that this and related compounds show promise as prophylactic agents for human HNSCC. LEVEL OF EVIDENCE: NA Laryngoscope, 130:E593-E597, 2020.


Assuntos
Carcinogênese/efeitos dos fármacos , Dieta/métodos , Carboidratos da Dieta/administração & dosagem , Glucosilceramidas/administração & dosagem , Neoplasias de Cabeça e Pescoço/prevenção & controle , Ração Animal/análise , Animais , Carcinógenos , Modelos Animais de Doenças , Neoplasias de Cabeça e Pescoço/induzido quimicamente , Camundongos
13.
Sci Rep ; 10(1): 2935, 2020 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-32076068

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignancies. TGF-ß is strongly expressed in both the epithelial and stromal compartments of PDAC, and dysregulation of TGF-ß signalling is a frequent molecular disturbance in PDAC progression and metastasis. In this study, we investigated whether blockade of TGF-ß signalling synergizes with nal-IRI/5-FU/LV, a chemotherapy regimen for malignant pancreatic cancer, in an orthotopic pancreatic tumour mouse model. Compared to nal-IRI/5-FU/LV treatment, combining nal-IRI/5-FU/LV with vactosertib, a TGF-ß signalling inhibitor, significantly improved long-term survival rates and effectively suppressed invasion to surrounding tissues. Through RNA-sequencing analysis, we identified that the combination treatment results in robust abrogation of tumour-promoting gene signatures and positive enrichment of tumour-suppressing and apoptotic gene signatures. Particularly, the expression of tumour-suppressing gene Ccdc80 was induced by vactosertib and further induced by vactosertib in combination with nal-IRI/5-FU/LV. Ectopic expression of CCDC80 suppressed migration and colony formation concomitant with decreased expression of epithelial-to-mesenchymal transition (EMT) markers in pancreatic cancer cells. Collectively, these results indicate that combination treatment of vactosertib with nal-IRI/5-FU/LV improves overall survival rates in a mouse model of pancreatic cancer by suppressing invasion through CCDC80. Therefore, combination therapy of nal-IRI/5-FU/LV with vactosertib could provide clinical benefits to pancreatic cancer patients.


Assuntos
Fluoruracila/uso terapêutico , Irinotecano/uso terapêutico , Leucovorina/uso terapêutico , Nanopartículas/química , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Compostos de Anilina/farmacologia , Compostos de Anilina/uso terapêutico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinogênese/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Modelos Animais de Doenças , Sinergismo Farmacológico , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Fluoruracila/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Irinotecano/farmacologia , Leucovorina/farmacologia , Lipossomos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Análise de Sobrevida , Transcriptoma/genética , Triazóis/farmacologia , Triazóis/uso terapêutico , Ensaio Tumoral de Célula-Tronco , Regulação para Cima/efeitos dos fármacos
14.
J Mater Chem B ; 7(37): 5677-5687, 2019 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-31475273

RESUMO

Poly(phosphoester)-based biomaterials have great potential in drug delivery systems (DDSs) because of their multifunctional adjustability, stealth effect, excellent biodegradability, and biocompatibility. To further increase the drug loading efficiency (DLE) and sustained release ability, a multi-arm block copolymer, poly(amido amine)-b-poly(2-butenyl phospholane)-b-poly(2-methoxy phospholane) conjugated with folic acid (abbreviated as PAMAM-PBEP-PMP-FA), was designed and prepared. Compared to the traditional linear copolymers, this multi-arm phosphoester block copolymer integrates a balanced combination of unique features. As an advanced DDS, the PAMAM-PBEP-PMP-FA based supramolecular micelle provides good architectural stability, low protein adsorption, extremely high DLE, and sustained drug release for chemotherapy and abundant surface chemistry for target engineering. Benefitting from these novel functions, the supramolecular micellar drug delivery system exhibits great performances both in in vitro and in vivo evaluations. Doxorubicin (DOX)-loaded supramolecular micelles PAMAM-PBEP-PMP-FA/DOX are fast taken up by HepG2 cells and inhibit the tumor growth effectively in HepG2-tumor-bearing nude mice without obvious system toxicity. This work not only suggests a targeted sustained release DDS for effective chemotherapy but also enlightens, through a delicate design at the molecular scale, the brilliance of multifunctional PPE-based nanomaterials towards versatile bio-applications.


