Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Cell Int ; 24(1): 94, 2024 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-38431613

RESUMO

BACKGROUND: Recent investigations have reported the benefits of using a tyrosine kinase inhibitor, dasatinib (DA), as well as programmed death-ligand 1 (PD-L1) inhibitors in the management of several solid tumors, including breast cancer. Nevertheless, the outcome of the combination of these inhibitors on HER2-positive breast cancer is not explored yet. METHODS: Herein, we investigated the impact of DA and PD-L1 inhibitor (BMS-202) combination on HER2-positive breast cancer cell lines, SKBR3 and ZR75. RESULTS: Our data reveal that the combination significantly inhibits cell viability of both cancer cell lines as compared to monotreatment. Moreover, the combination inhibits epithelial-mesenchymal transition (EMT) progression and reduces cancer cell invasion by restoring E-cadherin and ß-catenin expressions and loss of vimentin, major biomarkers of EMT. Additionally, the combination reduces the colony formation of both cell lines in comparison with their matched control. Also, the combination considerably inhibits the angiogenesis of the chorioallantoic membrane model compared with monotreatment. Molecular pathway analysis of treated cells shows that this combination blocks HER2, AKT, ß-catenin, and JNK1/2/3 activities. CONCLUSION: Our findings implicate that a combination of DA and BMS-202 could have a significant impact on the management of HER2-positive breast cancer.

2.
Biomol Biomed ; 24(3): 575-581, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38158791

RESUMO

Angiogenesis is the process of forming new blood capillaries from pre-existing vessels. Even though it is essential during normal development, it plays a major role in cancer progression. Neratinib is a pan-human epidermal growth factor receptor (HER) inhibitor that has recently been approved for the treatment of HER2-positive breast cancer. However, its effects on angiogenesis and embryogenesis remain unknown. This study examined the antiangiogenic effects of neratinib using the chorioallantoic membrane (CAM) of chicken embryos. We also evaluated neratinib's toxicity during the early stages of normal development using the chicken embryos, primary embryonic fibroblasts (EFBs), and human umbilical vein endothelial cells (HUVEC). Our findings revealed that neratinib significantly inhibited the CAM angiogenesis compared to controls by reducing vessel percentage area and the average vessel length. Furthermore, neratinib downregulated vascular endothelial growth factor (VEGF), a key mediator of angiogenesis. At lower concentrations, neratinib was well-tolerated during early stages of normal development. Additionally, EFBs treated with neratinib showed no morphological or viability changes when compared to controls. However, at the highest concentration tested, neratinib treatment reduced HUVEC cell viability. This effect may be associated with the dysregulation of key apoptotic genes, including caspase-3, caspase-8, caspase-9, and the B-cell lymphoma 2 (Bcl2) gene. Our findings indicate a novel potential application of neratinib as an antiangiogenic agent, exhibiting tolerable toxicity in the early stages of embryogenesis.


Assuntos
Inibidores da Angiogênese , Membrana Corioalantoide , Células Endoteliais da Veia Umbilical Humana , Neovascularização Fisiológica , Quinolinas , Fator A de Crescimento do Endotélio Vascular , Embrião de Galinha , Animais , Humanos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Quinolinas/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Inibidores da Angiogênese/farmacologia , Membrana Corioalantoide/efeitos dos fármacos , Membrana Corioalantoide/irrigação sanguínea , Neovascularização Fisiológica/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Angiogênese
3.
Biomed Pharmacother ; 162: 114676, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37037091

RESUMO

Due to the strong association between diabetes and cancer incidents, several anti-diabetic drugs, including metformin, have been examined for their anticancer activity. Metformin is a biguanide antihyperglycemic agent used as a first-line drug for type II diabetes mellitus. It exhibits anticancer activity by impacting different molecular pathways, such as AMP-inducible protein kinase (AMPK)-dependent and AMPK-independent pathways. Additionally, Metformin indirectly inhibits IGF-1R signaling, which is highly activated in breast malignancy. On the other hand, breast cancer is one of the major causes of cancer-related morbidity and mortality worldwide, where the human epidermal growth factor receptor-positive (HER2-positive) subtype is one of the most aggressive ones with a high rate of lymph node metastasis. In this review, we summarize the association between diabetes and human cancer, listing recent evidence of metformin's anticancer activity. A special focus is dedicated to HER2-positive breast cancer with regards to the interaction between HER2 and IGF-1R. Then, we discuss combination therapy strategies of metformin and other anti-diabetic drugs in HER2-positive breast cancer.


