Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
R Soc Open Sci ; 8(10): 211088, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34659781

ABSTRACT

The human brain carries out complex tasks and higher functions and is crucial for organismal survival, as it senses both intrinsic and extrinsic environments. Proper brain development relies on the orchestrated development of different precursor cells, which will give rise to the plethora of mature brain cell-types. Within this process, neuronal cells develop closely to and in coordination with vascular cells (endothelial cells (ECs), pericytes) in a bilateral communication process that relies on neuronal activity, attractive or repulsive guidance cues for both cell types and on tight-regulation of gene expression. Translational control is a master regulator of the gene-expression pathway and in particular for neuronal and ECs, it can be localized in developmentally relevant (axon growth cone, endothelial tip cell) and mature compartments (synapses, axons). Herein, we will review mechanisms of translational control relevant to brain development in neurons and ECs in health and disease.

2.
J Cell Sci ; 134(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34313314

ABSTRACT

Ligand-receptor complexes formed at the plasma membrane are internalised via various endocytic pathways that influence the ultimate signalling output by regulating the selection of interaction partners by the complex along the trafficking route. We report that, in differentiated cells, activin A-receptor complexes are internalised via clathrin-mediated endocytosis (CME) and macropinocytosis (MP), whereas in human embryonic stem cells (hESCs) internalisation occurs via CME. We further show that hESCs are devoid of MP, which becomes functional upon differentiation towards endothelial cells through mesoderm mediators. Our results reveal, for the first time, that MP is an internalisation route for activin A in differentiated cells, and that MP is not active in hESCs and is induced as cells differentiate.


Subject(s)
Activins , Endothelial Cells , Cell Differentiation , Embryonic Stem Cells , Endocytosis , Humans
3.
Amino Acids ; 50(2): 279-291, 2018 02.
Article in English | MEDLINE | ID: mdl-29185031

ABSTRACT

Quercetin is a flavonoid presenting cytotoxicity against different cancer cell lines. We hypothesized that its core could serve as a scaffold for generating more potent compounds. A quercetin-alanine bioconjugate was synthesized, its cellular internalization was monitored through confocal microscopy and its cytotoxic activity was explored against ten different cell lines. The bioconjugate consistently illustrated enhanced cytotoxic activity with respect to the parent compound. A threefold enhancement in its cytotoxicity was revealed for HeLa, A549, MCF-7 and LNCaP cells. In silico studies suggested that quercetin-alanine possesses enhanced binding affinity to human estrogen receptor alpha corroborating to its activity to MCF-7, overexpressing this receptor. Spectrofluorimetric, calorimetric and in silico studies revealed that quercetin-alanine binds primarily to Sudlow site I of serum albumin mainly through hydrogen bonding. Through this array of experiments we discovered that the specific compound bears a more refined pharmaceutical profile in contrast to quercetin in terms of cytotoxicity, while at the same time preserves its affinity to serum albumin. Natural products could thus offer a potent scaffold to develop bioconjugates with amplified therapeutic window.


Subject(s)
Antineoplastic Agents/pharmacology , Quercetin/analogs & derivatives , Quercetin/pharmacology , Alanine/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Biological Products/chemistry , Biological Products/metabolism , Biological Products/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Flavonoids/chemistry , Flavonoids/metabolism , Flavonoids/pharmacology , Humans , Inhibitory Concentration 50 , Mice , Molecular Docking Simulation , Protein Binding/drug effects , Quercetin/chemistry , Quercetin/metabolism , Serum Albumin/metabolism , Structure-Activity Relationship
4.
Fertil Steril ; 107(3): 821-830, 2017 03.
Article in English | MEDLINE | ID: mdl-28139237

ABSTRACT

OBJECTIVE: To investigate the expression of long interspersed element (LINE) 1, human endogenous retrovirus (HERV) K10, and short interspersed element-VNTR-Alu element (SVA) retrotransposons in ejaculated human spermatozoa by means of reverse-transcription (RT) polymerase chain reaction (PCR) analysis as well as the potential incorporation of cloned human and mouse active retroelements in human sperm cell genome. DESIGN: Laboratory study. SETTING: University research laboratories and academic hospital. PATIENT(S): Normozoospermic and oligozoospermic white men. INTERVENTION(S): RT-PCR analysis was performed to confirm the retrotransposon expression in human spermatozoa. Exogenous retroelements were tagged with a plasmid containing a green fluorescence (EGFP) retrotransposition cassette, and the de novo retrotransposition events were tested with the use of PCR, fluorescence-activated cell sorting analysis, and confocal microscopy. MAIN OUTCOME MEASURE(S): Retroelement expression in human spermatozoa, incorporation of cloned human and mouse active retroelements in human sperm genome, and de novo retrotransposition events in human spermatozoa. RESULT(S): RT-PCR products of expressed human LINE-1, HERV-K10, and SVA retrotransposons were observed in ejaculated human sperm samples. The incubation of human spermatozoa with either retrotransposition-active human LINE-1 and HERV-K10 or mouse reverse transcriptase-deficient VL30 retrotransposons tagged with an EGFP-based retrotransposition cassette led to EGFP-positive spermatozo; 16.67% of the samples were positive for retrotransposition. The respective retrotransposition frequencies for the LINE-1, HERV-K10, and VL30 retrotransposons in the positive samples were 0.34 ± 0.13%, 0.37 ± 0.17%, and 0.30 ± 0.14% per sample of 10,000 spermatozoa. CONCLUSION(S): Our results show that: 1) LINE-1, HERV-K10, and SVA retrotransposons are transcriptionally expressed in human spermatozoa; 2) cloned active retroelements of human and mammalian origin can be incorporated in human sperm genome; 3) active reverse transcriptases exist in human spermatozoa; and 4) de novo retrotransposition events occur in human spermatozoa.


Subject(s)
Cloning, Molecular , Endopeptidases/genetics , Long Interspersed Nucleotide Elements , Minisatellite Repeats , Oligospermia/genetics , Spermatozoa/metabolism , Animals , Cell Separation/methods , Endopeptidases/biosynthesis , Flow Cytometry , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Male , Mice , Microscopy, Confocal , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transfection , Viral Proteases
5.
Immunology ; 147(1): 73-81, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26447818

ABSTRACT

We have generated three monoclonal cell-penetrating antibodies (CPAbs) from a non-immunized lupus-prone (NZB × NZW)F1 mouse that exhibited high anti-DNA serum titres. These CPAbs are polyreactive because they bind to DNA and other cellular components, and localize mainly in the nucleus of HeLa cells, albeit with a distinct nuclear labelling profile. Herein, we have examined whether DNA-histone complexes (DHC) binding to CPAbs, before cell entry, could modify the cell penetration of CPAbs or their nuclear staining properties. By applying confocal microscopy and image analysis, we found that extracellular binding of purified CPAbs to DHC significantly enhanced their subsequent cell-entry, both in terms of percentages of positively labelled cells and fluorescence intensity (internalized CPAb amount), whereas there was a variable effect on their nuclear staining profile. Internalization of CPAbs, either alone or bound to DHC, remained unaltered after the addition of endocytosis-specific inhibitors at 37° or assay performance at 4°, suggesting the involvement of energy-independent mechanisms in the internalization process. These findings assign to CPAbs a more complex pathogenetic role in systemic lupus erythematosus where both CPAbs and nuclear components are abundant.


Subject(s)
Antibodies, Antinuclear/metabolism , Antibodies, Monoclonal/metabolism , Cell Nucleus/metabolism , Cell-Penetrating Peptides/metabolism , Chromatin/metabolism , DNA/metabolism , Endocytosis , Histones/metabolism , Lupus Erythematosus, Systemic/immunology , Active Transport, Cell Nucleus , Animals , Antibodies, Antinuclear/immunology , Antibodies, Monoclonal/immunology , Cell Nucleus/immunology , Chromatin/immunology , DNA/immunology , Disease Models, Animal , HeLa Cells , Histones/immunology , Humans , Ligands , Lupus Erythematosus, Systemic/genetics , Male , Mice, Inbred NZB , Microscopy, Confocal
6.
Mol Cell ; 54(4): 559-72, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24746698

ABSTRACT

Accumulation of unfolded proteins in the endoplasmic reticulum (ER) initiates IRE1α, ATF6, and PERK cascades, leading to a transcriptional/translational response known as unfolded protein response (UPR). Here we show that VEGF activates UPR mediators through a PLCγ-mediated crosstalk with the mTORC1 complex without accumulation of unfolded proteins in the ER. Activation of ATF6 and PERK contributes to the survival effect of VEGF on endothelial cells (ECs) by positively regulating mTORC2-mediated phosphorylation of AKT on Ser473, which is required for full activity of AKT. Low levels of CHOP allow ECs to evade the proapoptotic effect of this UPR product. Depletion of PLCγ, ATF6, or eIF2α dramatically inhibited VEGF-induced vascularization in mouse Matrigel plugs, suggesting that the ER and the UPR machinery constitute components of the VEGF signaling circuit that regulates EC survival and angiogenesis, extending their role beyond adaptation to ER stress.


Subject(s)
Activating Transcription Factor 6/metabolism , Endoplasmic Reticulum Stress , Endothelial Cells/physiology , Neovascularization, Pathologic , Unfolded Protein Response , Vascular Endothelial Growth Factor A/metabolism , eIF-2 Kinase/metabolism , Activating Transcription Factor 6/genetics , Animals , Cell Survival , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Phospholipase C gamma/metabolism , Phosphorylation , Protein Unfolding , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Unfolded Protein Response/genetics , Vascular Endothelial Growth Factor A/genetics , eIF-2 Kinase/genetics
7.
Mol Cell Oncol ; 1(3): e964024, 2014.
Article in English | MEDLINE | ID: mdl-27308350

ABSTRACT

Vascular endothelial growth factor (VEGF) activates unfolded protein response sensors in the endoplasmic reticulum through phospholipase C gamma (PLCγ)-mediated crosstalk with mammalian target of rapamycin complex 1 (mTORC1). Activation of transcription factor 6 (ATF6) and protein kinase RNA-like endoplasmic reticulum kinase (PERK) activate mTORC2, ensuring maximal endothelial cell survival and angiogenic activity through phosphorylation of AKT on Ser473. As mTORC1 is a metabolic sensor, metabolic signals may be integrated with signals from VEGF in the regulation of angiogenesis.

8.
Metabolism ; 62(5): 622-33, 2013 May.
Article in English | MEDLINE | ID: mdl-23260798

ABSTRACT

Human survival has relied upon the ability to withstand starvation through energy storage, the capacity to fight off infection by a proinflammatory immune response, and the ability to cope with physical stressors by an adaptive stress response. Energy storage, mainly as glycogen in liver and triglycerides in adipose tissue, is regulated by the anabolic actions of insulin. On the other hand, mobilization of stored energy during infection, trauma or stress is served by the temporary inhibition of insulin action (insulin resistance) in target tissues by proinflammatory cytokines and stress hormones. In the current environment, high energy intake, low physical activity, and chronic stress favor the storage of surplus fat in adipose tissue depots that far exceeds their storage capacity and liporegulation. Lipid overload in central fat depots initiates an inflammatory response and adipocyte dysfunction with resultant low-grade systemic inflammation and lipid overflow to peripheral tissues. In turn, proinflammatory cytokines and non-oxidized lipid metabolites, accumulated in liver and muscle cells, activate the mechanism of insulin resistance as would occur in the case of infection or stress. The same factors together with the ensuing insulin resistance further contribute to pancreatic ß-cell dysfunction and ultimately to type 2 diabetes and cardiovascular disease. The present review supports the hypothesis that insulin resistance evolved as a physiological adaptive mechanism in human survival and that the same mechanism is inappropriately activated on a chronic basis in the current environment, leading to the manifestations of the metabolic syndrome.


Subject(s)
Adaptation, Physiological/physiology , Biological Evolution , Environment , Insulin Resistance/physiology , Adaptation, Physiological/genetics , Adipose Tissue/metabolism , Adipose Tissue/physiology , Animals , Gene-Environment Interaction , Humans , Insulin Resistance/genetics , Models, Biological , Stress, Physiological/genetics , Stress, Physiological/physiology
9.
Mol Cancer ; 11: 35, 2012 May 14.
Article in English | MEDLINE | ID: mdl-22583931

ABSTRACT

BACKGROUND: Increased consumption of plant-based diets has been linked to the presence of certain phytochemicals, including polyphenols such as flavonoids. Several of these compounds exert their protective effect via inhibition of tumor angiogenesis. Identification of additional phytochemicals with potential antiangiogenic activity is important not only for understanding the mechanism of the preventive effect, but also for developing novel therapeutic interventions. RESULTS: In an attempt to identify phytochemicals contributing to the well-documented preventive effect of plant-based diets on cancer incidence and mortality, we have screened a set of hitherto untested phytoestrogen metabolites concerning their anti-angiogenic effect, using endothelial cell proliferation as an end point. Here, we show that a novel phytoestrogen, 6-methoxyequol (6-ME), inhibited VEGF-induced proliferation of human umbilical vein endothelial cells (HUVE) cells, whereas VEGF-induced migration and survival of HUVE cells remained unaffected. In addition, 6-ME inhibited FGF-2-induced proliferation of bovine brain capillary endothelial (BBCE) cells. In line with its role in cell proliferation, 6-ME inhibited VEGF-induced phosphorylation of ERK1/2 MAPK, the key cascade responsible for VEGF-induced proliferation of endothelial cells. In this context, 6-ME inhibited in a dose dependent manner the phosphorylation of MEK1/2, the only known upstream activator of ERK1/2. 6-ME did not alter VEGF-induced phosphorylation of p38 MAPK or AKT, compatible with the lack of effect on VEGF-induced migration and survival of endothelial cells. Peri-tumor injection of 6-ME in A-431 xenograft tumors resulted in reduced tumor growth with suppressed neovasularization compared to vehicle controls (P < 0.01). CONCLUSIONS: 6-ME inhibits VEGF- and FGF2-induced proliferation of ECs by targeting the phosphorylation of MEK1/2 and it downstream substrate ERK1/2, both key components of the mitogenic MAPK pathway. Injection of 6-ME in mouse A-431 xenograft tumors results to tumors with decreased neovascularization and reduced tumor volume suggesting that 6-ME may be developed to a novel anti-angiogenic agent in cancer treatment.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Isoflavones/pharmacology , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic , Tumor Burden/drug effects , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemical synthesis , Animals , Cattle , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Equol/analogs & derivatives , Female , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Isoflavones/administration & dosage , Isoflavones/chemical synthesis , MAP Kinase Signaling System/drug effects , Mice , Mice, Nude , Mitosis/drug effects , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Vascular Endothelial Growth Factor A/pharmacology , Xenograft Model Antitumor Assays
10.
Anticancer Res ; 32(4): 1273-81, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22493359