Assuntos
Carcinogênese/efeitos dos fármacos , Preparações de Ação Retardada/química , Neoplasias/tratamento farmacológico , Animais , Sobrevivência Celular , Doxorrubicina/administração & dosagem , Células Hep G2 , Humanos , Camundongos , Camundongos Nus , Micelas , Polímeros/química , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Eur Rev Med Pharmacol Sci ; 23(1): 397-401, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30657582

RESUMO

OBJECTIVE: Metronidazole is an antibiotic widely used in different medical conditions such as trichomoniasis, amoebiasis, and giardiasis among others. Its use has been associated with toxicity; however, it is not well characterized. In this review, we discuss the different therapeutic uses of metronidazole and its side effects in order to aid future investigation in this field. MATERIALS AND METHODS: Relevant information, original research articles, clinical trials, and reviews were collected from PubMed to know the state of the art of the different therapeutic uses of metronidazole and the reported side effects. RESULTS: Metronidazole was used by the first time in 1959, to treat an infection caused by Trichomonas vaginalis; subsequently, new therapeutic properties were discovered. Nowadays, Metronidazole is used to treat infections caused by Bacteroides, Fusobacteria and Clostridia, rosacea, oral and dental infections, bone and joint infections, gynecologic infections, endocarditis, septicemia, and respiratory tract infections. It also can be used to treat Crohn´s disease or even like prophylaxis, before surgical procedures. Metronidazole is well tolerated with mild to moderate side effects such as nausea, abdominal pain, and diarrhea. Nevertheless, serious neurotoxicity, optic neuropathy, peripheral neuropathy, and encephalopathy have been reported in rare cases. Their genotoxic effects observed in animal models are controversial in humans. CONCLUSIONS: The therapeutic use of metronidazole had increased worldwide. Even though it is widely used, metronidazole has been associated with neurotoxicity and genotoxicity; however, its side effects are not well established. Conversely, its veterinary use is restricted in some countries because of its tumor association. Subsequently, further studies are needed to discover the secure use of metronidazole and describe new usages for this drug.


Assuntos
Antiprotozoários/efeitos adversos , Metronidazol/efeitos adversos , Síndromes Neurotóxicas/etiologia , Infecções Protozoárias em Animais/tratamento farmacológico , Infecções por Protozoários/tratamento farmacológico , Animais , Carcinogênese/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Modelos Animais de Doenças , Uso de Medicamentos/estatística & dados numéricos , Humanos , Legislação Veterinária , Drogas Veterinárias/efeitos adversos
16.
Int J Nanomedicine ; 14: 7107-7121, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31564868

RESUMO

BACKGROUND: Cervical cancer (CxCa) ranks as the fourth most prevalent women-related cancer worldwide. Therefore, there is a crucial need to develop newer treatment modalities. Ormeloxifene (ORM) is a non-steroidal, selective estrogen receptor modulator (SERM) that is used as an oral contraceptive in humans. Recent investigations suggest that ORM exhibits potent anti-cancer activity against various types of cancers. Nanoparticulates offer targeted delivery of anti-cancer drugs with minimal toxicity and promise newer approaches for cancer diagnosis and treatment. Therefore, the nanotherapy approach is superior compared to traditional chemotherapy, which is not site-specific and is often associated with various side effects. METHODS: Pursuing this novel nanotherapy approach, our lab has recently developed ORM-loaded poly [lactic-co-glycolic acid] (PLGA), an FDA-approved biodegradable polymer, nanoparticles to achieve targeted drug delivery and improved bioavailability. Our optimized PLGA-ORM nanoformulation showed improved internalization in both dose- and energy-dependent manners, through endocytosis-mediated pathways in both Caski and SiHa cell lines. Additionally, we employed MTS and colony forming assays to determine the short- and long-term effects of PLGA-ORM on these cells. RESULTS: Our results showed that this formulation demonstrated improved inhibition of cellular proliferation and clonogenic potential compared to free ORM. Furthermore, the PLGA-ORM nanoformulation exhibited superior anti-tumor activities in an orthotopic cervical cancer mouse model than free ORM. CONCLUSION: Collectively, our findings suggest that our novel nanoformulation has great potential for repurposing the drug and becoming a novel modality for CxCa management.