Assuntos
Neoplasias da Mama , Diabetes Mellitus Tipo 2 , Metformina , Humanos , Feminino , Metformina/farmacologia , Neoplasias da Mama/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Proteínas Quinases Ativadas por AMP/metabolismo , Hipoglicemiantes/farmacologia
4.
Front Oncol ; 12: 977664, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36568154

RESUMO

Introduction: Immune checkpoint blockade (ICB)-based therapy is revolutionizing cancer treatment by fostering successful immune surveillance and effector cell responses against various types of cancers. However, patients with HER2+ cancers are yet to benefit from this therapeutic strategy. Precisely, several questions regarding the right combination of drugs, drug modality, and effective dose recommendations pertaining to the use of ICB-based therapy for HER2+ patients remain unanswered. Methods: In this study, we use a mathematical modeling-based approach to quantify the growth inhibition of HER2+ breast cancer (BC) cell colonies (ZR75) when treated with anti-HER2; trastuzumab (TZ) and anti-PD-1/PD-L1 (BMS-202) agents. Results and discussion: Our data show that a combination therapy of TZ and BMS-202 can significantly reduce the viability of ZR75 cells and trigger several morphological changes. The combination decreased the cell's invasiveness along with altering several key pathways, such as Akt/mTor and ErbB2 compared to monotherapy. In addition, BMS-202 causes dose-dependent growth inhibition of HER2+ BC cell colonies alone, while this effect is significantly improved when used in combination with TZ. Based on the in-vitro monoculture experiments conducted, we argue that BMS-202 can cause tumor growth suppression not only by mediating immune response but also by interfering with the growth signaling pathways of HER2+BC. Nevertheless, further studies are imperative to substantiate this argument and to uncover the potential crosstalk between PD-1/PD-L1 inhibitors and HER2 growth signaling pathways in breast cancer.

5.
Int J Mol Sci ; 22(20)2021 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-34681694

RESUMO

Gold nanorods have been implicated in several biomedical applications. Herein, the effect of two surface-modified gold nanorods on the early stages of embryogenesis and angiogenesis was investigated using avian embryos at three days and their chorioallantoic membrane (CAM) at five days of incubation. We found that gold nanorods (GNR) modified with PEGylated phospholipid moiety show a high mortality rate in embryos after four days of exposure compared to GNR modified with PEGylated cholesterol moiety. Meanwhile, our data revealed that surface modified-GNR significantly inhibit the formation of new blood vessels in the treated CAM model after 48 h of exposure. Moreover, we report that surface-modified GNR significantly deregulate the expression of several genes implicated in cell proliferation, invasion, apoptosis, cellular energy metabolism, and angiogenesis. On the other hand, our data point out that GNR treatments can modulate the expression patterns of JNK1/2/3, NF-KB/p38, and MAPK, which could be the main molecular pathways of the nanorods in our experimental models.


