Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
FASEB J ; 33(3): 3112-3128, 2019 03.
Article in English | MEDLINE | ID: mdl-30550356

ABSTRACT

The chondroitin sulfate proteoglycan 4 ( CSPG4) gene encodes a transmembrane proteoglycan (PG) constituting the largest and most structurally complex macromolecule of the human surfaceome. Its transcript shows an extensive evolutionary conservation and, due to the elaborated intracellular processing of the translated protein, it generates an array of glycoforms with the potential to exert variant-specific functions. CSPG4-mediated molecular events are articulated through the interaction with more than 40 putative ligands and the concurrent involvement of the ectodomain and cytoplasmic tail. Alternating inside-out and outside-in signal transductions may thereby be elicited through a tight functional connection of the PG with the cytoskeleton and its regulators. The potential of CSPG4 to influence both types of signaling mechanisms is also asserted by its lateral mobility along the plasma membrane and its intersection with microdomain-restricted internalization and endocytic trafficking. Owing to the multitude of molecular interplays that CSPG4 may engage, and thanks to a differential phosphorylation of its intracellular domain accounted by crosstalking signaling pathways, the PG stands out for its unique capability to affect numerous cellular phenomena, including those purporting pathologic conditions. We discuss here the progresses made in advancing our understanding about the structural-functional bases for the ability of CSPG4 to widely impact on cell behavior, such as to highlight how its multivalency may be exploited to interfere with disease progression.-Tamburini, E., Dallatomasina, A., Quartararo, J., Cortelazzi, B., Mangieri, D., Lazzaretti, M., Perris, R. Structural deciphering of the NG2/CSPG4 proteoglycan multifunctionality.


Subject(s)
Antigens/chemistry , Proteoglycans/chemistry , Amino Acid Sequence , Animals , Antigens/genetics , Antigens/metabolism , Cell Membrane/metabolism , Chondroitin Sulfate Proteoglycans/chemistry , Chondroitin Sulfate Proteoglycans/genetics , Chondroitin Sulfate Proteoglycans/metabolism , Evolution, Molecular , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Molecular , Nerve Regeneration/physiology , Neurites/metabolism , Phylogeny , Protein Interaction Domains and Motifs , Proteoglycans/genetics , Proteoglycans/metabolism
2.
Haematologica ; 104(12): 2465-2481, 2019 12.
Article in English | MEDLINE | ID: mdl-30948493

ABSTRACT

Considering that Aurora kinase inhibitors are currently under clinical investigation in hematologic cancers, the identification of molecular events that limit the response to such agents is essential for enhancing clinical outcomes. Here, we discover a NF-κB-inducing kinase (NIK)-c-Abl-STAT3 signaling-centered feedback loop that restrains the efficacy of Aurora inhibitors in multiple myeloma. Mechanistically, we demonstrate that Aurora inhibition promotes NIK protein stabilization via downregulation of its negative regulator TRAF2. Accumulated NIK converts c-Abl tyrosine kinase from a nuclear proapoptotic into a cytoplasmic antiapoptotic effector by inducing its phosphorylation at Thr735, Tyr245 and Tyr412 residues, and, by entering into a trimeric complex formation with c-Abl and STAT3, increases both the transcriptional activity of STAT3 and expression of the antiapoptotic STAT3 target genes PIM1 and PIM2. This consequently promotes cell survival and limits the response to Aurora inhibition. The functional disruption of any of the components of the trimer NIK-c-Abl-STAT3 or the PIM survival kinases consistently enhances the responsiveness of myeloma cells to Aurora inhibitors. Importantly, concurrent inhibition of NIK or c-Abl disrupts Aurora inhibitor-induced feedback activation of STAT3 and sensitizes myeloma cells to Aurora inhibitors, implicating a combined inhibition of Aurora and NIK or c-Abl kinases as potential therapies for multiple myeloma. Accordingly, pharmacological inhibition of c-Abl together with Aurora resulted in substantial cell death and tumor regression in vivo The findings reveal an important functional interaction between NIK, Abl and Aurora kinases, and identify the NIK, c-Abl and PIM survival kinases as potential pharmacological targets for improving the efficacy of Aurora inhibitors in myeloma.