Assuntos
Benzopiranos/uso terapêutico , Nanopartículas/uso terapêutico , Neoplasias do Colo do Útero/tratamento farmacológico , Animais , Benzopiranos/farmacologia , Carcinogênese/efeitos dos fármacos , Carcinogênese/metabolismo , Carcinogênese/patologia , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Clonais , Modelos Animais de Doenças , Endocitose/efeitos dos fármacos , Eritrócitos/metabolismo , Feminino , Hemólise/efeitos dos fármacos , Humanos , Teste de Materiais , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Nus , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Soro/química , Neoplasias do Colo do Útero/patologia
17.
JCI Insight ; 4(20)2019 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-31619584

RESUMO

Lung cancer remains the leading cause of cancer-related death in the United States. Although the alveolar macrophage (AM) comprises the major resident immune cell in the lung, few studies have investigated its role in lung cancer development. We recently discovered a potentially novel mechanism wherein AMs regulate STAT-induced inflammatory responses in neighboring epithelial cells (ECs) via secretion and delivery of suppressors of cytokine signaling 3 (SOCS3) within extracellular vesicles (EVs). Here, we explored the impact of SOCS3 transfer on EC tumorigenesis and the integrity of AM SOCS3 secretion during development of lung cancer. AM-derived EVs containing SOCS3 inhibited STAT3 activation as well as proliferation and survival of lung adenocarcinoma cells. Levels of secreted SOCS3 were diminished in lungs of patients with non-small cell lung cancer and in a mouse model of lung cancer, and the impaired ability of murine AMs to secrete SOCS3 within EVs preceded the development of lung tumors. Loss of this homeostatic brake on tumorigenesis prompted our effort to "rescue" it. Provision of recombinant SOCS3 loaded within synthetic liposomes inhibited proliferation and survival of lung adenocarcinoma cells in vitro as well as malignant transformation of normal ECs. Intratumoral injection of SOCS3 liposomes attenuated tumor growth in a lung cancer xenograft model. This work identifies AM-derived vesicular SOCS3 as an endogenous antitumor mechanism that is disrupted within the tumor microenvironment and whose rescue by synthetic liposomes can be leveraged as a potential therapeutic strategy for lung cancer.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/imunologia , Neoplasias Pulmonares/imunologia , Macrófagos Alveolares/imunologia , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Células A549 , Células Epiteliais Alveolares/citologia , Células Epiteliais Alveolares/metabolismo , Animais , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Carcinogênese/efeitos dos fármacos , Carcinogênese/imunologia , Carcinogênese/patologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Feminino , Humanos , Injeções Intralesionais , Lipossomos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Macrófagos Alveolares/citologia , Macrófagos Alveolares/metabolismo , Camundongos , Cultura Primária de Células , Ratos , Proteínas Recombinantes/administração & dosagem , Mucosa Respiratória/citologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Fator de Transcrição STAT3/imunologia , Fator de Transcrição STAT3/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/administração & dosagem , Proteína 3 Supressora da Sinalização de Citocinas/genética , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Sci Rep ; 9(1): 8272, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31164688

RESUMO

Smokeless tobacco associated Gingivobuccal squamous cell carcinoma (GB-SCC) is a major public health problem but available oral cancer cell lines are mostly from smoking associated tongue SCC raising the need for pertinent GB-SCC cell line models. As part of the International Cancer Genome Consortium (ICGC) Project, 4 novel cell lines, namely, Indian Tata Memorial Centre Oral Cancer (ITOC) -01 to -04 were established and characterized with conventional methods, karyotyping, ultrastructure, in vivo tumourigenicity, Whole exome sequencing (WES) and RNA sequencing. These hyperploid cell lines form xenografts in mice and show metabolically active and necrotic areas on fluorodeoxyglucose-positron emission tomography (FDG-PET) imaging. WES of ITOC cell lines recapitulate the genomic tumor profile of ICGC GB-SCC database. We further identified smokeless tobacco associated genetic alterations (PCLO, FAT3 and SYNE2) and oncogenic PIK3CA mutation in GB-SCC cell lines. Transcriptome profiling identified deregulation of pathways commonly altered in cancer and down-regulation of arachidonic acid metabolism pathway, implying its possible role in GB-SCC. Clinical application of high throughput sequencing data depends on relevant cell line models to validate potential targets. Extensively characterized, these oral SCC cell lines are particularly suited for mechanistic studies and pre-clinical drug development for smokeless tobacco associated oral cancer.