Assuntos
Desenvolvimento Embrionário/efeitos dos fármacos , Ouro/química , Nanotubos/toxicidade , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Embrião de Galinha , Galinhas , Membrana Corioalantoide/irrigação sanguínea , Nanotubos/química , Polietilenoglicóis/química , Transdução de Sinais/efeitos dos fármacos , Propriedades de Superfície , Fator de Transcrição RelA/metabolismo
6.
Int J Mol Sci ; 22(17)2021 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-34502529

RESUMO

Natural chalcones possess antitumor properties and play a role as inducers of apoptosis, antioxidants and cytotoxic compounds. We recently reported that novel nitrogen chalcone-based compounds, which were generated in our lab, have specific effects on triple-negative breast cancer cells. However, the outcome of these two new compounds on human epidermal growth factor receptor 2 (HER2)-positive breast cancer remains nascent. Thus, we herein investigated the effects of these compounds (DK-13 and DK-14) on two HER2-positive breast cancer cell lines, SKBR3 and ZR75. Our data revealed that these compounds inhibit cell proliferation, deregulate cell-cycle progression and significantly induce cell apoptosis in both cell lines. Furthermore, the two chalcone compounds cause a significant reduction in the cell invasion ability of SKBR3 and ZR75 cancer cells. In parallel, we found that DK-13 and DK-14 inhibit colony formation of both cell lines in comparison to their matched controls. On the other hand, we noticed that these two compounds can inhibit angiogenesis in the chorioallantoic membrane model. The molecular pathway analysis of chalcone compounds exposed cells revealed that these compounds inhibit the expression of both JNK1/2/3 and ERK1/2, the major plausible molecular pathways behind these events. Our findings implicate that DK-13 and DK-14 possess effective chemotherapeutic outcomes against HER2-positive breast cancer via the ERK1/2 and JNK1/2/3 signaling pathways.


Assuntos
Apoptose/efeitos dos fármacos , Chalconas/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptor ErbB-2/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Chalconas/química , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Nitrogênio/química
7.
Adv Drug Deliv Rev ; 178: 113908, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34390777

RESUMO

Drug delivery systems or vectors are usually needed to improve the bioavailability and effectiveness of a drug through improving its pharmacokinetics/pharmacodynamics at an organ, tissue or cellular level. However, emerging technologies with sensitive readouts as well as a greater understanding of physiological/biological systems have revealed that polymeric drug delivery systems are not biologically inert but can have innate or intrinsic biological actions. In this article, we review the emerging multiple innate biological/toxicological properties of naked polyamidoamine (PAMAM) dendrimer delivery systems in the absence of any drug cargo and discuss their correlation with the defined physicochemical properties of PAMAMs in terms of molecular size (generation), architecture, surface charge and chemistry. Further, we assess whether any of the reported intrinsic biological actions of PAMAMs such as their antimicrobial activity or their ability to sequester glucose and modulate key protein interactions or cell signaling pathways, can be exploited clinically such as in the treatment of diabetes and its complications.


Assuntos
Dendrímeros/metabolismo , Sistemas de Liberação de Fármacos por Nanopartículas/química , Disponibilidade Biológica , Dendrímeros/química , Humanos , Tamanho da Partícula
8.
Comput Struct Biotechnol J ; 19: 2881-2890, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34093999

RESUMO

HER2-positive breast cancer is one of its most challenging subtypes, forming around 15-25% of the total cases. It is characterized by aggressive behavior and treatment resistance. On the other hand, poly (amidoamine) (PAMAM) dendrimers are widely used in drug delivery systems and gene transfection as carriers. PAMAMs can modulate gene expression and interfere with transactivation of the human epidermal growth factor receptor family members (HER1-4). Nevertheless, the outcome of PAMAMs on HER2-positive breast cancer remains unknown. Thus, in this study, we investigated the anti-cancer effects of different generations of PAMAM dendrimers (G4 and G6) and the outcome of their surface chemistries (cationic, neutral, and anionic) on HER2-positive breast cancer cell lines, SKBR3 and ZR75. Our data showed that PAMAM dendrimers, mainly cationic types, significantly reduce cell viability in a dose-dependent manner. More significantly, PAMAMs induce substantial cell apoptosis, accompanied by the up-regulation of apoptotic markers (Bax, Caspases-3, 8 and 9) in addition to down-regulation of Bcl-2. Moreover, our data pointed out that cationic PAMAMs inhibit colony formation compared to controls and other types of PAMAMs. The molecular pathway analysis of PAMAM exposed cells revealed that PAMAMs enhance JNK1/2/3 expression while blocking ERK1/2, in addition to EGFR1 (HER1) and HER2 activities, which could be the major molecular pathway behind these events. These observed effects were comparable to lapatinib treatment, a clinically used inhibitor of HER1 and 2 receptors phosphorylation. Our findings implicate that PAMAMs may possess important therapeutic effects against HER2-positive breast cancer via JNK1/2/3, ERK1/2, and HER1/2 signalling pathways.