Subject(s)
Aurora Kinase A/antagonists & inhibitors , Aurora Kinase B/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Multiple Myeloma/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Animals , Apoptosis , Cell Proliferation , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Piperazines/pharmacology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-abl/genetics , Pyrazoles/pharmacology , Pyrroles/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , NF-kappaB-Inducing Kinase
3.
Tumour Biol ; 39(6): 1010428317705757, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28621232

ABSTRACT

Although usually referred to as a structural actin-binding protein, LIM and SH3 domain-containing protein may actually be dynamically involved in the control of a wide spectrum of cellular processes, by virtue of its interaction with several molecular partners. Alongside being ubiquitously expressed in physiological conditions, LIM and SH3 domain-containing protein is overexpressed in a growing number of human cancers, in which it may actively contribute to their aggressiveness by promoting cell proliferation and migration. In view of the recent findings, implicating the protein in cancer progression, we discuss here the most relevant discoveries highlighting the role of this versatile protein in various human tumors. The correlation between LIM and SH3 domain-containing protein expression levels in cancer and the poor outcome and metastatic behavior of tumors denotes the clinical significance of this protein and hints its potential value as a new cancer prognostic or even diagnostic biomarker. This may be decisive not only to optimize existing pharmacological regimes but also to delineate novel, more efficacious therapeutic and/or preventive approaches.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Cytoskeletal Proteins/genetics , LIM Domain Proteins/genetics , Neoplasms/genetics , Adaptor Proteins, Signal Transducing/biosynthesis , Biomarkers, Tumor/biosynthesis , Cell Proliferation/genetics , Cytoskeletal Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , LIM Domain Proteins/biosynthesis , Neoplasms/pathology , Prognosis
4.
Acta Neuropathol ; 132(1): 23-42, 2016 07.
Article in English | MEDLINE | ID: mdl-27026411

ABSTRACT

In adult CNS, nerve/glial-antigen 2 (NG2) is expressed by oligodendrocyte progenitor cells (OPCs) and is an early marker of pericyte activation in pathological conditions. NG2 could, therefore, play a role in experimental autoimmune encephalomyelitis (EAE), a disease associated with increased blood-brain barrier (BBB) permeability, inflammatory infiltrates, and CNS damage. We induced EAE in NG2 knock-out (NG2KO) mice and used laser confocal microscopy immunofluorescence and morphometry to dissect the effect of NG2 KO on CNS pathology. NG2KO mice developed milder EAE than their wild-type (WT) counterparts, with less intense neuropathology associated with a significant improvement in BBB stability. In contrast to WT mice, OPC numbers did not change in NG2KO mice during EAE. Through FACS and confocal microscopy, we found that NG2 was also expressed by immune cells, including T cells, macrophages, and dendritic cells (DCs). Assessment of recall T cell responses to the encephalitogen by proliferation assays and ELISA showed that, while WT and NG2KO T cells proliferated equally to the encephalitogenic peptide MOG35-55, NG2KO T cells were skewed towards a Th2-type response. Because DCs could be responsible for this effect, we assessed their expression of IL-12 by PCR and intracellular FACS. IL-12-expressing CD11c+ cells were significantly decreased in MOG35-55-primed NG2KO lymph node cells. Importantly, in WT mice, the proportion of IL-12-expressing cells was significantly lower in CD11c+ NG2- cells than in CD11c+ NG2+ cells. To assess the relevance of NG2 at immune system and CNS levels, we induced EAE in bone-marrow chimeric mice, generated with WT recipients of NG2KO bone-marrow cells and vice versa. Regardless of their original phenotype, mice receiving NG2KO bone marrow developed milder EAE than those receiving WT bone marrow. Our data suggest that NG2 plays a role in EAE not only at CNS/BBB level, but also at immune response level, impacting on DC activation and thereby their stimulation of reactive T cells, through controlling IL-12 expression.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Dendritic Cells/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Severity of Illness Index , Spinal Cord/immunology , Spinal Cord/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
5.
J Cell Mol Med ; 19(9): 2098-107, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26010680

ABSTRACT

Gynaecological leiomyosarcoma (gLMS) represent a heterogeneous group of soft tissue sarcoma, characterized by rare incidence, high aggressiveness and propensity to infiltrate secondary organs, poor prognosis and lethality, because of the lack of biological mechanisms that underlying their progression and effective pharmaceutical treatments. This study was focused on some of the aspects of progression and dissemination of a subtype of gLMS namely vulvar LMS (vLMS). We therefore used a vulvar LMS-derived cell line namely SK-LMS-1, coupled with in vitro and in vivo assays. We observed that SK-LMS-1 cells have a strong invasive capacity in vitro, through the activity of matrix metalloproteinases 2 and 9, while in vivo these cells induce a strong angiogenic response and disseminate to the chick embryo liver. Therefore, we postulate that metalloproteinases are involved in the spreading behaviour of SK-LMS-1. Further investigations are necessary to better understand the molecular and cellular machinery involved in the progression of this malignancy.