Assuntos
Carcinoma de Células Escamosas/genética , Classe I de Fosfatidilinositol 3-Quinases/genética , Doenças da Gengiva/genética , Neoplasias Bucais/genética , Animais , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genômica , Doenças da Gengiva/induzido quimicamente , Doenças da Gengiva/patologia , Xenoenxertos , Humanos , Camundongos , Mucosa Bucal/efeitos dos fármacos , Mucosa Bucal/patologia , Neoplasias Bucais/induzido quimicamente , Neoplasias Bucais/patologia , Mutação/genética , Tabaco sem Fumaça/efeitos adversos , Sequenciamento do Exoma
19.
Trends Cancer ; 4(4): 281-291, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29606313

RESUMO

Resistance to chemotherapy and pathway-targeted therapies poses a major problem in cancer research. While the fields of tumor biology and experimental therapeutics have already benefited from ex vivo preclinical tissue models, these models have yet to address the reasons for malignant transformations and the emergence of chemoresistance. With the increasing number of ex vivo models poised to incorporate physiological biophysical properties, along with the advent of genomic sequencing information, there are now unprecedented opportunities to better understand tumorigenesis and to design therapeutic approaches to overcome resistance. Here we discuss that new preclinical ex vivo models should consider - in addition to common biophysical parameters such as matrix stiffness and bioadhesivity - a more comprehensive milieu of tissue signaling, nuclear mechanics, immune response, and the gut microbiome.


Assuntos
Antineoplásicos/farmacologia , Materiais Biocompatíveis , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Neoplasias/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Adesividade , Antineoplásicos/uso terapêutico , Fenômenos Biofísicos , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinogênese/patologia , Desenho de Fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/patologia , Humanos , Modelos Biológicos , Neoplasias/genética , Neoplasias/patologia , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral/genética
20.
Sci Rep ; 8(1): 5873, 2018 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-29651001

RESUMO

Consumption of green tea (GT) extracts or purified catechins has shown the ability to prevent oral and other cancers and inhibit cancer progression in rodent models, but the evidence for this in humans is mixed. Working with humans, we sought to understand the source of variable responses to GT by examining its effects on oral epithelium. Lingual epithelial RNA and lingual and gingival microbiota were measured before and after 4 weeks of exposure in tobacco smokers, whom are at high risk of oral cancer. GT consumption had on average inconsistent effects on miRNA expression in the oral epithelium. Only analysis that examined paired miRNAs, showing changed and coordinated expression with GT exposure, provided evidence for a GT effect on miRNAs, identifying miRNAs co-expressed with two hubs, miR-181a-5p and 301a-3p. An examination of the microbiome on cancer prone lingual mucosa, in contrast, showed clear shifts in the relative abundance of Streptococcus and Staphylococcus, and other genera after GT exposure. These data support the idea that tea consumption can consistently change oral bacteria in humans, which may affect carcinogenesis, but argue that GT effects on oral epithelial miRNA expression in humans vary between individuals.


Assuntos
Antioxidantes/administração & dosagem , MicroRNAs/genética , Mucosa Bucal/efeitos dos fármacos , Neoplasias Bucais/prevenção & controle , Chá/química , Adulto , Antioxidantes/química , Carcinogênese/efeitos dos fármacos , Catequina/administração & dosagem , Epitélio/efeitos dos fármacos , Epitélio/microbiologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Gengiva/efeitos dos fármacos , Gengiva/microbiologia , Humanos , Masculino , MicroRNAs/efeitos dos fármacos , Microbiota/efeitos dos fármacos , Microbiota/genética , Pessoa de Meia-Idade , Mucosa Bucal/microbiologia , Neoplasias Bucais/genética , Neoplasias Bucais/microbiologia , Fumantes , Staphylococcus/efeitos dos fármacos , Staphylococcus/patogenicidade , Streptococcus/efeitos dos fármacos , Streptococcus/patogenicidade , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA