Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 14: 1176629, 2023.
Article in English | MEDLINE | ID: mdl-37886132

ABSTRACT

Amazonian flora includes several species with the potential to develop pharmaceutical and biotechnological products. The essential oils from Amazonian species possess some biological properties, such as antioxidant, antibacterial, and cytotoxic activities. The essential oil of red sacaca (RSO), Croton cajucara Benth., contains metabolites characterized by antioxidant and anti-inflammatory activities. Nanostructured lipid carriers (NLC) are an advantageous alternative for the effective delivery of drugs because they can solubilize lipophilic actives and reduce their cytotoxicity. This study aimed to optimize the synthesis of RSO-loaded nanostructured lipid carriers (NLC-RSO) using a 23 factorial design and investigate their antioxidant and cytotoxic effects. The red sacaca essential oil (RSO) metabolite profile was characterized using gas chromatography coupled with a mass spectrometer (GC-MS), identifying 33 metabolites, with linalool and 7-hydroxy-calamenene as the major ones, as reported in the literature. The optimized NLC-RSO formulation had a particle size less than 100 nm and a polydispersity index lower than 0.25. After characterizing NLC-RSO using Fourier-transform infrared spectroscopy, powder X-ray diffraction, zeta potential, moisture content, and wettability, in vitro cytotoxicity were performed in A549 and BEAS-2B cell lines using the resazurin metabolism assay. The data indicated a lower IC50 for RSO than for NLC-RSOs in both cell lines. Furthermore, low cytotoxicity of blank nanoparticles (blank NP) and medium chain triglycerides-loaded nanostructured lipid carriers (NLC-MCT) towards both pulmonary cell lines was noted. At a concentration of 50-100 µg/mL, free RSO exhibited higher cytotoxicity than NLC-RSO, demonstrating the protective effect of this lipid carrier in reducing cytotoxicity during metabolite delivery. Similarly, free RSO showed higher 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging than NLC-RSO, also indicating this protective effect. The 2',7'-dichlorofluorescein diacetate (DCFH-DA) intracellular reactive oxygen species (ROS) level assay did not show differences between the treatments at higher but non-cytotoxic dosages. Taken together, our results suggest that NLC-RSOs are potential RSO delivery systems for applications related to cancer treatment.

2.
Cytokine ; 171: 156371, 2023 11.
Article in English | MEDLINE | ID: mdl-37725872

ABSTRACT

Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer in the world, and accounts for 25% of all childhood cancers among children under 15 years of age. Longitudinal studies have shown that children with ALL are born with a deregulated immune response that, together with postnatal environmental exposures, favor the onset of the disease. In this context, IL-10, a key cytokine in the regulation of the immune response, presents itself as a paradoxical mediator, initially influencing the development of ALL through the regulation of inflammatory processes and later on the progression of malignancy, with the increase of this molecule in the leukemia microenvironment. According to the literature, this cytokine plays a critical role in the natural history of the disease and plays an important role in two different though complex scenarios. Thus, in this review, we explore the dual role of IL-10 in ALL, and describe its biological characteristics, immunological mechanisms and genetics, as well as its impact on the leukemia microenvironment and its clinical implications.


Subject(s)
Interleukin-10 , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Child , Humans , Carcinogenesis , Cytokines , Interleukin-10/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Tumor Microenvironment
4.
Biomed Pharmacother ; 144: 112269, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34794230

ABSTRACT

Head and neck cancer (HNC), a common malignancy worldwide, is associated with high morbidity and mortality rates. Squamous cell carcinoma is the most common HNC type, followed by salivary gland carcinomas, head and neck sarcomas, and lymphomas. The microenvironment of HNCs comprises various cells that regulate tumor development. Recent studies have reported that the tumor microenvironment, which modulates cancer progression, regulates cancer treatment response. However, the presence of different types of stromal cells in cancers is a major challenge to elucidate the role of individual cells in tumor progression. The role of mesenchymal stromal cells (MSCs), which are a component of the tumor microenvironment, in HNC is unclear. The major impediment for characterizing the role of MSCs in cancer progression is the lack of MSC-specific markers and their phenotypic similarity with stromal cells. This review aimed to summarize the latest findings on the role of MSCs in the progression of HNC to improve our understanding of HNC pathophysiology.


Subject(s)
Head and Neck Neoplasms/pathology , Mesenchymal Stem Cells/pathology , Tumor Microenvironment , Biomarkers, Tumor , Carcinoma, Squamous Cell/pathology , Epithelial-Mesenchymal Transition , Humans , Neoplasm Metastasis , Squamous Cell Carcinoma of Head and Neck/pathology
5.
Adv Biol Regul ; 82: 100834, 2021 12.
Article in English | MEDLINE | ID: mdl-34710785

ABSTRACT

Ca2+ is a highly versatile intracellular signal that regulates many biological processes such as cell death and proliferation. Broad Ca2+-signaling machinery is used to assemble signaling systems with a precise spatial and temporal resolution to achieve this versatility. Ca2+-signaling components can be organized in different regions of the cell and local increases in Ca2+ within the nucleus can regulate different cellular functions from the increases in cytosolic Ca2+. However, the mechanisms and pathways that promote localized increases in Ca2+ levels in the nucleus are still under investigation. This review presents evidence that the nucleus has its own Ca2+ stores and signaling machinery, which modulate processes such as cell proliferation and tumor growth. We focus on what is known about the functions of nuclear Phospholipase C (PLC) in the generation of nuclear Ca2+ transients that are involved in cell proliferation.


Subject(s)
Cell Nucleus , Type C Phospholipases , Calcium/metabolism , Calcium Signaling , Cell Nucleus/metabolism , Cell Proliferation , Cytosol/metabolism , Signal Transduction , Type C Phospholipases/genetics , Type C Phospholipases/metabolism
6.
Appl Biochem Biotechnol ; 193(11): 3704-3718, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34363139

ABSTRACT

In recent years, stem cell therapy has shown promise in regenerative medicine. The lack of standardized protocols for cell isolation and differentiation generates conflicting results in this field. Mesenchymal stem cells derived from adipose tissue (ASC) and fibroblasts (FIB) share very similar cell membrane markers. In this context, the distinction of mesenchymal stem cells from fibroblasts has been crucial for safe clinical application of these cells. In the present study, we developed aptamers capable of specifically recognize ASC using the Cell-SELEX technique. We tested the affinity of ASC aptamers compared to dermal FIB. Quantitative PCR was advantageous for the in vitro validation of four candidate aptamers. The binding capabilities of Apta 2 and Apta 42 could not distinguish both cell types. At the same time, Apta 21 and Apta 99 showed a better binding capacity to ASC with dissociation constants (Kd) of 50.46 ± 2.28 nM and 72.71 ± 10.3 nM, respectively. However, Apta 21 showed a Kd of 86.78 ± 9.14 nM when incubated with FIB. Therefore, only Apta 99 showed specificity to detect ASC by total internal reflection microscopy (TIRF). This aptamer is a promising tool for the in vitro identification of ASC. These results will help understand the differences between these two cell types for more specific and precise cell therapies.


Subject(s)
Adipose Tissue/metabolism , Aptamers, Nucleotide/pharmacology , Cell Differentiation/drug effects , Fibroblasts/metabolism , Mesenchymal Stem Cells/metabolism , Adipose Tissue/cytology , Aptamers, Nucleotide/chemistry , Cells, Cultured , Fibroblasts/cytology , Humans , Mesenchymal Stem Cells/cytology
7.
Biochimie ; 191: 69-77, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34454978

ABSTRACT

It is becoming increasingly evident that mesenchymal stem/stromal cells are recruited by cancer cells from nearby endogenous host stroma and promote events such as tumor proliferation, angiogenesis, invasion, and metastasis, as well as mediate therapeutic resistance. Consequently, understanding the regulatory mechanisms of ASCs that influence the tumor microenvironment may provide an avenue for further treatment. To understand the role of the ASC secretome in breast cancer cell proliferation, death, and phenotype alteration, adipose-derived stem cell-conditioned medium (mASC) was used to cultivate MCF-7 and MDA-MB-231 cells. These breast cancer cells in mASC showed a shorter doubling time, higher frequency of EdU positivity, and higher levels of phosphorylated histone 3. In addition, increased expression of cyclin B1 was observed, suggesting that proliferation was induced. The mASC was also able to increase apoptosis in MCF-7 cells, which was confirmed by caspase-7 activation. The number of tumor-initiating cells (CD44+ CD24-/low) and migration capacity were increased in cells cultivated in mASC. These data collectively suggest that ASC-conditioned medium can induce selective pressure by increasing cell proliferation, giving rise to a more aggressive phenotype in MCF-7 and MDA-MB-231 cells. Our study provides a foundation for further elucidation of the precise mechanism underlying ASCs in breast cancer cells and the modulation of ASCs in potential therapeutic uses.


Subject(s)
Adipose Tissue/metabolism , Breast Neoplasms/metabolism , Cell Differentiation , Cell Proliferation , Mesenchymal Stem Cells/metabolism , Secretome/metabolism , Tumor Microenvironment , Adipose Tissue/pathology , Breast Neoplasms/pathology , Coculture Techniques , Female , Humans , MCF-7 Cells , Mesenchymal Stem Cells/pathology
8.
Cell Biochem Funct ; 39(3): 442-454, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33389760

ABSTRACT

Human adipose-derived stromal/stem cells (ASC) have immunomodulatory properties and the potential to differentiate into several cell lines, important for application in regenerative medicine. However, the contamination with dermal fibroblasts (FIB) can impair the beneficial effects of ASC in cell therapy. It is then essential to develop new strategies that contribute to the distinction between these two cell types. In this study, we performed functional assays, high-throughput RNA sequencing (RNA-Seq) and quantitative PCR (qPCR) to find new markers that can distinguish ASC and FIB. We showed that ASC have adipogenic and osteogenic differentiation capacity and alkaline phosphatase activity, not observed in FIB. Gene expression variation analysis identified more than 2000 differentially expressed genes (DEG) between these two cell types. We validated 16 genes present in the list of DEG, including the alkaline phosphatase gene (ALPL). In conclusion, we showed that ASC and FIB have distinct biological properties as demonstrated by alkaline phosphatase activity and differentiation capacity, besides having different gene expression profiles. SIGNIFICANCE OF THE STUDY: Although many differences between stromal stem cells derived from human adipose tissue (ASC) and human dermal fibroblasts (FIB) are described, it is still difficult to find specific markers to differentiate them. This problem can interfere with the therapeutic use of ASC. This work aimed to find new markers to differentiate these two cell populations. Our findings suggest that these cells can be distinguished by biological and molecular characteristics, such as adipogenic and osteogenic differentiation, alkaline phosphatase activity and differential gene expression profiles. The DEG were related to the regulation of the cell cycle, development process, structural organization of the cell and synthesis of the extracellular matrix. This study helps to find new cellular markers to distinguish the two populations and to better understand the properties of these cells, which can improve cell therapy.


Subject(s)
Adipose Tissue/metabolism , Dermis/metabolism , Fibroblasts/metabolism , RNA-Seq , Stem Cells/metabolism , Adipose Tissue/cytology , Dermis/cytology , Fibroblasts/cytology , Humans , Organ Specificity , Stem Cells/cytology , Stromal Cells/cytology , Stromal Cells/metabolism
9.
J Biol Chem ; 294(45): 16650-16662, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31537645

ABSTRACT

Calcium (Ca2+) signaling within the cell nucleus regulates specific cellular events such as gene transcription and cell proliferation. Nuclear and cytosolic Ca2+ levels can be independently regulated, and nuclear translocation of receptor tyrosine kinases (RTKs) is one way to locally activate signaling cascades within the nucleus. Nuclear RTKs, including the epidermal growth factor receptor (EGFR), are important for processes such as transcriptional regulation, DNA-damage repair, and cancer therapy resistance. RTKs can hydrolyze phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) within the nucleus, leading to Ca2+ release from the nucleoplasmic reticulum by inositol 1,4,5-trisphosphate receptors. PI(4,5)P2 hydrolysis is mediated by phospholipase C (PLC). However, it is unknown which nuclear PLC isoform is triggered by EGFR. Here, using subcellular fractionation, immunoblotting and fluorescence, siRNA-based gene knockdowns, and FRET-based biosensor reporter assays, we investigated the role of PLCδ4 in epidermal growth factor (EGF)-induced nuclear Ca2+ signaling and downstream events. We found that EGF-induced Ca2+ signals are inhibited when translocation of EGFR is impaired. Nuclear Ca2+ signals also were reduced by selectively buffering inositol 1,4,5-trisphosphate (InsP3) within the nucleus. EGF induced hydrolysis of nuclear PI(4,5)P2 by the intranuclear PLCδ4, rather than by PLCγ1. Moreover, protein kinase C, a downstream target of EGF, was active in the nucleus of stimulated cells. Furthermore, PLCδ4 and InsP3 modulated cell cycle progression by regulating the expression of cyclins A and B1. These results provide evidence that EGF-induced nuclear signaling is mediated by nuclear PLCδ4 and suggest new therapeutic targets to modulate the proliferative effects of this growth factor.


Subject(s)
Calcium Signaling/drug effects , Cell Nucleus/metabolism , Epidermal Growth Factor/pharmacology , Phospholipase C delta/metabolism , Cell Line , Cell Proliferation/drug effects , Clathrin Heavy Chains/antagonists & inhibitors , Clathrin Heavy Chains/genetics , Clathrin Heavy Chains/metabolism , Cyclin A/metabolism , Cyclin B1/metabolism , ErbB Receptors/metabolism , Humans , Hydrolysis , Inositol 1,4,5-Trisphosphate/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C delta/antagonists & inhibitors , Phospholipase C delta/genetics , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Protein Kinase C/metabolism , RNA Interference , RNA, Small Interfering/metabolism
10.
Cell Signal ; 49: 59-67, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29859928

ABSTRACT

Ca2+ is an important second messenger, and it is involved in many cellular processes such as cell death and proliferation. The rise in intracellular Ca2+ levels can be due to the generation of inositol 1,4,5-trisphosphate (InsP3), which is a product of phosphatidylinositol 4,5-bisphosphate (PIP2) hydrolysis by phospholipases C (PLCs), that leads to Ca2+ release from endoplasmic reticulum by InsP3 receptors (InsP3R). Ca2+ signaling patterns can vary in different regions of the cell and increases in nuclear Ca2+ levels have specific biological effects that differ from those of Ca2+ increase in the cytoplasm. There are PLCs in the cytoplasm and nucleus, but little is known about the functions of nuclear PLCs. This work aimed to characterize phenotypically the human PLCδ4 (hPLCδ4) in mesenchymal stem cells. This nuclear isoform of PLC is present in different cell types and has a possible role in proliferative processes. In this work, hPLCδ4 was found to be mainly nuclear in human adipose-derived mesenchymal stem cells (hASC). PLCδ4 knockdown demonstrated that it is essential for hASC proliferation, without inducing cell death. An increase of cells in G1, and a reduction of cells on interphase and G2/M in knockdown cells were seen. Furthermore, PLCδ4 knockdown increased the percentage of senescent cells, p16INK4A+ and p21Cip1 mRNAs expression, which could explain the impaired cell proliferation. The results show that hPLCδ4 is in involved in cellular proliferation and senescence in hASC.


Subject(s)
Cell Proliferation , Cellular Senescence , Phospholipase C delta/metabolism , Adipose Tissue/cytology , Cell Cycle Checkpoints , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Phospholipase C delta/antagonists & inhibitors , Phospholipase C delta/genetics , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism
11.
J Struct Biol ; 202(1): 61-69, 2018 04.
Article in English | MEDLINE | ID: mdl-29277356

ABSTRACT

Nuclear Epidermal Growth Factor Receptor (EGFR) has been associated with worse prognosis and treatment resistance for several cancer types. After Epidermal Growth Factor (EGF) binding, the ligand-receptor complex can translocate to the nucleus where it functions in oncological processes. By three-dimensional quantification analysis of super-resolution microscopy images, we verified the translocation kinetics of fluorescent conjugated EGF to the nucleus in two mesenchymal cell types: human adipose tissue-derived stem cells (hASC) and SK-HEP-1 tumor cells. The number of EGF clusters in the nucleus does not change after 10 min of stimulation with EGF in both cells. The total volume occupied by EGF clusters in the nucleus of hASC also does not change after 10 min of stimulation with EGF. However, the total volume of EGF clusters increases only after 20 min in SK-HEP-1 cells nuclei. In these cells the nuclear volume occupied by EGF is 3.2 times higher than in hASC after 20 min of stimulation, indicating that translocation kinetics of EGF differs between these two cell types. After stimulation, EGF clusters assemble in larger clusters in the cell nucleus in both cell types, which suggests specific sub-nuclear localizations of the receptor. Super-resolution microscopy images show that EGF clusters are widespread in the nucleoplasm, and can be localized in nuclear envelope invaginations, and in the nucleoli. The quantitative study of EGF-EGFR complex translocation to the nucleus may help to unravel its roles in health and pathological conditions, such as cancer.


Subject(s)
Cell Nucleus/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Mesenchymal Stem Cells/metabolism , Adipose Tissue/cytology , Cell Line, Tumor , Cell Lineage , Epidermal Growth Factor/chemistry , Fluorescent Dyes/chemistry , Humans , Kinetics , Mesenchymal Stem Cells/cytology , Nuclear Envelope/metabolism , Protein Transport
12.
Pathol Res Pract ; 212(4): 340-4, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26944829

ABSTRACT

The epidermal growth factor receptor (EGFR) has been described in the nucleus of primary tumors. Accumulation of EGFR at the nucleus is linked to DNA synthesis and cell proliferation, but the pathological significance of nuclear EGFR is not completely understood. The aim of this study was to investigate the nuclear localization of EGFR in invasive micropapillary carcinoma (IMPC) that is an aggressive neoplasm of canine mammary gland. Confocal immunofluorescence of formalin and paraffin-embedded tissue was used to access the subcellular localization of EGFR. Our results demonstrated that EGFR co-localizes with the inner nuclear envelope marker, Lamin B1 in IMPC. Furthermore, EGFR was not localized within the nucleus or at the inner nuclear envelope membrane in mammary carcinoma in mixed tumor (CMT) that is associated with a better prognosis than other malignant histological types. This finding could be useful as a predictive biomarker of therapeutic response for IMPC.


Subject(s)
Carcinoma, Papillary/veterinary , Dog Diseases/metabolism , ErbB Receptors/metabolism , Mammary Neoplasms, Animal/metabolism , Animals , Blotting, Western , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Disease Models, Animal , Dog Diseases/pathology , Dogs , Female , Fluorescent Antibody Technique , Mammary Neoplasms, Animal/pathology , Microscopy, Confocal , Nuclear Envelope/metabolism , Prognosis
13.
J Cancer Sci Ther ; 5(2): 58-65, 2013 Jan 21.
Article in English | MEDLINE | ID: mdl-24738023

ABSTRACT

Cancer is comprised of a multitude of epigenetic abnormalities, including the global loss and regional gain of DNA methylation as well as alterations in histone methylation. Here, we characterize a new methyltransferase, SET domain-containing protein 4 (SETD4), which is involved in breast carcinogenesis. Quantitative real-time PCR (qPCR) showed elevated expression levels of SETD4 in several breast cancer cell lines. SETD4 overexpression was confirmed by western blot analysis suggesting a correlation between high expression of SETD4 and a lack of the estrogen receptor (ER) in breast cancer. In addition, cell fractionation studies and confocal immunofluorescence revealed the nuclear and non-nuclear localization of this new protein. SETD4 knockdown in breast cancer cell lines significantly suppressed their proliferation and delayed the G1/S cell cycle transition without affecting apoptosis. Furthermore, western blot analysis showed that knockdown of SETD4 decreased cyclin D1 expression, revealing the involvement of SETD4 in cell cycle regulation. These data imply that SETD4 plays a crucial role in breast carcinogenesis and could be a novel molecular target for the development of new strategies for the diagnosis and treatment of breast cancer.

SELECTION OF CITATIONS
SEARCH DETAIL
...