Subject(s)
Leiomyosarcoma/blood supply , Leiomyosarcoma/enzymology , Matrix Metalloproteinases/metabolism , Neovascularization, Pathologic/enzymology , Vulvar Neoplasms/blood supply , Vulvar Neoplasms/enzymology , Angiogenesis Inducing Agents/metabolism , Animals , Cell Line, Tumor , Chickens , Chorioallantoic Membrane/metabolism , Collagen/metabolism , Drug Combinations , Enzyme Activation , Female , Humans , Laminin/metabolism , Leiomyosarcoma/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Proteoglycans/metabolism , Vulvar Neoplasms/pathology
6.
BMC Cancer ; 15: 352, 2015 May 03.
Article in English | MEDLINE | ID: mdl-25935541

ABSTRACT

BACKGROUND: Tumour relapse is recognized to be the prime fatal burden in patients affected by head and neck squamous cell carcinoma (HNSCC), but no discrete molecular trait has yet been identified to make reliable early predictions of tumour recurrence. Expression of cell surface proteoglycans (PGs) is frequently altered in carcinomas and several of them are gradually emerging as key prognostic factors. METHODS: A PG expression analysis at both mRNA and protein level, was pursued on primary lesions derived from 173 HNSCC patients from whom full clinical history and 2 years post-surgical follow-up was accessible. Gene and protein expression data were correlated with clinical traits and previously proposed tumour relapse markers to stratify high-risk patient subgroups. RESULTS: HNSCC lesions were indeed found to exhibit a widely aberrant PG expression pattern characterized by a variable expression of all PGs and a characteristic de novo transcription/translation of GPC2, GPC5 and NG2/CSPG4 respectively in 36%, 72% and 71% on 119 cases. Importantly, expression of NG2/CSPG4, on neoplastic cells and in the intralesional stroma (Hazard Ratio [HR], 6.76, p = 0.017) was strongly associated with loco-regional relapse, whereas stromal enrichment of SDC2 (HR, 7.652, p = 0.007) was independently tied to lymphnodal infiltration and disease-related death. Conversely, down-regulated SDC1 transcript (HR, 0.232, p = 0.013) uniquely correlated with formation of distant metastases. Altered expression of PGs significantly correlated with the above disease outcomes when either considered alone or in association with well-established predictors of poor prognosis (i.e. T classification, previous occurrence of precancerous lesions and lymphnodal metastasis). Combined alteration of all three PGs was found to be a reliable predictor of shorter survival. CONCLUSIONS: An unprecedented PG-based prognostic portrait is unveiled that incisively diversifies disease course in HNSCC patients beyond the currently known clinical and molecular biomarkers.


Subject(s)
Antigens/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Neoplasm Recurrence, Local/metabolism , Proteoglycans/metabolism , Adult , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/surgery , Chondroitin Sulfate Proteoglycans/metabolism , Female , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/surgery , Humans , Kaplan-Meier Estimate , Male , Membrane Proteins/metabolism , Middle Aged , Mouth/metabolism , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/prevention & control , Proportional Hazards Models , Syndecan-2/metabolism , Treatment Outcome
7.
J Pers Med ; 14(2)2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38392627

ABSTRACT

Tumor-stroma crosstalk promotes the adaptation of cancer cells to the local microenvironment and sustains their growth. We assessed the quantitative and qualitative impact of intralesional stroma on clinic-pathological features and the prognosis of poorly cohesive gastric cancer (PCGC) variants. Tissue microarrays including 75 PCGC specimens were immunostained for cytokeratin 8/18 and α-smooth muscle actin to assess the relative proportion of neoplastic cells versus stromal components and the cases were subsequently divided into stroma-rich (SR) and stroma-poor (SP) tumors. Stromal status is significantly associated with the depth of tumor invasion. Patient survival rate was found to be higher in the SP compared to the SR tumor group and, hence, abundant stroma was identified as a significant risk factor in univariable analysis but had no independent prognostic impact. We also investigated the mRNA levels of KRT8 and the associated transcriptional signatures using the molecular data of 82 PCGC cases divided into KRT8-high and KRT8-low groups. KRT8-high tumors were enriched in proteins localized in the extracellular compartment and their expression levels correlated with longer survival in the KRT8-high group and shorter overall survival in the KRT8-low group. Comprehensively, we find that relative intralesional stromal content is a marker of aggressiveness in PCGC tumors and that extracellular proteins characterize functionally and clinically different PCGC subgroups.

8.
Angiogenesis ; 16(2): 309-27, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23124902

ABSTRACT

Sprouting of angiogenic perivascular cells is thought to be highly dependent upon autocrine and paracrine growth factor stimulation. Accordingly, we report that corneal angiogenesis induced by ectopic FGF implantation is strongly impaired in NG2/CSPG4 proteoglycan (PG) null mice known to harbour a putative deficit in pericyte proliferation/mobilization. Conversely, no significant differences were seen between wild type and knockout corneas when VEGF was used as an angiocrine factor. Perturbed responsiveness of NG2-deficient pericytes to paracrine and autocrine stimulation by several FGFs could be confirmed in cells isolated from NG2 null mice, while proliferation induced by other growth factors was equivalent in wild type and knockout cells. Identical results were obtained after siRNA-mediated knock-down of NG2 in human smooth muscle-like cell lines, as also demonstrated by the decreased levels of FGF receptor phosphorylation detected in these NG2 deprived cells. Binding assays with recombinant proteins and molecular interactions examined on live cells asserted that FGF-2 bound to NG2 in a glycosaminoglycan-independent, core protein-mediated manner and that the PG was alone capable of retaining FGF-2 on the cell membrane for subsequent receptor presentation. The use of dominant-negative mutant cells, engineered by combined transduction of NG2 deletion constructs and siRNA knock-down of the endogenous PG, allowed us to establish that the FGF co-receptor activity of NG2 is entirely mediated by its extracellular portion. In fact, forced overexpression of the NG2 ectodomain in human smooth muscle-like cells increased their FGF-2-induced mitosis and compensated for low levels of FGF receptor surface expression, in a manner equivalent to that produced by overexpression of the full-length NG2. Upon FGF binding, the cytoplasmic domain of NG2 is phosphorylated, but there is no evidence that this event elicits signal transductions that could bypass the FGFR-mediated ones. Pull-down experiments, protein-protein binding assays and flow cytometry FRET coherently revealed an elective ligand-independent association of NG2 with FGFR1 and FGFR3. The NG2 cooperation with these receptors was also corroborated functionally by the outcome of FGF-2 treatments of cells engineered to express diverse NG2/FGFR combinations. Comprehensively, the findings suggest that perivascular NG2 may serve as a dual modulator of the availability/accessibility of FGF at the cell membrane, as well as the resulting FGFR transducing activity.


Subject(s)
Antigens/metabolism , Fibroblast Growth Factors/metabolism , Mitogens/metabolism , Pericytes/metabolism , Proteoglycans/metabolism , Animals , Cornea/blood supply , Fluorescence Resonance Energy Transfer , Fluorescent Antibody Technique , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Signal Transduction
9.
Cell Mol Life Sci ; 69(4): 553-79, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21964924

ABSTRACT

Proteoglycans (PGs), a family of complex post-translationally sculptured macromolecules, are fundamental regulators of most normal and aberrant cellular functions. The unparalleled structural-functional diversity of PGs endows them with the ability to serve as critical mediators of the tumor cells' interaction with the host microenvironment, while directly contributing to the organization and dynamic remodeling of this milieu. Despite their indisputable importance during embryonic development and in the adult organism, and their frequent dysregulation in tumor lesions, their precise involvement in tumorigenesis awaits a more decisive demonstration. Particularly challenging is to ascertain to what extent selected PGs may catalyze tumor progression and to what extent they may inhibit it, implying antithetic functions of individual PGs. Integrated efforts are needed to consolidate the routine use of PGs in the clinical monitoring of cancer patients and to broaden the exploitation of these macromolecules as therapeutic targets. Several PGs have the required attributes to be contemplated as effective antigens for immunotherapeutic approaches, while the tangible results obtained in recent clinical trials targeting the NG2/CSPG4 transmembrane PG urge further development of PG-based cancer treatment modalities.


Subject(s)
Neoplasms/metabolism , Proteoglycans/metabolism , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic , Disease Progression , Humans , Immunotherapy , Neoplasms/immunology , Neoplasms/pathology
10.
Leukemia ; 37(8): 1671-1685, 2023 08.
Article in English | MEDLINE | ID: mdl-37386079

ABSTRACT

Resistance to tyrosine kinase inhibitors (TKIs) remains a clinical challenge in Ph-positive variants of chronic myeloid leukemia. We provide mechanistic insights into a previously undisclosed MEK1/2/BCR::ABL1/BCR/ABL1-driven signaling loop that may determine the efficacy of arsenic trioxide (ATO) in TKI-resistant leukemic patients. We find that activated MEK1/2 assemble into a pentameric complex with BCR::ABL1, BCR and ABL1 to induce phosphorylation of BCR and BCR::ABL1 at Tyr360 and Tyr177, and ABL1, at Thr735 and Tyr412 residues thus provoking loss of BCR's tumor-suppression functions, enhanced oncogenic activity of BCR::ABL1, cytoplasmic retention of ABL1 and consequently drug resistance. Coherently, pharmacological blockade of MEK1/2 induces dissociation of the pentameric MEK1/2/BCR::ABL1/BCR/ABL1 complex and causes a concurrent BCRY360/Y177, BCR::ABL1Y360/Y177 and cytoplasmic ABL1Y412/T735 dephosphorylation thereby provoking the rescue of the BCR's anti-oncogenic activities, nuclear accumulation of ABL1 with tumor-suppressive functions and consequently, growth inhibition of the leukemic cells and an ATO sensitization via BCR-MYC and ABL1-p73 signaling axes activation. Additionally, the allosteric activation of nuclear ABL1 was consistently found to enhance the anti-leukemic effects of the MEK1/2 inhibitor Mirdametinib, which when combined with ATO, significantly prolonged the survival of mice bearing BCR::ABL1-T315I-induced leukemia. These findings highlight the therapeutic potential of MEK1/2-inhibitors/ATO combination for the treatment of TKI-resistant leukemia.


Subject(s)
Fusion Proteins, bcr-abl , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Mice , Animals , Arsenic Trioxide/pharmacology , Fusion Proteins, bcr-abl/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Drug Resistance, Neoplasm , Apoptosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
11.
J Pers Med ; 12(12)2022 Dec 03.
Article in English | MEDLINE | ID: mdl-36556227

ABSTRACT

Loss of CDH1/Cadherin-1 is a common step towards the acquisition of an abnormal epithelial phenotype. In gastric cancer (GC), mutation and/or downregulation of CDH1/Cadherin-1 is recurrent in sporadic and hereditary diffuse GC type. To approach the molecular events downstream of CDH1/Cadherin-1 alterations and their relevance in gastric carcinogenesis, we queried public databases for genetic and DNA methylation data in search of molecular signatures with a still-uncertain role in the pathological mechanism of GC. In all GC subtypes, modulated genes correlating with CDH1/Cadherin-1 aberrations are associated with stem cell and epithelial-to-mesenchymal transition pathways. A higher level of genes upregulated in CDH1-mutated GC cases is associated with reduced overall survival. In the diffuse GC (DGC) subtype, genes downregulated in CDH1-mutated compared to cases with wild type CDH1/Cadherin-1 resulted in being strongly intertwined with the DREAM complex. The inverse correlation between hypermethylated CpGs and CDH1/Cadherin-1 transcription in diverse subtypes implies a common epigenetic program. We identified nonredundant protein-encoding isoforms of 22 genes among those differentially expressed in GC compared to normal stomach. These unique proteins represent potential agents involved in cell transformation and candidate therapeutic targets. Meanwhile, drug-induced and CDH1/Cadherin-1 mutation-related gene expression comparison predicts FIT, GR-127935 hydrochloride, amiodarone hydrochloride in GC and BRD-K55722623, BRD-K13169950, and AY 9944 in DGC as the most effective treatments, providing cues for the design of combined pharmacological treatments. By integrating genetic and epigenetic aspects with their expected functional outcome, we unveiled promising targets for combinatorial pharmacological treatments of GC.

12.
Neurobiol Dis ; 43(3): 678-89, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21679768

ABSTRACT

Experimentally induced autoimmune encephalomyelitis (EAE) in mice provides an animal model that shares many features with human demyelinating diseases such as multiple sclerosis (MS). To what extent the cerebral cortex is affected by the process of demyelination and how the corollary response of the oligodendrocyte lineage is explicated are still not completely known aspects of EAE. By performing a detailed in situ analysis of expression of myelin and oligodendrocyte markers we have identified areas of subpial demyelination in the cerebral cortex of animals with conventionally induced EAE conditions. On EAE-affected cerebral cortices, the distribution and relative abundance of cells of the oligodendrocyte lineage were assessed and compared with control mouse brains. The analysis demonstrated that A2B5(+) glial restricted progenitors (GRPs) and NG2(+)/PDGFR-α(+) oligodendrocyte precursor cells (OPCs) were increased in number during "early" disease, 20 days post MOG immunization, whereas in the "late" disease, 39 days post-immunization, they were strongly diminished, and there was an accompanying reduction in NG2(+)/O4(+) pre-oligodendrocytes and GST-π mature oligodendrocytes. These results, together with the observed steady-state amount of NG2(-)/O4(+) pre-myelinating oligodendrocytes, suggested that oligodendroglial precursors attempted to compensate for the progressive loss of myelin, although these cells appeared to fail to complete the last step of their differentiation program. Our findings confirm that this chronic model of EAE reproduces the features of neocortex pathology in progressive MS and suggest that, despite the proliferative response of the oligodendroglial precursors, the failure to accomplish final differentiation may be a key contributing factor to the impaired remyelination that characterizes these demyelinating conditions.


Subject(s)
Adult Stem Cells/pathology , Cerebral Cortex/pathology , Demyelinating Diseases/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Oligodendroglia/pathology , Adult Stem Cells/metabolism , Animals , Cell Lineage/physiology , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Nerve Fibers, Myelinated/pathology , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism
13.
BMC Cancer ; 10: 550, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20939912

ABSTRACT

BACKGROUND: In the last years, the transmembrane proteoglycan NG2 has gained interest as a therapeutic target for the treatment of diverse tumor types, including gliomas, because increases of its expression correlate with dismal prognosis. NG2 has been shown to function as a co-receptor for PDGF ligands whose aberrant expression is common in gliomas. We have recently generated a glioma model based on the overexpression of PDGF-B in neural progenitors and here we investigated the possible relevance of NG2 during PDGF-driven gliomagenesis. METHODS: The survival curves of NG2-KO mice overexpressing PDGF-B were compared to controls by using a Log-rank test. The characteristics of tumors induced in NG2-KO were compared to those of tumors induced in wild type mice by immunostaining for different cell lineage markers and by transplantation assays in adult mice. RESULTS: We showed that the lack of NG2 does not appreciably affect any of the characterized steps of PDGF-driven brain tumorigenesis, such as oligodendrocyte progenitor cells (OPC) induction, the recruitment of bystander OPCs and the progression to full malignancy, which take place as in wild type animals. CONCLUSIONS: Our analysis, using both NG2-KO mice and a miRNA based silencing approach, clearly demonstrates that NG2 is not required for PDGF-B to efficiently induce and maintain gliomas from neural progenitors. On the basis of the data obtained, we therefore suggest that the role of NG2 as a target molecule for glioma treatment should be carefully reconsidered.


Subject(s)
Antigens/physiology , Brain Neoplasms/pathology , Glioma/pathology , Proteoglycans/physiology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Antigens/genetics , Brain Neoplasms/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Gene Silencing , Glioma/genetics , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/metabolism , Oligodendroglia/cytology , Proteoglycans/genetics , Retroviridae , Stem Cells
14.
J Cell Mol Med ; 13(9B): 3151-73, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19220578

ABSTRACT

Aggrecan is a component of the CNS extracellular matrix (ECM) and we show here that the three primary alternative spliced transcripts of the aggrecan gene found in cartilage are also present in the adult CNS. Using a unique panel of core protein-directed antibodies against human aggrecan we further show that different aggrecan isoforms are deposited in perineuronal nets (PNNs) and neuropil ECM of Brodmann's area 6 of the human adult cerebral cortex. According to their distribution pattern, the identified cortical aggrecan isoforms were subdivided into five clusters spanning from cluster 1, comprised isoforms that appeared widespread throughout the cortex, to cluster 5, which was an aggrecan-free subset. Comparison of brain and cartilage tissues showed a different relative abundance of aggrecan isoforms, with cartilage-specific isoforms characterizing cluster 5, and PNN-associated isoforms lacking keratan sulphate chains. In the brain, isoforms of cluster 1 were disclosed in PNNs surrounding small-medium interneurons of layers II-V, small-medium pyramidal neurons of layers III and V and large interneurons of layer VI. Aggrecan PNNs enveloped both neuron bodies and neuronal processes, encompassing pre-terminal nerve fibres, synaptic boutons and terminal processes of glial cells and aggrecan was also observed in continuous 'coats' associated with satellite, neuron-associated cells of a putative glial nature. Immunolabelling for calcium-binding proteins and glutamate demonstrated that aggrecan PNNs were linked to defined subsets of cortical interneurons and pyramidal cells. We suggest that in the human cerebral cortex, discrete, layer-specific PNNs are assembled through the participation of selected aggrecan isoforms that characterize defined subsets of cortical neurons.


Subject(s)
Aggrecans/chemistry , Cerebral Cortex/metabolism , Gene Expression Regulation , Neurons/metabolism , Brain/metabolism , Cartilage/metabolism , Chondroitin/chemistry , Humans , Hybridomas/metabolism , Immunoassay , Microscopy, Confocal , Models, Biological , Oligodendroglia/metabolism , Protein Isoforms , Synapses/metabolism
15.
J Cell Biol ; 158(6): 1133-44, 2002 Sep 16.
Article in English | MEDLINE | ID: mdl-12235127

ABSTRACT

Osteoclast (OC) precursors migrate to putative sites of bone resorption to form functionally active, multinucleated cells. The preOC FLG 29.1 cells, known to be capable of irreversibly differentiating into multinucleated OC-like cells, displayed several features of primary OCs, including expression of specific integrins and the hyaluronan (HA) receptor CD44. OC-like FLG 29.1 cells adhered to and extensively migrated through membranes coated with fibronectin, vitronectin, and laminins, but, although strongly binding to HA, totally failed to move on this substrate. Moreover, soluble HA strongly inhibited OC-like FLG 29.1 cell migration on the permissive matrix substrates, and this behavior was dependent on its engagement with CD44, as it was fully restored by function-blocking anti-CD44 antibodies. HA did not modulate the cell-substrate binding affinity/avidity nor the expression levels of the corresponding integrins. MMP-9 was the major secreted metalloproteinase used by OC-like FLG 29.1 cells for migration, because this process was strongly inhibited by both TIMP-1 and GM6001, as well as by MMP-9-specific antisense oligonucleotides. After HA binding to CD44, a strong down-regulation of MMP-9 mRNA and protein was detected. These findings highlight a novel role of the HA-CD44 interaction in the context of OC-like cell motility, suggesting that it may act as a stop signal for bone-resorbing cells.


Subject(s)
Cell Movement , Down-Regulation , Hyaluronan Receptors/physiology , Hyaluronic Acid/metabolism , Matrix Metalloproteinase 9/metabolism , Osteoclasts/physiology , Antibodies, Monoclonal/pharmacology , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Count , Cells, Cultured , Dipeptides/pharmacology , Extracellular Matrix/metabolism , Filaggrin Proteins , Humans , Hyaluronan Receptors/metabolism , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Oligonucleotides, Antisense/pharmacology , Osteoclasts/cytology , Protease Inhibitors/pharmacology , Tissue Inhibitor of Metalloproteinase-1/pharmacology
16.
Methods Mol Biol ; 522: 221-50, 2009.
Article in English | MEDLINE | ID: mdl-19247614

ABSTRACT

Cell adhesion and cell migration are two primary cellular phenomena for which in vitro approaches may be exploited to effectively dissect the individual events and underlying molecular mechanisms. The use of assays dedicated to the analysis of cell adhesion and migration in vitro also afford an efficient way of conducting larger basic and applied research screenings on the factors affecting these processes and are potentially exploitable in the context of routine diagnostic, prognostic, and predictive tests in the biological and medical fields. Therefore, there is a longstanding continuum in the interest in devising more rationale such assays and major contributions in this direction have been provided by the advent of procedures based on fluorescence cell tagging, the design of instruments capable of detecting fluorescent signals with high sensitivity, and informatic tools allowing sophisticated elaboration of data generated through these instruments. In this report, we describe three representative fluorescence-based model assays for the qualitative and quantitative assessment of cell adhesion and cell locomotion in static and dynamic conditions. The assays are easily performed, accurate and reproducible, and can be automated for high-to-medium throughput screenings of cell behavior in vitro. Performance of the assays involves the use of certain dedicated disposable accessories, which are commercially available, and a few instruments that, due to their versatility, can be regarded as constituents of a more generic laboratory setup.


Subject(s)
Cell Adhesion , Cell Movement , Fluorescence
17.
Cereb Cortex ; 18(3): 553-62, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17576749

ABSTRACT

Six3, a homeodomain-containing transcriptional regulator belonging to the Six/so family, shows a defined spatiotemporal expression pattern in the developing murine telencephalon, suggesting that it may control the development of specific subsets of neural progenitors. We find that retrovirus-mediated misexpression of Six3 causes clonal expansion of isolated cortical progenitor cells by shortening their cell cycle and by prolonging their amplification period, while maintaining them in an immature precursor state. Our results show that the observed effects exerted by Six3 overexpression in mammalian brain depend strictly on the integrity of its DNA-binding domain, suggesting that Six3 action likely relies exclusively on its transcriptional activity. In vivo upregulation of Six3 expression in single progenitor cells of the embryonic telencephalon keeps them in an undifferentiated state. Our observations point to a role of Six3 in the control of the subtle equilibrium between proliferation and differentiation of defined precursor populations during mammalian neurogenesis.