9.
Mater Sci Eng C Mater Biol Appl ; 122: 111910, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33641906

RESUMO

Carbon nanofibers (CNFs) have been implicated in biomedical applications, yet, they are still considered as a potential hazard. Conversely, mesoporous silica is a biocompatible compound that has been used in various biomedical applications. In this regard, we recently reported that CNFs induce significant toxicity on the early stage of embryogenesis in addition to the inhibition of its angiogenesis. Thus, we herein use mesoporous silica coating of CNFs (MCNFs) in order to explore their outcome on normal development and angiogenesis using avian embryos at 3 days and its chorioallantoic membrane (CAM) at 6 days of incubation. Our data show that mesoporous silica coating of CNFs significantly reduces embryotoxicity provoked by CNFs. However, MCNFs exhibit slight increase in angiogenesis inhibition in comparison with CNFs. Further investigation revealed that MCNFs slightly deregulate the expression patterns of key controller genes involved in cell proliferation, survival, angiogenesis, and apoptosis as compared to CNFs. We confirmed these data using avian primary normal embryonic fibroblast cells established in our lab. Regarding the molecular pathways, we found that MCNFs downregulate the expression of ERK1/ERK2, p-ERK1/ERK2 and JNK1/JNK2/JNK3, thus indicating a protective role of MCNFs via ERK and JNK pathways. Our data suggest that coating CNFs with a layer of mesoporous silica can overcome their toxicity making them suitable for use in biomedical applications. Nevertheless, further investigations are required to evaluate the effects of MCNFs and their mechanisms using different in vitro and in vivo models.


Assuntos
Nanofibras , Animais , Carbono , Membrana Corioalantoide , Sistema de Sinalização das MAP Quinases , Nanofibras/toxicidade , Dióxido de Silício/toxicidade
10.
Biomed Pharmacother ; 134: 111134, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33341672

RESUMO

Dasatinib is a targeted cancer therapy, while programmed death ligand 1 (PD-L1) inhibitors are a form of immune checkpoint therapy used to treat various types of cancers. Several studies showed the potential efficacy of these drugs in the management of triple-negative breast cancer- an aggressive subtype of breast cancer, which can develop during pregnancy. Nevertheless, side effects of Dasatinib (DA) and PD-L1 drugs during pregnancy, especially in the early stages of embryogenesis are not explored yet. The aim of this study is to assess the individual and combined toxicity of DA and PD-L1 inhibitors during the early stages of embryogenesis and to evaluate their effect(s) on angiogenesis using the chorioallantoic membrane (CAM) model of the embryo. Our results show that embryos die at greater rates after exposure to DA and PD-L1 inhibitors as compared to their matched controls. Moreover, treatment with these drugs significantly inhibits angiogenesis of the CAM. To further elucidate key regulator genes of embryotoxicity induced by the actions of PD-L1 and DA, an RT-PCR analysis was performed for seven target genes that regulate cell proliferation, angiogenesis, and survival (ATF3, FOXA2, MAPRE2, RIPK1, INHBA, SERPINA4, and VEGFC). Our data revealed that these genes are significantly deregulated in the brain, heart, and liver tissues of exposed embryos, compared to matched control tissues. Nevertheless, further studies are necessary to evaluate the effects of these anti breast cancer drugs and elucidate their role during pregnancy.


Assuntos
Acetamidas/toxicidade , Inibidores da Angiogênese/toxicidade , Antígeno B7-H1/antagonistas & inibidores , Membrana Corioalantoide/irrigação sanguínea , Dasatinibe/toxicidade , Inibidores de Checkpoint Imunológico/toxicidade , Neovascularização Fisiológica/efeitos dos fármacos , Inibidores de Proteínas Quinases/toxicidade , Piridinas/toxicidade , Animais , Antígeno B7-H1/metabolismo , Embrião de Galinha , Desenvolvimento Embrionário/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento , Neovascularização Fisiológica/genética , Transdução de Sinais
11.
J Biomed Nanotechnol ; 16(6): 975-984, 2020 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-33187592

RESUMO

Implementation of carbon nanofibers (CNFs) in biomedical applications have successful outcomes, however, they are still considered as a potential hazard. We herein used avian embryos at 3 days and its chorioallantoic membrane (CAM) at 6 days of incubation to evaluate the impact of synthesized CNFs on the early stage of embryogenesis and angiogenesis. Our data point out that 50 µg/embryo concentration of CNFs provoke adverse effects as 75% of CNFs-exposed embryos die within 1-5 days after exposure compared with their matched controls. Furthermore, CNFs significantly inhibit angiogenesis of the CAM after 48-hours post-treatment. Additionally, RT-PCR analysis on seven key controller genes responsible for proliferation, survival, angiogenesis, and apoptosis showed that these genes are deregulated in brain, heart, and liver tissues of CNFs-exposed embryos compared to their matched control. Our investigation suggests that CNFs could have a toxic effect on the early stages of embryogenesis as well as angiogenesis. Nevertheless, further investigations are required to evaluate the effects of CNFs and elucidate their mechanism on the early stage of the normal development and human health.


Assuntos
Nanofibras , Animais , Apoptose , Carbono , Membrana Corioalantoide , Humanos , Nanofibras/toxicidade , Neovascularização Patológica
12.
Toxics ; 8(3)2020 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-32961854

RESUMO

Water-pipe smoking (WPS) is becoming the most popular form of tobacco use among the youth, especially in the Middle East, replacing cigarettes rapidly and becoming a major risk of tobacco addiction worldwide. Smoke from WPS contains similar toxins as those present in cigarette smoke and is linked directly with different types of cancers including lung and head and neck (HN) carcinomas. However, the underlying molecular pathways and/or target genes responsible for the carcinogenic process are still unknown. In this study, human normal oral epithelial (HNOE) cells, NanoString PanCancer Pathways panel of 770 gene transcripts and quantitative real-time polymerase chain reaction (qRT-PCR) analysis were applied to discover differentially expressed genes (DEG) modulated by WPS. In silico analysis was performed to analyze the impact of these genes in HN cancer patient's biology and outcome. We found that WPS can induce the epithelial-mesenchymal transition (EMT: hallmark of cancer progression) of HNOE cells. More significantly, our analysis of NanoString revealed 23 genes deregulated under the effect of WPS, responsible for the modulation of cell cycle, proliferation, migration/invasion, apoptosis, signal transduction, and inflammatory response. Further analysis was performed using qRT-PCR of HNOE WPS-exposed and unexposed cells supported the reliability of our NanoString data. Moreover, we demonstrate those DEG to be upregulated in cancer compared with normal tissue. Using the Kaplan-Meier analysis, we observed a significant association between WPS-deregulated genes and relapse-free survival/overall survival in HN cancer patients. Our findings imply that WPS can modulate EMT as well as a set of genes that are directly involved in human HN carcinogenesis, thereby affecting HN cancer patients' survival.