ABSTRACT

BACKGROUND: The epithelial to mesenchymal transition (EMT) has been associated with metastatic dissemination and poor outcome in several solid tumour types. Our aim was to study its incidence and its prognostic significance in cancer of unknown primary (CUP). PATIENTS AND METHODS: One hundred tumour samples of CUP were loaded in tissue microarrays and were studied for immunohistochemical (IHC) expression of E-cadherin, N-cadherin, vimentin, the EMT transcription factor (SNAIL) and the stem cell marker octamer-binding transcription marker 4(OCT4). An EMT phenotype was defined as low expression of E-cadherin, expression of N-cadherin with/without vimentin with concomitant expression of SNAIL, as assessed by percentage of tumour cell staining. RESULTS: Among 100 CUP cases, the histological diagnosis was adenocarcinoma in 55, squamous carcinoma in 20 and undifferentiated carcinoma in 15, with a high grade seen in 46. Therapy consisted of palliative chemotherapy, mostly platinum based. The median progression-free survival and overall survival (OS) were 7 and 12 months respectively. Distributional studies resulted in selection of IHC cut-offs for E-cadherin (negative when expressed in <60% of tumour cells), N-cadherin, vimentin (positive when expressed in ≥40% of tumour cells), SNAIL (positive when stained in ≥80% of tumour cells). An EMT phenotype was observed in 8 cases (8.1%) and was strongly associated with poor OS (median OS EMT(-)=13 months vs. median OS EMT(+)=8 months, p=0.023). When we used staining intensity (H-Score), an EMT phenotype was observed in 16 patients and carried borderline adverse prognostic utility for outcome (median OS 9 vs. 14 months, p=0.07). The presence of the EMT phenotype correlated significantly with male gender, high grade and presence of visceral metastases (χ(2) p<0.05), while EMT mediator expression was correlated to high NOTCH 2/3 expression. Other factors, prognostic for poor survival, were male gender, PS≥2, non-platinum therapy (χ(2) p<0.05). CONCLUSION: EMT is infrequently seen in tumours of CUP. However, an adverse prognostic significance for patient outcome has been identified and may warrant studies of therapeutic targeting.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasms, Unknown Primary/metabolism , Adult , Aged , Aged, 80 and over , Cadherins/metabolism , Female , Humans , Immunohistochemistry , Incidence , Male , Middle Aged , Neoplasms, Unknown Primary/epidemiology , Neoplasms, Unknown Primary/pathology , Octamer Transcription Factor-3/metabolism , Phenotype , Prognosis , Snail Family Transcription Factors , Tissue Array Analysis , Transcription Factors/metabolism , Vimentin/metabolism
11.
Am J Physiol Cell Physiol ; 297(6): C1477-89, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19741200

ABSTRACT

Vascular endothelial growth factor (VEGF) is a key angiogenic factor that regulates proliferation and migration of endothelial cells via phosphorylation of extracellular signal-regulated kinase-1/2 (ERK1/2) and p38, respectively. Here, we demonstrate that VEGF strongly induces the transcription of two dual-specificity phosphatase (DUSP) genes DUSP1 and DUSP5 in endothelial cells. Using fluorescence microscopy, fluorescence lifetime imaging (FLIM), and fluorescence cross-correlation spectroscopy (FCCS), we found that DUSP1/mitogen-activated protein kinases phosphatase-1 (MKP-1) was localized in both the nucleus and cytoplasm of endothelial cells, where it existed in complex with p38 (effective dissociation constant, K(D)(eff), values of 294 and 197 nM, respectively), whereas DUSP5 was localized in the nucleus of endothelial cells in complex with ERK1/2 (K(D)(eff) 345 nM). VEGF administration affected differentially the K(D)(eff) values of the DUSP1/p38 and DUSP5/ERK1/2 complexes. Gain-of-function and lack-of-function approaches revealed that DUSP1/MKP-1 dephosphorylates primarily VEGF-phosphorylated p38, thereby inhibiting endothelial cell migration, whereas DUSP5 dephosphorylates VEGF-phosphorylated ERK1/2 inhibiting proliferation of endothelial cells. Moreover, DUSP5 exhibited considerable nuclear anchoring activity on ERK1/2 in the nucleus, thereby diminishing ERK1/2 export to the cytoplasm decreasing its further availability for activation.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Homeostasis , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Movement/physiology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Drug Interactions , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Tissue Distribution , Transcription, Genetic/physiology , Umbilical Veins/cytology , Up-Regulation/physiology , Vascular Endothelial Growth Factor A/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL