Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters











Publication year range
1.
PLoS Biol ; 22(7): e3002074, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39038054

ABSTRACT

While interactions between neural crest and placode cells are critical for the proper formation of the trigeminal ganglion, the mechanisms underlying this process remain largely uncharacterized. Here, by using chick embryos, we show that the microRNA (miR)-203, whose epigenetic repression is required for neural crest migration, is reactivated in coalescing and condensing trigeminal ganglion cells. Overexpression of miR-203 induces ectopic coalescence of neural crest cells and increases ganglion size. By employing cell-specific electroporations for either miR-203 sponging or genomic editing using CRISPR/Cas9, we elucidated that neural crest cells serve as the source, while placode cells serve as the site of action for miR-203 in trigeminal ganglion condensation. Demonstrating intercellular communication, overexpression of miR-203 in the neural crest in vitro or in vivo represses an miR-responsive sensor in placode cells. Moreover, neural crest-secreted extracellular vesicles (EVs), visualized using pHluorin-CD63 vector, become incorporated into the cytoplasm of placode cells. Finally, RT-PCR analysis shows that small EVs isolated from condensing trigeminal ganglia are selectively loaded with miR-203. Together, our findings reveal a critical role in vivo for neural crest-placode communication mediated by sEVs and their selective microRNA cargo for proper trigeminal ganglion formation.


Subject(s)
Cell Communication , Extracellular Vesicles , MicroRNAs , Neural Crest , Trigeminal Ganglion , Neural Crest/metabolism , Neural Crest/embryology , Neural Crest/cytology , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/embryology , Trigeminal Ganglion/cytology , Extracellular Vesicles/metabolism , Chick Embryo , Cell Communication/genetics , Cell Movement/genetics , Gene Expression Regulation, Developmental
2.
ACS Synth Biol ; 12(1): 71-82, 2023 01 20.
Article in English | MEDLINE | ID: mdl-36534043

ABSTRACT

Cell communication is a widespread mechanism in biology, allowing the transmission of information about environmental conditions. In order to understand how cell communication modulates relevant biological processes such as survival, division, differentiation, and apoptosis, different synthetic systems based on chemical induction have been successfully developed. In this work, we coupled cell communication and optogenetics in the budding yeast Saccharomyces cerevisiae. Our approach is based on two strains connected by the light-dependent production of α-factor pheromone in one cell type, which induces gene expression in the other type. After the individual characterization of the different variants of both strains, the optogenetic intercellular system was evaluated by combining the cells under contrasting illumination conditions. Using luciferase as a reporter gene, specific co-cultures at a 1:1 ratio displayed activation of the response upon constant blue light, which was not observed for the same cell mixtures grown in darkness. Then, the system was assessed at several dark/blue-light transitions, where the response level varies depending on the moment in which illumination was delivered. Furthermore, we observed that the amplitude of response can be tuned by modifying the initial ratio between both strains. Finally, the two-population system showed higher fold inductions in comparison with autonomous strains. Altogether, these results demonstrated that external light information is propagated through a diffusible signaling molecule to modulate gene expression in a synthetic system involving microbial cells, which will pave the road for studies allowing optogenetic control of population-level dynamics.


Subject(s)
Light , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Cell Communication/genetics , Signal Transduction , Cell Differentiation , Optogenetics/methods
3.
Cell Commun Signal ; 19(1): 86, 2021 08 14.
Article in English | MEDLINE | ID: mdl-34391444

ABSTRACT

BACKGROUND: Maspin (SERPINB5) is a potential tumor suppressor gene with pleiotropic biological activities, including regulation of cell proliferation, death, adhesion, migration and gene expression. Several studies indicate that nuclear localization is essential for maspin tumor suppression activity. We have previously shown that the EGFR activation leads to maspin nuclear localization in MCF-10A cells. The present study investigated which EGFR downstream signaling molecules are involved in maspin nuclear localization and explored a possible role of cell-cell contact in this process. METHODS: MCF-10A cells were treated with pharmacological inhibitors against EGFR downstream pathways followed by EGF treatment. Maspin subcellular localization was determined by immunofluorescence. Proteomic and interactome analyses were conducted to identify maspin-binding proteins in EGF-treated cells only. To investigate the role of cell-cell contact these cells were either treated with chelating agents or plated on different cell densities. Maspin and E-cadherin subcellular localization was determined by immunofluorescence. RESULTS: We found that PI3K-Akt and JAK2-STAT3, but not MAP kinase pathway, regulate EGF-induced maspin nuclear accumulation in MCF-10A cells. We observed that maspin is predominantly nuclear in sparse cell culture, but it is redistributed to the cytoplasm in confluent cells even in the presence of EGF. Proteomic and interactome results suggest a role of maspin on post-transcriptional and translation regulation, protein folding and cell-cell adhesion. CONCLUSIONS: Maspin nuclear accumulation is determined by an interplay between EGFR (via PI3K-Akt and JAK2-STAT3 pathways) and cell-cell contact. Video Abstract.


Subject(s)
Cell Communication/genetics , Janus Kinase 2/genetics , STAT3 Transcription Factor/genetics , Serpins/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/ultrastructure , Cell Proliferation/genetics , Epidermal Growth Factor/genetics , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Mitogen-Activated Protein Kinases/genetics , Phosphatidylinositol 3-Kinases/genetics , Proteomics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics
4.
Signal Transduct Target Ther ; 5(1): 99, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32555170

ABSTRACT

Normal cells are hijacked by cancer cells forming together heterogeneous tumor masses immersed in aberrant communication circuits that facilitate tumor growth and dissemination. Besides the well characterized angiogenic effect of some tumor-derived factors; others, such as BDNF, recruit peripheral nerves and leukocytes. The neurogenic switch, activated by tumor-derived neurotrophins and extracellular vesicles, attracts adjacent peripheral fibers (autonomic/sensorial) and neural progenitor cells. Strikingly, tumor-associated nerve fibers can guide cancer cell dissemination. Moreover, IL-1ß, CCL2, PGE2, among other chemotactic factors, attract natural immunosuppressive cells, including T regulatory (Tregs), myeloid-derived suppressor cells (MDSCs), and M2 macrophages, to the tumor microenvironment. These leukocytes further exacerbate the aberrant communication circuit releasing factors with neurogenic effect. Furthermore, cancer cells directly evade immune surveillance and the antitumoral actions of natural killer cells by activating immunosuppressive mechanisms elicited by heterophilic complexes, joining cancer and immune cells, formed by PD-L1/PD1 and CD80/CTLA-4 plasma membrane proteins. Altogether, nervous and immune cells, together with fibroblasts, endothelial, and bone-marrow-derived cells, promote tumor growth and enhance the metastatic properties of cancer cells. Inspired by the demonstrated, but restricted, power of anti-angiogenic and immune cell-based therapies, preclinical studies are focusing on strategies aimed to inhibit tumor-induced neurogenesis. Here we discuss the potential of anti-neurogenesis and, considering the interplay between nervous and immune systems, we also focus on anti-immunosuppression-based therapies. Small molecules, antibodies and immune cells are being considered as therapeutic agents, aimed to prevent cancer cell communication with neurons and leukocytes, targeting chemotactic and neurotransmitter signaling pathways linked to perineural invasion and metastasis.


Subject(s)
Molecular Targeted Therapy , Neoplasms/genetics , Neurogenesis/genetics , Tumor Escape/genetics , Cell Communication/genetics , Humans , Killer Cells, Natural/immunology , Neoplasms/complications , Neoplasms/therapy , Tumor Escape/immunology
5.
Int J Mol Sci ; 20(19)2019 Sep 30.
Article in English | MEDLINE | ID: mdl-31575081

ABSTRACT

Hematopoietic progenitor cell (HPC) transplantation is a treatment option for malignant and nonmalignant diseases. Umbilical cord blood (UCB) is an important HPC source, mainly for pediatric patients. It has been demonstrated that human leukocyte antigen (HLA) matching and cell dose are the most important features impacting clinical outcomes. However, UCB matching is performed using low resolution HLA typing and it has been demonstrated that the unnoticed mismatches negatively impact the transplant. Since we found differences in CD34+ viability after thawing of UCB units matched for two different patients (p = 0.05), we presumed a possible association between CD34+ cell viability and HLA. We performed a multivariate linear model (n = 67), comprising pre-cryopreservation variables and high resolution HLA genotypes separately. We found that pre-cryopreservation red blood cells (RBC), granulocytes, and viable CD34+ cell count significantly impacted CD34+ viability after thawing, along with HLA-B or -C (R2 = 0.95, p = 0.01; R2 = 0.56, p = 0.007, respectively). Although HLA-B*40:02 may have a negative impact on CD34+ cell viability, RBC depletion significantly improves it.


Subject(s)
Cell Communication , Erythrocytes/metabolism , Fetal Blood/cytology , HLA Antigens/genetics , Hematopoietic Stem Cells/metabolism , Alleles , Antigens, CD34/metabolism , Cell Communication/genetics , Cell Survival/genetics , Cord Blood Stem Cell Transplantation , Cryopreservation , Hematopoietic Stem Cells/cytology , Humans
6.
Glia ; 67(8): 1598-1619, 2019 08.
Article in English | MEDLINE | ID: mdl-31033038

ABSTRACT

Diverse studies have suggested that cytoplasmic inclusions of misfolded α-synuclein in neuronal and glial cells are main pathological features of different α-synucleinopathies, including Parkinson's disease and dementia with Lewy bodies. Up to now, most studies have focused on the effects of α-synuclein on neurons, whereas the possible alterations of astrocyte functions and neuron-glia crosstalk have received minor attention. Recent evidence indicates that cellular signaling mediated by hemichannels and pannexons is critical for astroglial function and dysfunction. These channels constitute a diffusional route of communication between the cytosol and the extracellular space and during pathological scenarios they may lead to homeostatic disturbances linked to the pathogenesis and progression of different diseases. Here, we found that α-synuclein enhances the opening of connexin 43 (Cx43) hemichannels and pannexin-1 (Panx1) channels in mouse cortical astrocytes. This response was linked to the activation of cytokines, the p38 MAP kinase, the inducible nitric oxide synthase, cyclooxygenase 2, intracellular free Ca2+ concentration ([Ca2+ ]i ), and purinergic and glutamatergic signaling. Relevantly, the α-synuclein-induced opening of hemichannels and pannexons resulted in alterations in [Ca2+ ]i dynamics, nitric oxide (NO) production, gliotransmitter release, mitochondrial morphology, and astrocyte survival. We propose that α-synuclein-mediated opening of astroglial Cx43 hemichannels and Panx1 channels might constitute a novel mechanism involved in the pathogenesis and progression of α-synucleinopathies.


Subject(s)
Astrocytes/pathology , Cell Death/genetics , Connexin 43/genetics , Connexins/genetics , Nerve Tissue Proteins/genetics , alpha-Synuclein/genetics , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Cell Communication/genetics , Cells, Cultured , Cytokines/metabolism , Mice , Mitochondria/genetics , Mitochondria/ultrastructure , Neurotransmitter Agents/metabolism , Nitric Oxide/biosynthesis , RNA, Small Interfering/genetics
7.
Sci Rep ; 9(1): 6270, 2019 04 18.
Article in English | MEDLINE | ID: mdl-31000742

ABSTRACT

Mesangial cells stimulated with high glucose (HG) exhibit increased intracellular angiotensin II (AngII) synthesis that is correlated with the upregulation of AngII target genes, such as profibrotic cytokines. The intracrine effects of AngII can be mediated by several molecules transferred to other cells via exosomes (Exos), which play a key role in cellular communication under many physiological and pathological conditions. The aim of this study was to investigate the effects of exosomes derived from HG-stimulated human mesangial cells (HG-HMCs) on normal unstimulated HMCs. Exosomes from HMCs (C-Exos) and HG-HMCs (HG-Exos) were obtained from cell culture supernatants. HMCs were incubated with C-Exos or HG-Exos. HG stimulus induced a change in the amount but not the size of Exos. Both C-Exos and HG-Exos contained angiotensinogen and renin, but no angiotensin converting enzyme was detected. Compared with HMCs treated with C-Exos, HMCs treated with HG-Exos presented higher levels of fibronectin, angiotensinogen, renin, AT1 and AT2 receptors, indicating that HG-Exos modified the function of normal HMCs. These results suggest that the intercellular communication through Exos may have pathophysiological implications in the diabetic kidney.


Subject(s)
Angiotensin II/genetics , Cell Communication/genetics , Diabetic Nephropathies/genetics , Exosomes/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Exosomes/pathology , Fibronectins/genetics , Gene Expression Regulation/genetics , Glomerular Mesangium/metabolism , Glucose/metabolism , Humans , Kidney/metabolism , Kidney/pathology , Mesangial Cells/metabolism , Peptidyl-Dipeptidase A/genetics , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 2/genetics , Renin/genetics
8.
Mitochondrion ; 48: 16-30, 2019 09.
Article in English | MEDLINE | ID: mdl-30771504

ABSTRACT

Mitochondria play an important role as an intracellular energy plant and signaling organelle. However, mitochondria also exist outside cells where they could mediate cell-to-cell communication, repair and serve as an activator of the immune response. Their effects depend on the mitochondrial state or the form in which it is present, either as a whole functional structure as fragments or only as mitochondrial DNA. Herein, we provide evidence of why extracellular mitochondria and their varying forms are considered regenerative factors or pro-inflammatory activators. Understanding these aspects will provide the base of their use in therapy or as a biomarker of disease severity and prognosis.


Subject(s)
Mitochondria/physiology , Animals , Biomarkers/metabolism , Cell Communication/genetics , Cell Communication/physiology , DNA, Mitochondrial/genetics , Humans , Inflammation/genetics , Inflammation/physiopathology , Mitochondria/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
9.
Cell Death Dis ; 7(12): e2570, 2016 12 29.
Article in English | MEDLINE | ID: mdl-28032857

ABSTRACT

Prostate cancer (PCa) cells display abnormal expression of cytoskeletal proteins resulting in an augmented capacity to resist chemotherapy and colonize distant organs. We have previously shown that heme oxygenase 1 (HO-1) is implicated in cell morphology regulation in PCa. Here, through a multi 'omics' approach we define the HO-1 interactome in PCa, identifying HO-1 molecular partners associated with the integrity of the cellular cytoskeleton. The bioinformatics screening for these cytoskeletal-related partners reveal that they are highly misregulated in prostate adenocarcinoma compared with normal prostate tissue. Under HO-1 induction, PCa cells present reduced frequency in migration events, trajectory and cell velocity and, a significant higher proportion of filopodia-like protrusions favoring zippering among neighboring cells. Moreover forced expression of HO-1 was also capable of altering cell protrusions in transwell co-culture systems of PCa cells with MC3T3 cells (pre-osteoblastic cell line). Accordingly, these effects were reversed under siHO. Transcriptomics profiling evidenced significant modulation of key markers related to cell adhesion and cell-cell communication under HO-1 induction. The integration from our omics-based research provides a four molecular pathway foundation (ANXA2/HMGA1/POU3F1; NFRSF13/GSN; TMOD3/RAI14/VWF; and PLAT/PLAU) behind HO-1 regulation of tumor cytoskeletal cell compartments. The complementary proteomics and transcriptomics approaches presented here promise to move us closer to unravel the molecular framework underpinning HO-1 involvement in the modulation of cytoskeleton pathways, pushing toward a less aggressive phenotype in PCa.


Subject(s)
Cell Communication/genetics , Gene Regulatory Networks , Heme Oxygenase-1/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Pseudopodia/metabolism , Animals , Cell Communication/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Coculture Techniques , Crystallography, X-Ray , Culture Media, Conditioned/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks/drug effects , Humans , Male , Mice , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/genetics , Protein Binding/drug effects , Proteomics , Pseudopodia/drug effects , Sequence Analysis, RNA , Tandem Mass Spectrometry , Transcriptome/drug effects , Transcriptome/genetics
10.
Biochim Biophys Acta ; 1860(10): 2255-68, 2016 10.
Article in English | MEDLINE | ID: mdl-27130882

ABSTRACT

BACKGROUND: We previously demonstrated that the activated leukocyte cell adhesion molecule (ALCAM/CD166) can interact with galectin-8 (Gal-8) in endothelial cells. ALCAM is a member of the immunoglobulin superfamily that promotes homophilic and heterophilic cell-cell interactions. Gal-8 is a "tandem-repeat"-type galectin, known as a matricellular protein involved in cell adhesion. Here, we analyzed the physical interaction between both molecules in breast cancer cells and the functional relevance of this phenomenon. METHODS: We performed binding assays by surface plasmon resonance to study the interaction between Gal-8 and the recombinant glycosylated ALCAM ectodomain or endogenous ALCAM from MDA-MB-231 breast cancer cells. We also analyzed the binding of ALCAM-silenced or control breast cancer cells to immobilized Gal-8 by SPR. In internalization assays, we evaluated the influence of Gal-8 on ALCAM surface localization. RESULTS: We showed that recombinant glycosylated ALCAM and endogenous ALCAM from breast carcinoma cells physically interacted with Gal-8 in a glycosylation-dependent fashion displaying a differential behavior compared to non-glycosylated ALCAM. Moreover, ALCAM-silenced breast cancer cells exhibited reduced binding to Gal-8 relative to control cells. Importantly, exogenously added Gal-8 provoked ALCAM segregation, probably trapping this adhesion molecule at the surface of breast cancer cells. CONCLUSIONS: Our data indicate that Gal-8 interacts with ALCAM at the surface of breast cancer cells through glycosylation-dependent mechanisms. GENERAL SIGNIFICANCE: A novel heterophilic interaction between ALCAM and Gal-8 is demonstrated here, suggesting its physiologic relevance in the biology of breast cancer cells.


Subject(s)
Antigens, CD/metabolism , Breast Neoplasms/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Fetal Proteins/metabolism , Galectins/metabolism , Protein Interaction Maps/genetics , Antigens, CD/genetics , Breast Neoplasms/pathology , Cell Adhesion/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cell Communication/genetics , Cell Line, Tumor , Cell Movement/genetics , Endothelial Cells/metabolism , Female , Fetal Proteins/genetics , Galectins/genetics , Glycosylation , Humans , Protein Binding , Surface Properties
11.
Mol Cancer ; 14: 145, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26227631

ABSTRACT

The recent exponential increase in our knowledge of cellular and molecular mechanisms involved in carcinogenesis has largely failed to translate into new therapies and clinical practices. This lack of success may result in part from the fact that most studies focus on tumor cells as potential therapeutic targets and neglect the complex microenvironment that undergoes profound changes during tumor development. Furthermore, an unfortunate association of factors such as tumor genetic complexity, overestimation of biomarker and drug potentials, as well as a poor understanding of tumor microenvironment in diagnosis and prognosis leads to the current levels of treatment failure regarding a vast majority of cancer types. A growing body of evidence points to the importance of the functional diversity of immune and structural cells during tumor development. In this sense, the lack of technologies that would allow for molecular screening of individual stromal cell types poses a major challenge for the development of therapies targeting the tumor microenvironment. Progress in microenvironment genetic studies represents a formidable opportunity for the development of new selective drugs because stromal cells have lower mutation rates than malignant cells, and should prove to be good targets for therapy.


Subject(s)
Genomic Instability , Neoplasms/genetics , Neoplasms/pathology , Tumor Microenvironment/genetics , Animals , Cell Communication/drug effects , Cell Communication/genetics , Cell Communication/immunology , Genomic Instability/drug effects , Genomic Instability/genetics , Humans , Molecular Targeted Therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Tumor Microenvironment/immunology
12.
Biomed Res Int ; 2013: 279505, 2013.
Article in English | MEDLINE | ID: mdl-23762835

ABSTRACT

Tumor-associated immune cells often lack immune effector activities, and instead they present protumoral functions. To understand how tumors promote this immunological switch, invasive and noninvasive breast cancer cell (BRC) lines were cocultured with a promonocytic cell line in a Matrigel-based 3D system. We hypothesized that if communication exists between tumor and immune cells, coculturing would result in augmented expression of genes associated with tumor malignancy. Upregulation of proteases MMP1 and MMP9 and inflammatory COX2 genes was found likely in response to soluble factors. Interestingly, changes were more apparent in promonocytes and correlated with the aggressiveness of the BRC line. Increased gene expression was confirmed by collagen degradation assays and immunocytochemistry of prostaglandin 2, a product of COX2 activity. Untransformed MCF-10A cells were then used as a sensor of soluble factors with transformation-like capabilities, finding that acini formed in the presence of supernatants of the highly aggressive BRC/promonocyte cocultures often exhibited total loss of the normal architecture. These data support that tumor cells can modify immune cell gene expression and tumor aggressiveness may importantly reside in this capacity. Modeling interactions in the tumor stroma will allow the identification of genes useful as cancer prognostic markers and therapy targets.


Subject(s)
Acinar Cells/pathology , Breast Neoplasms/pathology , Collagen/metabolism , Cyclooxygenase 2/metabolism , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 9/metabolism , Monocyte-Macrophage Precursor Cells/enzymology , Acinar Cells/metabolism , Breast Neoplasms/genetics , Cell Communication/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Coculture Techniques , Dinoprostone/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Extracellular Matrix/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Monocyte-Macrophage Precursor Cells/pathology , Neoplasm Invasiveness , Phenotype , Proteolysis , Solubility , Up-Regulation
13.
Immunobiology ; 218(4): 609-19, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22954710

ABSTRACT

B-1 cells can be differentiated from B-2 cells because they are predominantly located in the peritoneal and pleural cavities and have distinct phenotypic patterns and activation properties. The role of both cell populations in cancer progression is still controversial. Previous studies have indicated that direct contact between B-1 cells and B16 melanoma tumor cells (B16) increases the metastatic potential of the tumor cells. However, cellular changes that are induced in B-1 cells during the interaction between these two cell types have not been evaluated. In the present study, it is hypothesized that B-1 cells are modified after their interaction with tumor cells, leading to both increased cell viability and rate of proliferation. Additionally, soluble factors that were secreted by B16 cells were sufficient to augment B-1 cell viability and to modify the production of IL-10 by B-1 cells. Impressively, after direct or indirect contact with the B16 cells, B-1 cells became resistant to radiation-induced cell death. Thus, future studies that assess the importance of concomitant immunity and other conventional therapies in cancer treatment are needed.


Subject(s)
B-Lymphocyte Subsets/immunology , Cell Communication/immunology , Cell Proliferation , Interleukin-10/immunology , Melanoma/immunology , Animals , B-Lymphocyte Subsets/pathology , Cell Communication/genetics , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/immunology , Interleukin-10/genetics , Melanoma/genetics , Melanoma/pathology , Melanoma/radiotherapy , Mice , Mice, Knockout , Neoplasm Metastasis , Peritoneum/immunology , Peritoneum/pathology , Pleural Cavity/immunology , Pleural Cavity/pathology
14.
PLoS One ; 7(9): e45449, 2012.
Article in English | MEDLINE | ID: mdl-23029016

ABSTRACT

Accruing evidence indicates that connexin (Cx) channels in the gap junctions (GJ) are involved in neurodegeneration after injury. However, studies using KO animal models endowed apparently contradictory results in relation to the role of coupling in neuroprotection. We analyzed the role of Cx-mediated communication in a focal lesion induced by mechanical trauma of the retina, a model that allows spatial and temporal definition of the lesion with high reproducibility, permitting visualization of the focus, penumbra and adjacent areas. Cx36 and Cx43 exhibited distinct gene expression and protein levels throughout the neurodegeneration progress. Cx36 was observed close to TUNEL-positive nuclei, revealing the presence of this protein surrounding apoptotic cells. The functional role of cell coupling was assessed employing GJ blockers and openers combined with lactate dehydrogenase (LDH) assay, a direct method for evaluating cell death/viability. Carbenoxolone (CBX), a broad-spectrum GJ blocker, reduced LDH release after 4 hours, whereas quinine, a Cx36-channel specific blocker, decreased LDH release as early as 1 hour after lesion. Furthermore, analysis of dying cell distribution confirmed that the use of GJ blockers reduced apoptosis spread. Accordingly, blockade of GJ communication during neurodegeneration with quinine, but not CBX, caused downregulation of initial and effector caspases. To summarize, we observed specific changes in Cx gene expression and protein distribution during the progress of retinal degeneration, indicating the participation of these elements in acute neurodegeneration processes. More importantly, our results revealed that direct control of GJ channels permeability may take part in reliable neuroprotection strategies aimed to rapid, fast treatment of mechanical trauma in the retina.


Subject(s)
Cell Communication/physiology , Connexins/metabolism , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Retina/metabolism , Retina/pathology , Wounds and Injuries/complications , Animals , Cell Communication/genetics , Chickens , Immunohistochemistry , In Situ Nick-End Labeling , Neurodegenerative Diseases/therapy , Polymerase Chain Reaction
15.
BMC Med Genomics ; 4: 76, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-22032772

ABSTRACT

BACKGROUND: A wide variety of high-throughput microarray platforms have been used to identify molecular targets associated with biological and clinical tumor phenotypes by comparing samples representing distinct pathological states. METHODS: The gene expression profiles of human cutaneous melanomas were determined by cDNA microarray analysis. Next, a robust analysis to determine functional classifications and make predictions based on data-oriented hypotheses was performed. Relevant networks that may be implicated in melanoma progression were also considered. RESULTS: In this study we aimed to analyze coordinated gene expression changes to find molecular pathways involved in melanoma progression. To achieve this goal, ontologically-linked modules with coordinated expression changes in melanoma samples were identified. With this approach, we detected several gene networks related to different modules that were induced or repressed during melanoma progression. Among them we observed high coordinated expression levels of genes involved in a) cell communication (KRT4, VWF and COMP); b) epidermal development (KLK7, LAMA3 and EVPL); and c) functionally related to kallikreins (EVPL, KLK6, KLK7, KLK8, SERPINB13, SERPING1 and SLPI). Our data also indicated that hKLK7 protein expression was significantly associated with good prognosis and survival. CONCLUSIONS: Our findings, derived from a different type of analysis of microarray data, highlight the importance of analyzing coordinated gene expression to find molecular pathways involved in melanoma progression.


Subject(s)
Gene Regulatory Networks , Melanoma/pathology , Tissue Kallikreins/genetics , Tissue Kallikreins/metabolism , Cell Communication/genetics , Disease Progression , Epidermis/growth & development , Epidermis/metabolism , Gene Expression Profiling , Humans , Kallikreins/genetics , Kallikreins/metabolism , Melanoma/genetics , Melanoma/mortality , Oligonucleotide Array Sequence Analysis , Prognosis , RNA, Messenger/metabolism
16.
PLoS One ; 5(11): e13954, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-21085651

ABSTRACT

The retinoblastoma protein (pRb) is a cell cycle regulator inactivated in most human cancers. Loss of pRb function results from mutations in the gene coding for pRb or for any of its upstream regulators. Although pRb is predominantly known as a cell cycle repressor, our data point to additional pRb functions in cell adhesion. Our data show that pRb regulates the expression of a wide repertoire of cell adhesion genes and regulates the assembly of the adherens junctions required for cell adhesion. We conducted our studies in osteoblasts, which depend on both pRb and on cell-to-cell contacts for their differentiation and function. We generated knockout mice in which the RB gene was excised specifically in osteoblasts using the cre-lox P system and found that osteoblasts from pRb knockout mice did not assemble adherens junction at their membranes. pRb depletion in wild type osteoblasts using RNAi also disrupted adherens junctions. Microarrays comparing pRb-expressing and pRb-deficient osteoblasts showed that pRb controls the expression of a number of cell adhesion genes, including cadherins. Furthermore, pRb knockout mice showed bone abnormalities consistent with osteoblast adhesion defects. We also found that pRb controls the function of merlin, a well-known regulator of adherens junction assembly, by repressing Rac1 and its effector Pak1. Using qRT-PCR, immunoblots, co-immunoprecipitation assays, and immunofluorescent labeling, we observed that pRb loss resulted in Rac1 and Pak1 overexpression concomitant with merlin inactivation by Pak1, merlin detachment from the membrane, and adherens junction loss. Our data support a pRb function in cell adhesion while elucidating the mechanism for this function. Our work suggests that in some tumor types pRb inactivation results in both a loss of cell cycle control that promotes initial tumor growth as well as in a loss of cell-to-cell contacts, which contributes to later stages of metastasis.


Subject(s)
Osteoblasts/metabolism , Retinoblastoma Protein/metabolism , 3T3 Cells , Adherens Junctions/genetics , Adherens Junctions/physiology , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Communication/genetics , Cell Communication/physiology , Cell Proliferation , Cells, Cultured , Female , Gene Expression Profiling , Immunoblotting , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , Models, Biological , Neuropeptides/genetics , Neuropeptides/metabolism , Osteoblasts/cytology , Osteogenesis/genetics , Osteogenesis/physiology , Osteosarcoma/genetics , Osteosarcoma/metabolism , RNA Interference , Retinoblastoma Protein/genetics , Retinoblastoma Protein/physiology , Reverse Transcriptase Polymerase Chain Reaction , Skull/embryology , Skull/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
17.
Neurochem Int ; 57(7): 795-803, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20817065

ABSTRACT

Glutamate, the major excitatory transmitter in the vertebrate brain, is involved in neuronal development and synaptic plasticity. Glutamatergic stimulation leads to differential gene expression patterns in neuronal and glial cells. A glutamate-dependent transcriptional control has been established for several genes. However, much less is known about the molecular events that modify the translational machinery upon exposure to this neurotransmitter. In a glial model of cerebellar cultured Bergmann cells, glutamate induces a biphasic effect on [(35)S]-methionine incorporation into proteins that suggests that the elongation phase of protein biosynthesis is the target for regulation. Indeed, after a 15 min exposure to glutamate a transient increase in elongation factor 2 phosphorylation has been reported, an effect mediated through the activation of the elongation factor 2 kinase. In this contribution, we sought to characterize the phosphorylation status of the eukaryotic elongation factor 1A (eEF1A) and the ribosomal transit time under glutamate exposure. A dose-dependent increase in eEF1A phosphorylation was found after a 60 min glutamate treatment; this phenomenon is Ca(2+)/CaM dependent, blocked with Src and phosphatidyl-inositol 3-kinase inhibitors and with rapamicyn. Concomitantly, the ribosomal transit time was increased with a 15 min glutamate exposure. After 60 more minutes, the average time used by the ribosomes to complete a polypeptide chain had almost returned to its initial level. These results strongly suggest that glutamate exerts an exquisite time-dependent translational control in glial cells, a process that might be critical for glia-neuron interactions.


Subject(s)
Glutamic Acid/physiology , Neuroglia/metabolism , Peptide Elongation Factor 1/metabolism , Ribosomes/metabolism , Animals , Cell Communication/genetics , Cells, Cultured , Chick Embryo , Glutamic Acid/metabolism , Peptide Elongation Factor 1/genetics , Phosphorylation/genetics , Protein Biosynthesis , Protein Transport/genetics , Rats , Receptors, Glutamate/physiology , Ribosomes/genetics , Signal Transduction/genetics , Threonine/metabolism , Time Factors , Tumor Cells, Cultured
18.
Exp Cell Res ; 316(19): 3207-26, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20727350

ABSTRACT

Growth and survival of tumors at a site of metastasis involve interactions with stromal cells in the surrounding environment. Stromal cells aid tumor cell growth by producing cytokines as well as by modifying the environment surrounding the tumor through modulation of the extracellular matrix (ECM). Small leucine-rich proteoglycans (SLRPs) are biologically active components of the ECM which can be altered in the stroma surrounding tumors. The influence tumor cells have on stromal cells has been well elucidated. However, little is understood about the effect metastatic cancer cells have on the cell biology and behavior of the local stromal cells. Our data reveal a significant down-regulation in the expression of ECM components such as collagens I, II, III, and IV, and the SLRPs, decorin, biglycan, lumican, and fibromodulin in stromal cells when grown in the presence of two metastatic prostate cancer cell lines PC3 and DU145. Interestingly, TGF-ß down-regulation was observed in stromal cells, as well as actin depolymerization and increased vimentin and α5ß1 integrin expression. MT1-MMP expression was upregulated and localized in stromal cell protrusions which extended into the ECM. Moreover, enhanced stromal cell migration was observed after cross-talk with metastatic prostate tumor cells. Xenografting metastatic prostate cancer cells together with "activated" stromal cells led to increased tumorigenicity of the prostate cancer cells. Our findings suggest that metastatic prostate cancer cells create a metastatic niche by altering the phenotype of local stromal cells, leading to changes in the ECM.


Subject(s)
Cell Differentiation/genetics , Down-Regulation/genetics , Extracellular Matrix/genetics , Fibroblasts/pathology , Prostatic Neoplasms/genetics , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Cell Communication/drug effects , Cell Communication/genetics , Cell Differentiation/drug effects , Cell Movement/drug effects , Coculture Techniques , Cytoskeleton/drug effects , Cytoskeleton/genetics , Down-Regulation/drug effects , Extracellular Matrix/drug effects , Fibroblasts/drug effects , Fibroblasts/enzymology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Neoplasm Metastasis , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Protein Transport/drug effects , Proteoglycans/genetics , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism , Signal Transduction/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Xenograft Model Antitumor Assays
19.
Int J Dev Neurosci ; 28(1): 39-52, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19800961

ABSTRACT

Connexin (Cx) channels and hemichannels are involved in essential processes during nervous system development such as apoptosis, propagation of spontaneous activity and interkinetic nuclear movement. In the first part of this study, we extensively characterized Cx gene and protein expression during retinal histogenesis. We observed distinct spatio-temporal patterns among studied Cx and an overriding, ubiquitous presence of Cx45 in progenitor cells. The role of Cx-mediated communication was assessed by using broad-spectrum (carbenoxolone, CBX) and Cx36/Cx50 channel-specific (quinine) blockers. In vivo application of CBX, but not quinine, caused remarkable reduction in retinal thickness, suggesting changes in cell proliferation/apoptosis ratio. Indeed, we observed a decreased number of mitotic cells in CBX-injected retinas, with no significant changes in the expression of PCNA, a marker for cells in proliferative state. Taken together, our results pointed a pivotal role of Cx45 in the developing retina. Moreover, this study revealed that Cx-mediated communication is essential in retinal histogenesis, particularly in the control of cell proliferation.


Subject(s)
Cell Communication/physiology , Cell Proliferation , Connexins/metabolism , Retina/growth & development , Retina/physiology , Animals , Carbenoxolone/pharmacology , Cell Communication/drug effects , Cell Communication/genetics , Cell Proliferation/drug effects , Central Nervous System Agents/pharmacology , Connexins/antagonists & inhibitors , Connexins/genetics , Gene Expression Regulation, Developmental/drug effects , Neural Pathways/drug effects , Neural Pathways/growth & development , Neural Pathways/physiology , Neuroglia/drug effects , Neuroglia/physiology , Proliferating Cell Nuclear Antigen/metabolism , Quinine/pharmacology , Rats , Rats, Wistar , Retina/drug effects , Retinal Horizontal Cells/drug effects , Retinal Horizontal Cells/physiology , Stem Cells/drug effects , Stem Cells/physiology , Time Factors
20.
São Paulo; s.n; 2010. 141 p.
Thesis in Portuguese | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP, SESSP-IBACERVO | ID: biblio-1079144

ABSTRACT

Escherichia coli enteropatogênica é capaz de causar uma lesão histopatológica no epitélio intestinal conhecida como attaching-effacing (A/E) induzida por proteínas codificadas pelo locus of enterocyte effacement (LEE). EPEC é atualmente classificada em típica e atípica (aEPEC), baseado na presença ou ausência do EPEC adherence plasmid, respectivamente...


Enterophatogenic Escherichia coli is capable of cause a histopathological lession on the intestinal epithelium called attaching-effacing (A/E), triggered by proteins encoded by the locus of enterocyte effacement (LEE).EPEC is currently classified as typical and atypical (aEPEC), based on the presence or absence of the EPEC adherence plasmid, respectively...


Subject(s)
Humans , Cell Communication/genetics , Escherichia coli/classification , Escherichia coli/ultrastructure , Diarrhea/pathology , Escherichia coli Infections/microbiology , Escherichia coli Infections/prevention & control , Real-Time Polymerase Chain Reaction/methods
SELECTION OF CITATIONS
SEARCH DETAIL