13.
Molecules ; 25(18)2020 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-32947764

RESUMO

Elaeagnus angustifolia (EA) is a medicinal plant used for treating several human diseases in the Middle East. Meanwhile, the outcome of EA extract on HER2-positive breast cancer remains nascent. Thus, we herein investigated the effects of the aqueous EA extract obtained from the flowers of EA on two HER2-positive breast cancer cell lines, SKBR3 and ZR75-1. Our data revealed that EA extract inhibits cell proliferation and deregulates cell-cycle progression of these two cancer cell lines. EA extract also prevents the progression of epithelial-mesenchymal transition (EMT), an important event for cancer invasion and metastasis; this is accompanied by upregulations of E-cadherin and ß-catenin, in addition to downregulations of vimentin and fascin, which are major markers of EMT. Thus, EA extract causes a drastic decrease in cell invasion ability of SKBR3 and ZR75-1 cancer cells. Additionally, we found that EA extract inhibits colony formation of both cell lines in comparison with their matched control. The molecular pathway analysis of HER2 and JNK1/2/3 of EA extract exposed cells revealed that it can block HER2 and JNK1/2/3 activities, which could be the major molecular pathway behind these events. Our findings implicate that EA extract may possess chemo-preventive effects against HER2-positive breast cancer via HER2 inactivation and specifically JNK1/2/3 signaling pathways.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Elaeagnaceae/química , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Extratos Vegetais/química , Receptor ErbB-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Antineoplásicos Fitogênicos/química , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caderinas/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Elaeagnaceae/metabolismo , Feminino , Flores/química , Flores/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Extratos Vegetais/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Regulação para Cima/efeitos dos fármacos , Vimentina/metabolismo , beta Catenina/metabolismo
14.
Cancers (Basel) ; 12(3)2020 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-32164163

RESUMO

Breast cancer is one of the major causes of mortality in women worldwide. The most aggressive breast cancer subtypes are human epidermal growth factor receptor-positive (HER2+) and triple-negative breast cancers. Therapies targeting HER2 receptors have significantly improved HER2+ breast cancer patient outcomes. However, several recent studies have pointed out the deficiency of existing treatment protocols in combatting disease relapse and improving response rates to treatment. Overriding the inherent actions of the immune system to detect and annihilate cancer via the immune checkpoint pathways is one of the important hallmarks of cancer. Thus, restoration of these pathways by various means of immunomodulation has shown beneficial effects in the management of various types of cancers, including breast. We herein review the recent progress in the management of HER2+ breast cancer via HER2-targeted therapies, and its association with the programmed death receptor-1 (PD-1)/programmed death ligand-1 (PD-L1) axis. In order to link research in the areas of medicine and mathematics and point out specific opportunities for providing efficient theoretical analysis related to HER2+ breast cancer management, we also review mathematical models pertaining to the dynamics of HER2+ breast cancer and immune checkpoint inhibitors.

15.
J Biomed Nanotechnol ; 16(10): 1454-1462, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-33422157

RESUMO

To investigate the impact of poly(amidoamine) dendrimers (PAMAMs) in the embryo, we explored the outcome of different generations (G4 and G6) on the early stages of embryogenesis using the chicken embryo as a model. We also monitored their effect on angiogenesis in the chorioallantoic membrane (CAM). Our data revealed that cationic PAMAMs provoke substantial embryotoxicity, as they significantly induce death (up to 50%, p < 0 05) and inhibit angiogenesis of the CAM (up to 30%, p < 0 05) in a generation-dependent manner in comparison to controls and other types of PAMAMs (anionic and neutral). Moreover, cationic PAMAMs alter the expression of genes related to cell survival, cell cycle, proliferation, transcription factor, apoptosis, and angiogenesis, as shown by RT-PCR analysis. Our data suggest that PAMAM dendrimers exhibit intrinsic toxicity in embryos at the early stages and inhibits angiogenesis of the CAM. Thus, future studies are necessary to illustrate the exact mechanism of PAMAM dendrimers in embryotoxicity.


Assuntos
Dendrímeros , Nanopartículas , Animais , Sobrevivência Celular , Embrião de Galinha , Dendrímeros/toxicidade , Poliaminas/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...