Subject(s)
Eye Proteins/physiology , Homeodomain Proteins/physiology , Nerve Tissue Proteins/physiology , Neurons/physiology , Stem Cells/physiology , Telencephalon/physiology , Animals , Base Sequence , Cells, Cultured , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Eye Proteins/biosynthesis , Eye Proteins/genetics , Eye Proteins/metabolism , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Molecular Sequence Data , NIH 3T3 Cells , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Telencephalon/cytology , Telencephalon/metabolism , Up-Regulation/physiology , Homeobox Protein SIX3
18.
Int J Dev Biol ; 52(7): 993-8, 2008.
Article in English | MEDLINE | ID: mdl-18956330

ABSTRACT

The chondroitin sulfate proteoglycan (PG) PG-M/versican is known to be a primary component of the vertebrate embryonic extracellular matrix and, in the mouse, functional abrogation of the versican gene leads to severe cardiovascular malformations and embryonic lethality. In order to provide a means for approaching the study of the role of versican during embryogenesis, we have cloned the Xenopus versican cDNA and we have performed in situ hybridization on embryos at different stages of development. We showed maternal Xversican transcription, as well as a previously undocumented early expression of the PG during gastrulation and neurulation. At later stages of development, spatial transcription of Xversican correlates with the patterns of migrating neural crest cells (NCC) and it is expressed in embryonic regions representing the final sites of arrest of NCC. Xversican mRNA was also detected in a subpopulation of trunk NCC migrating into the fin, in tissues flanking the trunk NCC ventral migratory pathway and in post-migratory cranial skeletogenic NCC. Further embryonic sites expressing Xversican were the pronephros, pronephric ducts, heart anlage and branchial pouches. These findings constitute the basis for future studies aimed at clarifying unresolved aspects of versican function during embryogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Versicans/genetics , Xenopus/embryology , Animals , Cloning, Molecular , DNA, Complementary/genetics , Embryo, Nonmammalian , Heart/embryology , In Situ Hybridization , Kidney Tubules/embryology , Kidney Tubules/metabolism , Neural Crest/cytology , Neural Crest/metabolism , RNA, Messenger/metabolism , Transcription, Genetic , Versicans/metabolism , Xenopus/genetics , Xenopus/metabolism
19.
Cancers (Basel) ; 11(7)2019 Jul 09.
Article in English | MEDLINE | ID: mdl-31323949

ABSTRACT

5-methyl cytosine (5mC) is a key epigenetic mark entwined with gene expression and the specification of cellular phenotypes. Its distribution around gene promoters sets a barrier for transcriptional enhancers or inhibitor proteins binding to their target sequences. As a result, an additional level of regulation is added to the signals that organize the access to the chromatin and its structural components. The tumor suppressor gene RASSF1A is a microtubule-associated and multitasking scaffold protein communicating with the RAS pathway, estrogen receptor signaling, and Hippo pathway. RASSF1A action stimulates mitotic arrest, DNA repair and apoptosis, and controls the cell cycle and cell migration. De novo methylation of the RASSF1A promoter has received much attention due to its increased frequency in most cancer types. RASSF1A methylation is preceded by histones modifications and could represent an early molecular event in cell transformation. Accordingly, RASSF1A methylation is proposed as an epigenetic candidate marker in many cancer types, even though an inverse correlation of methylation and expression remains to be fully ascertained. Some findings indicate that the epigenetic abrogation of RASSF1A can promote the alternative expression of the putative oncogenic isoform RASSF1C. Understanding the complexity and significance of RASSF1A methylation is instrumental for a more accurate determination of its biological and clinical role. The review covers the molecular events implicated in RASSF1A methylation and gene silencing and provides a deeper view into the significance of the RASSF1A methylation patterns in a number of gastrointestinal cancer types.

20.
Connect Tissue Res ; 49(3): 225-9, 2008.
Article in English | MEDLINE | ID: mdl-18661348

ABSTRACT

Proteoglycans (PGs) as a whole, or when considering their GAG chains as single entities, are emerging as key regulators of tumor progression. Expectations on using them as putative prognostic markers and potential therapeutic targets are increasing coincidentally. Due to the multitude of biological roles that they may invest and the ample spectrum of cellular processes that they may control, we still need to learn better how they regulate phenomena such as intracellular signaling, proliferation, apoptosis, motility, and drug resistance. Depending on the type, their expression pattern, and the accessibility of their molecular ligands, PGs can either promote or inhibit tumorigenesis. The structural and functional diversity of PGs coupled with their ubiquitous abundance place them at the crossroads of many critical steps within the metastatic cascade. As this phenomenon is the pivotal factor for patient survivals, particular attention should be given to the understanding of how PGs govern metastasis formation.


Subject(s)
Antigens/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , Proteoglycans/metabolism , Animals , Cell Proliferation , Humans , Immunotherapy , Neoplasms/therapy , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL