Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
BMC Complement Med Ther ; 23(1): 301, 2023 Aug 25.
Article in English | MEDLINE | ID: mdl-37626388

ABSTRACT

BACKGROUND: Açaí, a Brazilian native fruit, has already been demonstrated to play a role in the progress of breast cancer and cardiotoxicity promoted by chemotherapy agents. Thus, the present study aimed to evaluate the combined use of açaí and the FAC-D chemotherapy protocol in a breast cancer model in vivo. METHODS: Mammary carcinogenesis was induced in thirty female Wistar rats by subcutaneous injection of 25 mg/kg 7,12-dimethylbenzanthracene (DMBA) in the mammary gland. After sixty days, the rats were randomized into two groups: treated with 200 mg/kg of either açaí extract or vehicle, via gastric tube for 45 consecutive days. The FAC-D protocol was initiated after 90 days of induction by intraperitoneal injection for 3 cycles with a 7-day break each. After treatment, blood was collected for haematological and biochemical analyses, and tumours were collected for macroscopic and histological analyses. In the same way, heart, liver, and kidney samples were also collected for macroscopic and histological analyses. RESULTS: Breast cancer was found as a cystic mass with a fibrotic pattern in the mammary gland. The histological analysis showed an invasive carcinoma area in both groups; however, in the saline group, there was a higher presence of inflammatory clusters. No difference was observed regarding body weight, glycaemia, aspartate aminotransferase (AST), alanine aminotransferase (ALT), creatinine, and urea in either group. However, açaí treatment decreased creatine kinase (CK), creatine kinase MB (CKMB), troponin I and C-reactive protein levels and increased the number of neutrophils and monocytes. Heart histopathology showed normal myocardium in the açaí treatment, while the saline group presented higher toxicity effects with loss of architecture of cardiac tissue. Furthermore, the açaí treatment presented greater collagen distribution, increased hydroxyproline concentration and lower H2AX immunostaining in the heart samples. CONCLUSION: Açaí decreased the number of inflammatory cells in the tumor environment and exhibited protection against chemotherapy drug cardiotoxicity with an increased immune response in animals. Thus, açaí can be considered a promising low-cost therapeutic treatment that can be used in association with chemotherapy agents to avoid heart damage.


Subject(s)
Euterpe , Neoplasms , Female , Animals , Rats , Rats, Wistar , Cardiotoxicity , Heart , Creatine Kinase
2.
Pharmaceuticals (Basel) ; 15(11)2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36422544

ABSTRACT

BACKGROUND: Current drugs for the treatment of endometriosis are not able to completely cure the condition, and significant side effects hinder the continuation of treatment. Therefore, it is necessary to search for new drug candidates. In the present paper, the use of plant extracts is highlighted. Babassu oil and Copaiba oil resin have several therapeutic properties. We investigated the in vitro effects of two nanoemulsions containing oil extracted from Babassu (Orbignya speciosa) nuts (called SNEDDS-18) and/or oil resin extracted from Copaiba trunk (Copaifera langsdorffii) (called SNEDDS-18/COPA) on cultured human eutopic endometrium stromal cells from endometrial biopsies of patients without (CESC) and with (EuESC) endometriosis as well as human stromal cells from biopsies of endometriotic lesions (EctESC). METHODS: CESC, EuESC, and EctESC were taken and treated with SNEDDS-18 and SNEDDS-18/COPA to evaluate their effects on cytotoxicity, cell morphology, proliferation, and signaling pathways. RESULTS: After 48 h of incubation with SNEDDS-18 and SNEDDS-18/COPA, cell viability and proliferation were inhibited, especially in EctESC. The lowest concentration of both nanoemulsions reduced cell viability and proliferation and broke down the cytoskeleton in EctESCs. After 24 h of treatment a decrease in IL-1, TNF-α, and MCP-1 was observed, as well as an increase in IL-10 production. CONCLUSIONS: Both nanoemulsions can affect endometriotic stromal cell behaviors, thus revealing two potential candidates for new phytotherapeutic agents for the management of endometriosis.

3.
Cell Mol Life Sci ; 79(2): 116, 2022 Feb 03.
Article in English | MEDLINE | ID: mdl-35113247

ABSTRACT

Esophageal cancer (EC) is one of the most incident and lethal tumors worldwide. Although surgical resection is an important approach in EC treatment, late diagnosis, metastasis and recurrence after surgery have led to the management of adjuvant and neoadjuvant therapies over the past few decades. In this scenario, 5-fluorouracil (5-FU) and cisplatin (CISP), and more recently paclitaxel (PTX) and carboplatin (CBP), have been traditionally used in EC treatment. However, chemoresistance to these agents along EC therapeutic management represents the main obstacle to successfully treat this malignancy. In this sense, despite the fact that most of chemotherapy drugs were discovered several decades ago, in many cases, including EC, they still represent the most affordable and widely employed treatment approach for these tumors. Therefore, this review summarizes the main mechanisms through which the response to the most widely chemotherapeutic agents used in EC treatment is impaired, such as drug metabolism, apoptosis resistance, cancer stem cells (CSCs), cell cycle, autophagy, energetic metabolism deregulation, tumor microenvironment and epigenetic modifications.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Drug Resistance, Neoplasm/genetics , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Mutation , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Carboplatin/therapeutic use , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Esophageal Neoplasms/metabolism , Fluorouracil/therapeutic use , Humans , Molecular Targeted Therapy/methods , Paclitaxel/therapeutic use , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
4.
Cancers (Basel) ; 13(8)2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33920762

ABSTRACT

Head and neck squamous cell carcinomas (HNSCC) are among the most common and lethal tumors worldwide, occurring mostly in oral cavity, pharynx, and larynx tissues. The squamous epithelia homeostasis is supported by the extracellular matrix (ECM), and alterations in this compartment are crucial for cancer development and progression. Laminin is a fundamental component of ECM, where it represents one of the main components of basement membrane (BM), and data supporting its contribution to HNSCC genesis and progression has been vastly explored in oral cavity squamous cell carcinoma. Laminin subtypes 111 (LN-111) and 332 (LN-332) are the main isoforms associated with malignant transformation, contributing to proliferation, adhesion, migration, invasion, and metastasis, due to its involvement in the regulation of several pathways associated with HNSCC carcinogenesis, including the activation of the EGFR/MAPK signaling pathway. Therefore, it draws attention to the possibility that laminin may represent a convergence point in HNSCC natural history, and an attractive potential therapeutic target for these tumors.

5.
Mol Cell Endocrinol ; 528: 111230, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33675864

ABSTRACT

Despite advances in treatment of lethal prostate cancer, the incidence of prostate cancer brain metastases is increasing. In this sense, we analyzed the molecular profile, as well as the functional consequences involved in the reciprocal interactions between prostate tumor cells and human astrocytes. We observed that the DU145 cells, but not the LNCaP cells or the RWPE-1 cells, exhibited more pronounced, malignant and invasive phenotypes along their interactions with astrocytes. Moreover, global gene expression analysis revealed several genes that were differently expressed in our co-culture models with the overexpression of GLIPR1 and SPARC potentially representing a molecular signature associated with the invasion of central nervous system by prostate malignant cells. Further, these results were corroborated by immunohistochemistry and in silico analysis. Thus, we conjecture that the data here presented may increase the knowledge about the molecular mechanisms associated with the invasion of CNS by prostate malignant cells.


Subject(s)
Brain Neoplasms/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Osteonectin/genetics , Osteonectin/metabolism , Prostatic Neoplasms/genetics , A549 Cells , Animals , Astrocytes/chemistry , Astrocytes/cytology , Brain Neoplasms/metabolism , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Prostatic Neoplasms/metabolism , Up-Regulation
6.
Nat Prod Res ; 35(24): 6034-6039, 2021 Dec.
Article in English | MEDLINE | ID: mdl-32924590

ABSTRACT

Leaves of Kalanchoe pinnata are used worldwide for healing skin wounds. This study aimed to develop and compare two creams containing a leaf aqueous extract of K. pinnata (KP; 6%) and its major flavonoid [quercetin 3-O-α-L-arabinopyranosyl-(1→2)-α-L-rhamnopyranoside] (0.15%). Both creams were topically evaluated in a rat excision model for 15 days. On the 12th day, groups treated with KP leaf-extract and KP major flavonoid creams exhibited 95.3 ± 1.2% and 97.5 ± 0.8% of healing, respectively (positive control = 96.7 ± 0.8%; negative control = 76.1 ± 3.8%). Both resulted in better re-epithelialization and denser collagen fibres. Flavonol glycosides are the main phenolics in KP leaf-extract according to HPLC-ESI-MS/MS analysis. KP major flavonoid plays a fundamental role in the wound healing. The similar results found for both creams indicate that the use of KP crude extract should be more profitable than the isolated compound.


Subject(s)
Kalanchoe , Animals , Flavonoids , Plant Extracts , Plant Leaves , Rats , Tandem Mass Spectrometry , Wound Healing
7.
Cell Mol Life Sci ; 78(3): 817-831, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32920697

ABSTRACT

The high mobility group A (HMGA) proteins are found to be aberrantly expressed in several tumors. Studies (in vitro and in vivo) have shown that HMGA protein overexpression has a causative role in carcinogenesis process. HMGA proteins regulate cell cycle progression through distinct mechanisms which strongly influence its normal dynamics along malignant transformation. Tumor protein p53 (TP53) is the most frequently altered gene in cancer. The loss of its activity is recognized as the fall of a barrier that enables neoplastic transformation. Among the different functions, TP53 signaling pathway is tightly involved in control of cell cycle, with cell cycle arrest being the main biological outcome observed upon p53 activation, which prevents accumulation of damaged DNA, as well as genomic instability. Therefore, the interaction and opposing effects of HMGA and p53 proteins on regulation of cell cycle in normal and tumor cells are discussed in this review. HMGA proteins and p53 may reciprocally regulate the expression and/or activity of each other, leading to the counteraction of their regulation mechanisms at different stages of the cell cycle. The existence of a functional crosstalk between these proteins in the control of cell cycle could open the possibility of targeting HMGA and p53 in combination with other therapeutic strategies, particularly those that target cell cycle regulation, to improve the management and prognosis of cancer patients.


Subject(s)
Cell Cycle Checkpoints/physiology , HMGA Proteins/metabolism , Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , DNA Damage , Disease Progression , Gene Expression Regulation, Neoplastic , Genomic Instability , HMGA Proteins/genetics , Humans , Neoplasms/genetics , Neoplasms/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics
8.
Reproduction ; 159(6): 779-786, 2020 06.
Article in English | MEDLINE | ID: mdl-32240980

ABSTRACT

This study aimed to analyse the effects of clotrimazole (CTZ) on estrogen production pathway in endometriosis progression. Experimental endometriosis was induced by autologous transplantation in female Wistar rats, and then the rats were treated with clotrimazole (200 mg/kg) or vehicle, both orally and intraperitoneally, for 15 consecutive days. Serum estrogen levels and vaginal smear analyses were performed and ERα (estrogen receptor alpha) and CYP19 (cytochrome P450 aromatase) levels in the endometriotic lesions were analysed morphologically and immunohistochemically. The clotrimazole group presented a reduction in serum estrogen levels, which were not influenced by the estrous cycle of the animals. The expression of ERα and CYP19 in endometriotic lesions was also reduced in the clotrimazole group compared to the control group. Moreover, clotrimazole treatment decreased the size of the lesions, as confirmed by histological examination, which showed glandular atrophy for both routes of administration. These results suggest that clotrimazole interferes with the estrogen production pathway by downregulating CYP19 and, therefore, reducing serum estrogen levels. Thus, the drug decreases endometriotic lesion size and consequently disease progression.


Subject(s)
Aromatase/metabolism , Clotrimazole/therapeutic use , Down-Regulation/drug effects , Endometriosis/drug therapy , Endometrium/drug effects , Estrogens/blood , Animals , Clotrimazole/pharmacology , Disease Models, Animal , Endometriosis/metabolism , Endometrium/metabolism , Estrogen Receptor alpha/metabolism , Estrous Cycle/drug effects , Female , Rats , Rats, Wistar
9.
An Acad Bras Cienc ; 92(1): e20190336, 2020.
Article in English | MEDLINE | ID: mdl-32321026

ABSTRACT

Uncaria tomentosa (UT) extracts have been shown to have promising anti-tumor activity. We hypothesized that its incorporation into nanostructured systems could improve the anticancer properties. Here, poly-e-caprolactone (PCL) and poly-d,l-lactide-co-glycolide (PLGA) were employed to generate nanoparticles loaded with UT extract in a single emulsion solvent evaporation method. The nanoparticles were characterized by particle size, zeta potential, morphology and entrapment efficiency along with stability and release profiles. The nanoparticles presented entrapment efficiencies above 60% and a mean diameter below 300nm. UT-PCL nanoparticles presented higher entrapment efficiency and mean particle size as well as a slow release rate. The UT-PLGA nanoparticles showed higher drug loading. Two prostate cancer cell-lines, LNCaP and DU145 that were derived from metastatic sites, served as model systems to assess cytotoxicity and anti-cancer activity. In vitro, both formulations reduced the viability of DU145 and LNCaP cells. Yet, the UT-PLGA nanoparticles showed higher cytotoxicity towards DU145 cells while the UTPCL against LNCaP cells. The results confirm that the incorporation of UT into nanoparticles could enhance its anti-cancer activities that can offer a viable alternative for the treatment of prostrate canner and highlights the potential of nanostructured systems to provide a promising methodology to enhance the activity of natural extracts.


Subject(s)
Antineoplastic Agents/pharmacology , Cat's Claw/chemistry , Plant Extracts/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers , Humans , Nanoparticles
10.
Cells ; 9(2)2020 02 17.
Article in English | MEDLINE | ID: mdl-32079295

ABSTRACT

In the last years, the extracellular matrix (ECM) has been reported as playing a relevant role in esophageal cancer (EC) development, with this compartment being related to several aspects of EC genesis and progression. This sounds very interesting due to the complexity of this highly incident and lethal tumor, which takes the sixth position in mortality among all tumor types worldwide. The well-established increase in ECM stiffness, which is able to trigger mechanotransduction signaling, is capable of regulating several malignant behaviors by converting alteration in ECM mechanics into cytoplasmatic biochemical signals. In this sense, it has been shown that some molecules play a key role in these events, particularly the different collagen isoforms, as well as enzymes related to its turnover, such as lysyl oxidase (LOX) and matrix metalloproteinases (MMPs). In fact, MMPs are not only involved in ECM stiffness, but also in other events related to ECM homeostasis, which includes ECM remodeling. Therefore, the crucial role of distinct MMPs isoform has already been reported, especially MMP-2, -3, -7, and -9, along EC development, thus strongly associating these proteins with the control of important cellular events during tumor progression, particularly in the process of invasion during metastasis establishment. In addition, by distinct mechanisms, a vast diversity of glycoproteins and proteoglycans, such as laminin, fibronectin, tenascin C, galectin, dermatan sulfate, and hyaluronic acid exert remarkable effects in esophageal malignant cells due to the activation of oncogenic signaling pathways mainly involved in cytoskeleton alterations during adhesion and migration processes. Finally, the wide spectrum of interactions potentially mediated by ECM may represent a singular intervention scenario in esophageal carcinogenesis natural history and, due to the scarce knowledge on the cellular and molecular mechanisms involved in EC development, the growing body of evidence on ECM's role along esophageal carcinogenesis might provide a solid base to improve its management in the future.


Subject(s)
Carcinogenesis/pathology , Esophageal Neoplasms/embryology , Extracellular Matrix/metabolism , Matrix Metalloproteinases/metabolism , Proteoglycans/metabolism , Humans
11.
Biomed Res Int ; 2019: 2059516, 2019.
Article in English | MEDLINE | ID: mdl-31737655

ABSTRACT

GI tumors represent a heterogeneous group of neoplasms concerning their natural history and molecular alterations harbored. Nevertheless, these tumors share very high incidence and mortality rates worldwide and patients' poor prognosis. Therefore, the identification of specific biomarkers could increase the development of personalized medicine, in order to improve GI cancer management. In this sense, HMGA family members (HMGA1 and HMGA2) comprise an important group of genes involved in the genesis and progression of malignant tumors. Additionally, it has also been reported that HMGA1 and HMGA2 display an important role in the detection and progression of GI tumors. In this way, HMGA family members could be used as reliable biomarkers able to efficiently track not only the tumor per se but also the main risk conditions related with their development of GI cancers in the future. Finally, it shall be a promising option to revert the current scenario, once HMGA genes and proteins could represent a convergence point in the complex landscape of GI tumors.


Subject(s)
Gastrointestinal Neoplasms/metabolism , HMGA Proteins/metabolism , Animals , Biomarkers, Tumor/metabolism , Disease Progression , Gastrointestinal Neoplasms/pathology , Humans , Prognosis
12.
Life Sci ; 239: 117048, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31730867

ABSTRACT

Benign prostatic hyperplasia (BPH) is an aging-related and progressive disease linked to an up-regulation of α1-adrenoceptors. The participation of EGF receptors (EGFR) in the GPCRs' signalosome has been described but so far data about the contribution of these receptors to prostatic stromal hyperplasia are scanty. We isolated and cultured vimentin-positive prostate stromal cells obtained from BPH patients. According to intracellular Ca2+ measurements, cell proliferation and Western blotting assays, these cultured hyperplastic stromal cells express functional α1-adrenoceptors and EGFR, and proliferate in response to the α1-adrenoceptor agonist phenylephrine. Interestingly, in these cells the inhibition of EGFR signaling with GM6001, CRM197, AG1478 or PD98059 was associated with full blockage of α1-adrenoceptor-mediated cell proliferation, while cell treatment with each inhibitor alone did not alter basal cell growth. Moreover, the co-incubation of AG1478 (EGFR inhibitor) with α1A/α1D-adrenoceptor antagonists showed no additive inhibitory effect. These findings highlight a putative role of EGFR signaling to α1-adrenoceptor-mediated human prostate hyperplasia, suggesting that the inhibition of this transactivation cascade could be useful to reduce BPH progression.


Subject(s)
ErbB Receptors/metabolism , Prostatic Hyperplasia/metabolism , Receptors, Adrenergic, alpha-1/physiology , Adrenergic alpha-1 Receptor Antagonists/metabolism , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Cell Line , Cell Proliferation/drug effects , ErbB Receptors/antagonists & inhibitors , Humans , Hyperplasia/metabolism , Male , Piperazines/pharmacology , Primary Cell Culture , Prostate/drug effects , Protein Kinase Inhibitors/pharmacology , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction/drug effects , Stromal Cells
13.
Genes (Basel) ; 10(5)2019 05 15.
Article in English | MEDLINE | ID: mdl-31096664

ABSTRACT

Endometrioid endometrial carcinomas (EEC) are the most common malignant gynecologic tumors. Despite the increase in EEC molecular knowledge, the identification of new biomarkers involved in disease's development and/or progression would represent an improvement in its course. High-mobility group A protein (HMGA) family members are frequently overexpressed in a wide range of malignancies, correlating with a poor prognosis. Thus, the aim of this study was to analyze HMGA1 and HMGA2 expression pattern and their potential role as EEC biomarkers. HMGA1 and HMGA2 expression was initially evaluated in a series of 46 EEC tumors (stages IA to IV), and the findings were then validated in The Cancer Genome Atlas (TCGA) EEC cohort, comprising 381 EEC tumors (stages IA to IV). Our results reveal that HMGA1 and HMGA2 mRNA and protein are overexpressed in ECC, but only HMGA1 expression is associated with increased histological grade and tumor size. Moreover, HMGA1 but not HMGA2 overexpression was identified as a negative prognostic factor to EEC patients. Finally, a positive correlation between expression of HMGA1 pseudogenes-HMGA1-P6 and HMGA1-P7-and HMGA1 itself was detected, suggesting HMGA1 pseudogenes may play a role in HMGA1 expression regulation in EEC. Thus, these results indicate that HMGA1 overexpression possesses a potential role as a prognostic biomarker for EEC.


Subject(s)
Carcinoma, Endometrioid/genetics , Endometrial Neoplasms/genetics , HMGA1a Protein/genetics , HMGA2 Protein/genetics , Adult , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Carcinoma, Endometrioid/metabolism , Endometrial Neoplasms/metabolism , Female , HMGA1a Protein/biosynthesis , HMGA2 Protein/biosynthesis , Humans , Middle Aged , Prognosis , Transcriptome
14.
Article in English | MEDLINE | ID: mdl-30719063

ABSTRACT

Benign Prostatic Hyperplasia (BPH) affects mainly older men. It is estimated to affect 50% of 51-60-year-old men and 70% of 61-70-year-old men. BPH is a nonmalignant proliferation of epithelial and stromal cells of the prostate gland regions. Despite the use of conventional pharmacological therapy, herbal medicines are used in BPH therapy, and several mechanisms of action have been suggested based on their complex chemical composition. Considering the ethnomedicinal uses of Kalanchoe gastonis-bonnieri (KGB), we evaluated the inhibitory effects on the proliferation of stromal cells from primary benign prostatic hyperplasia (BPH) of four different aqueous extracts from this plant: underground parts from specimens in flower (T1 treatment), leaves from specimens in flower (T2 treatment), and flowers (T3 treatment) and leaves from specimens not in flower (T4 treatment). T1, T2, T3, and T4 treatments at 250 µg/ml for 72 hours inhibited BPH cells by 56.7%, 29.2%, 39.4%, and 13.5%, respectively, showing that the KGB underground parts extract (T1 treatment) was the most active. Our findings show that the extract of the KGB underground parts (150 and 250 µg/ml) stimulates important changes in the BPH cells, modulating crucial processes such as proliferation, viability, and apoptosis. HPLC-DAD-MS/MS analysis provided a tentative identification of glycosylated syringic acid derivatives, glycosylated forms of volatile compounds, and lignans in this extract. Finally, these results suggest that there is a potential therapeutic use for KGB in BPH, which could improve the clinical management of the disease.

15.
Mol Cell Endocrinol ; 486: 1-10, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30753853

ABSTRACT

This study aimed to analyze galectin-3 importance in endometriotic lesions development and the effect of recombinant Gal-3 carbohydrate recognition domain (Gal3C) in experimental endometriosis treatment. Experimental endometriosis was induced in WT and Gal-3-/- mice. Initially developed lesions were macroscopically and histologically analyzed, including immunohistochemical analysis. Then, WT mice were treated with Gal3C for 15 days. Gal-3 deficiency and Gal3C treatment significantly impaired endometriosis development. A significant decrease in lesions implantation and size, VEGF and VEGFR-2 expression, vascular density and macrophage distribution were observed in Gal-3 absence or inhibition. A greater presence of iNOS positive cells was observed in knockout mice lesions, while the presence of Arginase positive cells was higher in the WT animal lesions. In addition, COX-2 and TGFb1 were reduced by Gal3C treatment. Data showed here indicate a relevant role of Gal-3 in endometriosis development and highlight a target of endometriosis treatment using Gal-3 inhibitor.


Subject(s)
Endometriosis/drug therapy , Endometriosis/metabolism , Galectin 3/metabolism , Molecular Targeted Therapy , Animals , Biomarkers/metabolism , Endometrium/drug effects , Endometrium/pathology , Female , Galectin 3/chemistry , Inflammation/pathology , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred BALB C , Mice, Knockout , Neovascularization, Pathologic/drug therapy , Protein Domains , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
Methods Mol Biol ; 1842: 105-121, 2018.
Article in English | MEDLINE | ID: mdl-30196405

ABSTRACT

To understand the molecular mechanisms involved in gastric disorders and regeneration, we need an in vitro tridimensional (3D) culture model, which can mimic the in vivo gastric microenvironment. A 3D coculture system named gastrosphere is proposed herein, composed of primary human gastric epithelial and stromal cells. The primary cultures were obtained from endoscopic gastric biopsies, and after mechanical and enzymatic dispersion, epithelial (HGE3) and stromal (HGS12) cells were expanded. After extensive immunocytochemical characterization, cells were seeded onto 96-well round bottom plates previously covered with 1% agarose. Cells were cultured in KM-F12 culture medium with 10% fetal bovine serum (FBS), antibiotics, and antimycotics, in humidified air at 37 °C and atmosphere containing 5% CO2 for 72 h or until spheres formation. Then gastrospheres were carefully transferred to a rotary cell culture system (RCCS-4), and maintained for 07, 14, 21, and 28 days. Gastrospheres were morphologically characterized by immunocytochemistry [cytokeratins (CK), vimentin, α-smooth muscle actin (α-SMA), laminin (LN), fibronectin (FN), and type IV collagen (CIV), proliferating cell nuclear antigen (PCNA)], and electron microscopy. In gastrospheres, the cytokeratin-positive epithelial cells were found in the outer layer, while vimentin-positive stromal cells were localized in the center of the gastrospheres. PCNA+ cells were mainly seen at the peripheral and in the intermediary region while nestin+ cells were also depicted in the latter zone. Scanning electron microscopy revealed groups of cohesive gastric cells at the periphery, while transmission electron microscopy demonstrated some differentiated mucous-like or zymogenic-like cells in the periphery and stromal structures located at the center of the 3D structures. Extracellular matrix was deposed between cells. Our data suggest that in vitro gastrospheres recapitulate the in vivo gastric microenvironment.


Subject(s)
Cell Culture Techniques , Coculture Techniques , Spheroids, Cellular , Animals , Biomarkers , Biopsy , Cellular Microenvironment , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Fluorescent Antibody Technique , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Gastric Mucosa/ultrastructure , Immunohistochemistry , Mice , Stomach , Stomach Diseases/etiology , Stomach Diseases/metabolism , Stomach Diseases/pathology , Stromal Cells/cytology , Stromal Cells/metabolism , Stromal Cells/ultrastructure
17.
World J Urol ; 36(12): 2009-2019, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29980839

ABSTRACT

Malignancies of the central nervous system include primary brain tumors and brain metastases, the latter being the major cause of intracranial neoplasms in adults. Although prostate cancer (PCa) brain metastases are not the most common source, recent data show that the relevance of prostate cancer brain metastases (PCBM) cannot be neglected. In this review, we focus on the molecular repertory as well as on the phenotypical similarities between PCBM and primary PCa, such as the cellular evolution and the maintenance of androgen-receptor expression. Moreover, the simultaneous occurrence of PCBM with other PCa metastatic sites and the significance of the clinical heterogeneity of the disease are also discussed. In addition, a potential relationship between the heterogeneous behavior exhibited by PCBM and the co-occurrence of malignant cell clusters with distinct genetic profiles is also hypothesized, as well as the prominent role of astrocytes in the establishment of PCBM.


Subject(s)
Adenocarcinoma/secondary , Brain Neoplasms/secondary , Prostatic Neoplasms/pathology , Adenocarcinoma/genetics , Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics
18.
PLoS One ; 13(7): e0200101, 2018.
Article in English | MEDLINE | ID: mdl-29966007

ABSTRACT

Cancer is an increasingly frequent malignancy worldwide, and despite the advances in drug development, it is still necessary to develop new plant-derived medicines. Euterpe oleracea (açaí) is abundant in South and Central America and has health benefits due to its high levels of phytochemicals, including lignans and polyphenols. The aim of this review was to systematically describe the safety and antitumor effects of açaí in preclinical models using rodents to provide a more comprehensive assessment of açaí for both therapeutic uses and the development of future clinical studies in cancer. Eligible studies were identified using four international databases (PubMed, Medline, Lilacs and SciELO) from their inception date through December 2017. The included studies were analyzed with methodological rigor (QATRS) to enable better quality control for these experimental studies. Sixty publications were identified in the databases, but only 9 articles were eligible: 6 evaluated the pharmacological effects of açaí in animal models of cancer (1 model each of esophageal cancer, urothelial cancer, melanoma and Walker-256 tumor and 2 models of colon cancer), and 3 were toxicological assays using preclinical models with rodents. Overall, 747 animals were analyzed. On a QATRS score scale of 0-20, the quality of the studies ranged from 16 to 20 points. Pulp was the main fraction of açaí administered, and an oral administration route was most common. The açaí dosage administered by gavage ranged from 30 mg/kg to 40,000 mg/kg, and açaí fed in the diet accounted for 2.5% to 5% of the diet. The anticarcinogenic and chemopreventive activities of açaí were observed in all experimental models of cancer and reduced the incidence, tumor cell proliferation, multiplicity and size of the tumors due to the antiinflammatory, antiproliferative and proapoptotic properties of açaí. No genotoxic effects were observed after açaí administration. The results of this review suggest that açaí is safe and can be used as a chemoprotective agent against cancer development. Açaí therapy may be a novel strategy for treating cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Euterpe , Plant Extracts/pharmacology , Plant Extracts/toxicity , Animals , Humans
19.
Mol Cell Endocrinol ; 476: 17-26, 2018 11 15.
Article in English | MEDLINE | ID: mdl-29689297

ABSTRACT

The present work aimed to evaluate molecular, angiogenic and inflammatory changes induced by clotrimazole (CTZ) on endometriosis lesions. For this, thirty female Wistar rats with surgically implanted autologous endometrium were treated with CTZ or vehicle (200 mg/kg) via esophageal gavage for 15 consecutive days. CTZ treatment significantly decreased the growth and the size of the implants, and histological examination indicated regression and atrophy, with no toxicity to the animals. The levels of the angiogenic markers VEGF and VEGFR-2 were significantly decreased in CTZ group. The treatment also promotes a reduction on PGE2 and TNF-α levels. All these effects involve the amelioration of ERK1/2, Akt, AMPK and PERK signaling upon CTZ treatment. In conclusion, CTZ promoted an overall amelioration of endometriosis in a rat model due to the anti-angiogenic properties of the drug. Therefore, our results support the proposal of a clinical trial using CTZ for the treatment of endometriosis.


Subject(s)
Clotrimazole/therapeutic use , Endometriosis/drug therapy , Endometrium/pathology , Prostheses and Implants , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Biomarkers/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Clotrimazole/adverse effects , Clotrimazole/pharmacology , Disease Models, Animal , Down-Regulation/drug effects , Endometriosis/pathology , Endometrium/blood supply , Endometrium/drug effects , Female , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Rats, Wistar
20.
BMC Complement Altern Med ; 18(1): 116, 2018 Apr 02.
Article in English | MEDLINE | ID: mdl-29609579

ABSTRACT

BACKGROUND: Among the processes involved in the breast tumor microenvironment, angiogenesis and inflammation play a central role, and the main factors of these processes are the vascular endothelial growth factor (VEGF), cyclooxygenase 2 (COX-2) and macrophages. Recently, the extract of Euterpe oleracea (açaí), a fruit that is widely found in the Amazon region, already showed antitumorigenic effects in vitro in human breast cancer cell lines. The present study aimed to investigate the effect of açaí on breast cancer using a chemically DMBA (7,12-dimethylbenzanthracene) experimental model. METHODS: One day after initiation of treatment with açaí, mammary carcinogenesis was induced in female Wistar rats using a subcutaneous injection of 25 mg/kg of DMBA in the mammary gland. Forty rats were randomized into two groups: treated with 200 mg/kg of either açaí extract or vehicle, via gastric tube for 16 consecutive weeks. After treatment, the tumor was collected for macroscopic, histological and immunohistochemical (VEGF, vascular endothelial growth factor receptor 2 -VEGFR-2, COX-2 and matrix metalloproteinase -MMP-9) analyses; peritoneal fluid was subjected to flow cytometry (F4-80/MAC-2+) and ELISA immunoassay (VEGF, prostaglandin E2 -PGE2 and interleukin-10 -IL-10). Heart, liver and kidney samples were collected for histological analysis. RESULTS: After 16 weeks of induction, the mammary carcinoma was confirmed by macroscopic and histological evaluation. Survival analysis indicates that açaí increased the survival (P = .0002, long-rank test) and reduced the deaths number (P = .0036, Chi-square test). Açaí treatment decreased the number of inflammatory cells and macrophage positive cells (Mac-2 + F4-80+), as well as promoting a reduction in immunostaining of VEGF, VEGFR-2 and COX-2. The açaí group also exhibited lower concentrations of PGE2, VEGF and IL-10 compared to the control. The histopathological results of the liver and kidneys showed protective effect of açaí, since in the control group, there was an increase in fibrosis, atypical cells and hemorrhagic microenvironment. CONCLUSION: The results of this study demonstrated the antiangiogenic and anti-inflammatory potential of açaí, like due to the decreases of the number of activated macrophages, resulting in the inhibition of DMBA carcinogenicity in breast cancer.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene/toxicity , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Carcinogens/toxicity , Euterpe/chemistry , Mammary Neoplasms, Experimental , Plant Extracts/pharmacology , Animals , Cell Line, Tumor , Female , Humans , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...