Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Life (Basel) ; 13(5)2023 May 16.
Article in English | MEDLINE | ID: mdl-37240836

ABSTRACT

The pore-forming subunits (α subunits) of voltage-gated sodium channels (VGSC) are encoded in humans by a family of nine highly conserved genes. Among them, SCN1A, SCN2A, SCN3A, and SCN8A are primarily expressed in the central nervous system. The encoded proteins Nav1.1, Nav1.2, Nav1.3, and Nav1.6, respectively, are important players in the initiation and propagation of action potentials and in turn of the neural network activity. In the context of neurological diseases, mutations in the genes encoding Nav1.1, 1.2, 1.3 and 1.6 are responsible for many forms of genetic epilepsy and for Nav1.1 also of hemiplegic migraine. Several pharmacological therapeutic approaches targeting these channels are used or are under study. Mutations of genes encoding VGSCs are also involved in autism and in different types of even severe intellectual disability (ID). It is conceivable that in these conditions their dysfunction could indirectly cause a certain level of neurodegenerative processes; however, so far, these mechanisms have not been deeply investigated. Conversely, VGSCs seem to have a modulatory role in the most common neurodegenerative diseases such as Alzheimer's, where SCN8A expression has been shown to be negatively correlated with disease severity.

2.
Biomater Adv ; 148: 213355, 2023 May.
Article in English | MEDLINE | ID: mdl-36893487

ABSTRACT

Visceral myopathy (VSCM) is a rare genetic disease, orphan of pharmacological therapy. VSCM diagnosis is not always straightforward due to symptomatology similarities with mitochondrial or neuronal forms of intestinal pseudo-obstruction. The most prevalent form of VSCM is associates with variants in the gene ACTG2, encoding the protein gamma-2 actin. Overall, VSCM is a mechano-biological disorder, in which different genetic variants lead to similar alterations to the contractile phenotype of enteric smooth muscles, resulting in the emergence of life-threatening symptoms. In this work we analyzed the morpho-mechanical phenotype of human dermal fibroblasts from patients affected with VSCM, demonstrating that they retain a clear signature of the disease when compared with different controls. We evaluated several biophysical traits of fibroblasts, and we show that a measure of cellular traction forces can be used as a non-specific biomarker of the disease. We propose that a simple assay based on traction forces could be designed to provide a valuable support for clinical decision or pre-clinical research.


Subject(s)
Intestinal Pseudo-Obstruction , Humans , Intestinal Pseudo-Obstruction/diagnosis , Intestinal Pseudo-Obstruction/genetics , Intestinal Pseudo-Obstruction/metabolism , Actins/genetics , Actins/metabolism , Muscle Contraction , Phenotype , Muscle, Smooth/metabolism
3.
Front Neurosci ; 16: 966019, 2022.
Article in English | MEDLINE | ID: mdl-36148145

ABSTRACT

Transmissible spongiform encephalopathies (TSEs), or prion diseases, are progressive neurodegenerative disorders of the central nervous system that affect humans and animals as sporadic, inherited, and infectious forms. Similarly to Alzheimer's disease and other neurodegenerative disorders, any attempt to reduce TSEs' lethality or increase the life expectancy of affected individuals has been unsuccessful. Typically, the onset of symptoms anticipates the fatal outcome of less than 1 year, although it is believed to be the consequence of a decades-long process of neuronal death. The duration of the symptoms-free period represents by itself a major obstacle to carry out effective neuroprotective therapies. Prions, the infectious entities of TSEs, are composed of a protease-resistant protein named prion protein scrapie (PrPSc) from the prototypical TSE form that afflicts ovines. PrPSc misfolding from its physiological counterpart, cellular prion protein (PrPC), is the unifying pathogenic trait of all TSEs. PrPSc is resistant to intracellular turnover and undergoes amyloid-like fibrillation passing through the formation of soluble dimers and oligomers, which are likely the effective neurotoxic entities. The failure of PrPSc removal is a key pathogenic event that defines TSEs as proteopathies, likewise other neurodegenerative disorders, including Alzheimer's, Parkinson's, and Huntington's disease, characterized by alteration of proteostasis. Under physiological conditions, protein quality control, led by the ubiquitin-proteasome system, and macroautophagy clears cytoplasm from improperly folded, redundant, or aggregation-prone proteins. There is evidence that both of these crucial homeostatic pathways are impaired during the development of TSEs, although it is still unclear whether proteostasis alteration facilitates prion protein misfolding or, rather, PrPSc protease resistance hampers cytoplasmic protein quality control. This review is aimed to critically analyze the most recent advancements in the cause-effect correlation between PrPC misfolding and proteostasis alterations and to discuss the possibility that pharmacological restoring of ubiquitin-proteasomal competence and stimulation of autophagy could reduce the intracellular burden of PrPSc and ameliorate the severity of prion-associated neurodegeneration.

4.
Biomedicines ; 9(9)2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34572381

ABSTRACT

Hypersensitivity pneumonitis (HP) is a diffuse interstitial lung disease (ILD) caused by the inhalation of a variety of antigens in susceptible individuals. Patients with fibrotic HP (fHP) may show histopathological and radiological manifestations similar to patients with idiopathic pulmonary fibrosis (usual interstitial pneumonia-like pattern of fibrosis) that are associated with a worse prognosis. We describe here the establishment and characterization of a fibroblastic cell line derived from the broncho-alveolar lavage (BAL) of a patient with fHP, a 53 year old man who presented at our Pneumology Unit with cough and dyspnea. The fHP diagnosis was based on international criteria and multidisciplinary discussion. Primary fibroblasts were expanded in vitro until passage 36. These fibroblasts displayed morpho/phenotypical features of myofibroblasts, showing high positivity for α-smooth muscle actin, type I collagen, and fibronectin as determined by quantitative RT-PCR and cyto-fluorographic analysis. Cytogenetic analyses further evidenced trisomy of chromosome 10, which interestingly harbors the FGF2R gene. To our knowledge, this is the first fibroblastic cell line derived from an fHP patient and might, therefore, represent a suitable tool to model the disease in vitro. We preliminarily assessed here the activity of pirfenidone, further demonstrating a consistent inhibition of cells growth by this antifibrotic drug.

5.
Int J Mol Sci ; 22(13)2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34201814

ABSTRACT

High-risk neuroblastoma (HR-NB) still remains the most dangerous tumor in early childhood. For this reason, the identification of new therapeutic approaches is of fundamental importance. Recently, we combined the conventional pharmacological approach to NB, represented by cisplatin, with fendiline hydrochloride, an inhibitor of several transporters involved in multidrug resistance of cancer cells, which demonstrated an enhancement of the ability of cisplatin to induce apoptosis. In this work, we co-administrated acetazolamide, a carbonic anhydrase isoform IX (CAIX) inhibitor which was reported to increase chemotherapy efficacy in various cancer types, to the cisplatin/fendiline approach in SKNBE2 xenografts in NOD-SCID mice with the aim of identifying a novel and more effective treatment. We observed that the combination of the three drugs increases more than twelvefold the differences in the cytotoxic activity of cisplatin alone, leading to a remarkable decrease of the expression of malignancy markers. Our conclusion is that this approach, based on three FDA-approved drugs, may constitute an appropriate improvement of the pharmacological approach to HR-NB.


Subject(s)
Acetazolamide/pharmacology , Antineoplastic Agents/pharmacology , Calcium Channel Blockers/pharmacology , Carbonic Anhydrase Inhibitors/pharmacology , Cisplatin/pharmacology , Fendiline/pharmacology , Neuroblastoma/drug therapy , Animals , Apoptosis , Cell Proliferation , Drug Therapy, Combination , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neuroblastoma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
J Alzheimers Dis ; 68(3): 931-938, 2019.
Article in English | MEDLINE | ID: mdl-30814362

ABSTRACT

Certain proteases are involved in Alzheimer's disease (AD) and their erroneous control may contribute to the pathology onset and progression. In this study we evaluated the cerebral expression of eight proteases, involved in both AßPP processing and extracellular matrix remodeling. Among these proteases, ADAM10, ADAMTS1, Cathepsin D, and Meprin ß show a significantly higher mRNAs expression in sporadic AD subjects versus controls, while ADAMTS1, Cathepsin D, and Meprin ß show an increment also at the protein level. These data indicate that transcriptional events affecting brain proteases are activated in AD patients, suggesting a link between proteolysis and AD.


Subject(s)
Alzheimer Disease/enzymology , Brain/enzymology , Peptide Hydrolases/metabolism , ADAM10 Protein/metabolism , ADAM17 Protein/metabolism , Aged , Aged, 80 and over , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Blotting, Western , Case-Control Studies , Cathepsin D/metabolism , Cathepsin L/metabolism , Female , Humans , Male , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Middle Aged , Real-Time Polymerase Chain Reaction , Up-Regulation
7.
Int J Mol Sci ; 20(4)2019 Feb 19.
Article in English | MEDLINE | ID: mdl-30791416

ABSTRACT

The aim of this review is to critically analyze promises and limitations of pharmacological inducers of autophagy against protein misfolding-associated neurodegeneration. Effective therapies against neurodegenerative disorders can be developed by regulating the "self-defense" equipment of neurons, such as autophagy. Through the degradation and recycling of the intracellular content, autophagy promotes neuron survival in conditions of trophic factor deprivation, oxidative stress, mitochondrial and lysosomal damage, or accumulation of misfolded proteins. Autophagy involves the activation of self-digestive pathways, which is different for dynamics (macro, micro and chaperone-mediated autophagy), or degraded material (mitophagy, lysophagy, aggrephagy). All neurodegenerative disorders share common pathogenic mechanisms, including the impairment of autophagic flux, which causes the inability to remove the neurotoxic oligomers of misfolded proteins. Pharmacological activation of autophagy is typically achieved by blocking the kinase activity of mammalian target of rapamycin (mTOR) enzymatic complex 1 (mTORC1), removing its autophagy suppressor activity observed under physiological conditions; acting in this way, rapamycin provided the first proof of principle that pharmacological autophagy enhancement can induce neuroprotection through the facilitation of oligomers' clearance. The demand for effective disease-modifying strategies against neurodegenerative disorders is currently stimulating the development of a wide number of novel molecules, as well as the re-evaluation of old drugs for their pro-autophagic potential.


Subject(s)
Autophagy/drug effects , Drug Discovery , Neuroprotection/drug effects , Animals , Autophagy/genetics , Biomarkers , Drug Discovery/methods , Humans , Lysosomes/drug effects , Lysosomes/genetics , Lysosomes/metabolism , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Protein Aggregates , Protein Aggregation, Pathological , Protein Binding , Protein Conformation , Protein Folding , Protein Multimerization , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/etiology , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology , Structure-Activity Relationship
8.
Mol Neurobiol ; 56(3): 1957-1971, 2019 Mar.
Article in English | MEDLINE | ID: mdl-29981054

ABSTRACT

Soluble Aß oligomers are widely recognized as the toxic forms responsible for triggering AD, and Aß receptors are hypothesized to represent the first step in a neuronal cascade leading to dementia. Cellular prion protein (PrP) has been reported as a high-affinity binder of Aß oligomers. The interactions of PrP with both Aß42 and the highly toxic N-truncated pyroglutamylated species (AßpE3-42) are here investigated, at a molecular level, by means of ThT fluorescence, NMR and TEM. We demonstrate that soluble PrP binds both Aß42 and AßpE3-42, preferentially interacting with oligomeric species and delaying fibril formation. Residue level analysis of Aß42 oligomerization process reveals, for the first time, that PrP is able to differently interact with the forming oligomers, depending on the aggregation state of the starting Aß42 sample. A distinct behavior is observed for Aß42 1-30 region and C-terminal residues, suggesting that PrP protects Aß42 N-tail from entangling on the mature NMR-invisible fibril, consistent with the hypothesis that Aß42 N-tail is the locus of interaction with PrP. PrP/AßpE3-42 interactions are here reported for the first time. All interaction data are validated and complemented by cellular tests performed on Wt and PrP-silenced neuronal cell lines, clearly showing PrP dependent Aß oligomer cell internalization and toxicity. The ability of soluble PrP to compete with membrane-anchored PrP for binding to Aß oligomers bears relevance for studies of druggable pathways.


Subject(s)
Amyloid beta-Peptides/metabolism , Neurons/metabolism , Peptide Fragments/metabolism , Prion Proteins/metabolism , Animals , Binding Sites , Cell Line , Cell Survival/physiology , Magnetic Resonance Spectroscopy , Mice , Protein Binding
9.
Cell Death Dis ; 9(2): 166, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29416016

ABSTRACT

According to the "gain-of-toxicity mechanism", neuronal loss during cerebral proteinopathies is caused by accumulation of aggregation-prone conformers of misfolded cellular proteins, although it is still debated which aggregation state actually corresponds to the neurotoxic entity. Autophagy, originally described as a variant of programmed cell death, is now emerging as a crucial mechanism for cell survival in response to a variety of cell stressors, including nutrient deprivation, damage of cytoplasmic organelles, or accumulation of misfolded proteins. Impairment of autophagic flux in neurons often associates with neurodegeneration during cerebral amyloidosis, suggesting a role in clearing neurons from aggregation-prone misfolded proteins. Thus, autophagy may represent a target for innovative therapies. In this work, we show that alterations of autophagy progression occur in neurons following in vitro exposure to the amyloidogenic and neurotoxic prion protein-derived peptide PrP90-231. We report that the increase of autophagic flux represents a strategy adopted by neurons to survive the intracellular accumulation of misfolded PrP90-231. In particular, PrP90-231 internalization in A1 murine mesencephalic neurons occurs in acidic structures, showing electron microscopy hallmarks of autophagosomes and autophagolysosomes. However, these structures do not undergo resolution and accumulate in cytosol, suggesting that, in the presence of PrP90-231, autophagy is activated but its progression is impaired; the inability to clear PrP90-231 via autophagy induces cytotoxicity, causing impairment of lysosomal integrity and cytosolic diffusion of hydrolytic enzymes. Conversely, the induction of autophagy by pharmacological  blockade of mTOR kinase or trophic factor deprivation restored autophagy resolution, reducing intracellular PrP90-231 accumulation and neuronal death. Taken together, these data indicate that PrP90-231 internalization induces an autophagic defensive response in A1 neurons, although incomplete and insufficient to grant survival; the pharmacological enhancement of this process exerts neuroprotection favoring the clearing of the internalized peptide and could represents a promising neuroprotective tool for neurodegenerative proteinopathies.


Subject(s)
Autophagy , Intracellular Space/metabolism , Neurons/metabolism , Peptides/metabolism , Prion Proteins/metabolism , Protein Aggregates , Protein Folding , Acids/metabolism , Animals , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Autophagy/drug effects , Cytoplasmic Vesicles/drug effects , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/ultrastructure , Endocytosis/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/ultrastructure , Membrane Potential, Mitochondrial , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Neurons/drug effects , Neurons/ultrastructure , Neuroprotection/drug effects , Permeability , Prion Proteins/toxicity , Protein Aggregates/drug effects , Protein Folding/drug effects , Sirolimus/pharmacology
10.
J Alzheimers Dis ; 61(1): 1-15, 2018.
Article in English | MEDLINE | ID: mdl-29103038

ABSTRACT

The processing of the amyloid-ß protein precursor (AßPP) by ß- and γ-secretases is a pivotal event in the genesis of Alzheimer's disease (AD). Besides familial mutations on the AßPP gene, or upon its overexpression, familial forms of AD are often caused by mutations or deletions in presenilin 1 (PSEN1) and 2 (PSEN2) genes: the catalytic components of the proteolytic enzyme γ-secretase (GS). The "amyloid hypothesis", modified over time, states that the aberrant processing of AßPP by GS induces the formation of specific neurotoxic soluble amyloid-ß (Aß) peptides which, in turn, cause neurodegeneration. This theory, however, has recently evidenced significant limitations and, in particular, the following issues are debated: 1) the concept and significance of presenilin's "gain of function" versus "loss of function"; and 2) the presence of several and various GS substrates, which interact with AßPP and may influence Aß formation. The latter consideration is suggestive: despite the increasing number of GS substrates so far identified, their reciprocal interaction with AßPP itself, even in the AD field, is significantly unexplored. On the other hand, GS is also an important pharmacological target in the cancer field; inhibitors or GS activity are investigated in clinical trials for treating different tumors. Furthermore, the function of AßPP and PSENs in brain development and in neuronal migration is well known. In this review, we focused on a specific subset of GS substrates that directly interact with AßPP and are involved in its proteolysis and signaling, by evaluating their role in neurodegeneration and in cell motility or proliferation, as a possible connection between AD and cancer.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Neoplasms/metabolism , Presenilin-1/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Humans , Presenilin-1/genetics
11.
Neurotox Res ; 32(3): 381-397, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28540665

ABSTRACT

Glia over-stimulation associates with amyloid deposition contributing to the progression of central nervous system neurodegenerative disorders. Here we analyze the molecular mechanisms mediating microglia-dependent neurotoxicity induced by prion protein (PrP)90-231, an amyloidogenic polypeptide corresponding to the protease-resistant portion of the pathological prion protein scrapie (PrPSc). PrP90-231 neurotoxicity is enhanced by the presence of microglia within neuronal culture, and associated to a rapid neuronal [Ca++] i increase. Indeed, while in "pure" cerebellar granule neuron cultures, PrP90-231 causes a delayed intracellular Ca++ entry mediated by the activation of NMDA receptors; when neuron and glia are co-cultured, a transient increase of [Ca++] i occurs within seconds after treatment in both granule neurons and glial cells, then followed by a delayed and sustained [Ca++] i raise, associated with the induction of the expression of inducible nitric oxide synthase and phagocytic NADPH oxidase. [Ca++] i fast increase in neurons is dependent on the activation of multiple pathways since it is not only inhibited by the blockade of voltage-gated channel activity and NMDA receptors but also prevented by the inhibition of nitric oxide and PGE2 release from glial cells. Thus, Ca++ homeostasis alteration, directly induced by PrP90-231 in cerebellar granule cells, requires the activation of NMDA receptors, but is greatly enhanced by soluble molecules released by activated glia. In glia-enriched cerebellar granule cultures, the activation of inducible nitric oxide (iNOS) and NADPH oxidase represents the main mechanism of toxicity since their pharmacological inhibition prevented PrP90-231 neurotoxicity, whereas NMDA blockade by D(-)-2-amino-5-phosphonopentanoic acid is ineffective; conversely, in pure cerebellar granule cultures, NMDA blockade but not iNOS inhibition strongly reduced PrP90-231 neurotoxicity. These data indicate that amyloidogenic peptides induce neurotoxic signals via both direct neuron interaction and glia activation through different mechanisms responsible of calcium homeostasis disruption in neurons and potentiating each other: the activation of excitotoxic pathways via NMDA receptors and the release of radical species that establish an oxidative milieu.


Subject(s)
Cerebellum/drug effects , Neuroglia/drug effects , Neurons/drug effects , Peptide Fragments/toxicity , Prions/toxicity , Animals , Calcium/metabolism , Calcium Channels/metabolism , Cell Death , Cells, Cultured , Cerebellum/metabolism , Cerebellum/pathology , Coculture Techniques , Intracellular Space/metabolism , NADPH Oxidases/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Peptide Fragments/metabolism , Prions/metabolism , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism
12.
Oncotarget ; 8(5): 8189-8205, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28029658

ABSTRACT

We recently reported the in vitro over-expression of 45A, a RNA polymerase III-transcribed non-coding (nc)RNA, that perturbs the intracellular content of FE65L1 affecting cell proliferation rate, short-term response to genotoxic stress, substrate adhesion capacity and, ultimately, increasing the tumorigenic potential of human neuroblastoma cells. In this work, to deeply explore the mechanism by which 45A ncRNA contributes to cancer development, we targeted in vitro and in vivo 45A levels by the stable overexpression of antisense 45A RNA.45A downregulation leads to deep modifications of cytoskeleton organization, adhesion and migration of neuroblastoma cells. These effects are correlated with alterations in the expression of several genes including GTSE1 (G2 and S phase-expressed-1), a crucial regulator of tumor cell migration and metastatic potential. Interestingly, the downregulation of 45A ncRNA strongly affects the in vivo tumorigenic potential of SKNBE2 neuroblastoma cells, increasing tumor nodule compactness and reducing GTSE1 protein expression in a subcutaneous neuroblastoma mouse model. Moreover, intracardiac injection of neuroblastoma cells showed that downregulation of 45A ncRNA also influences tumor metastatic ability. In conclusion, our data highlight a key role of 45A ncRNA in cancer development and suggest that its modulation might represent a possible novel anticancer therapeutic approach.


Subject(s)
Cell Movement , Cell Proliferation , Neuroblastoma/metabolism , RNA, Untranslated/genetics , Tumor Burden , Animals , Cell Adhesion , Cell Line, Tumor , DNA Damage , DNA Repair , Down-Regulation , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Ki-67 Antigen/metabolism , Mice, Inbred NOD , Mice, SCID , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neoplasm Invasiveness , Neuroblastoma/genetics , Neuroblastoma/secondary , RNA, Antisense/genetics , RNA, Antisense/metabolism , RNA, Untranslated/metabolism , Signal Transduction , Time Factors , Transfection
13.
Jpn J Vet Res ; 64(2): 101-12, 2016 May.
Article in English | MEDLINE | ID: mdl-27506084

ABSTRACT

Cancer stem cells (CSCs) represent a small subpopulation of cells responsible for tumor formation and progression, drug resistance, tumor recurrence and metastasization. CSCs have been identified in many human tumors including osteosarcoma (OSA). CSC distinctive properties are the expression of stem cell markers, sustained growth, self-renewal and tumorigenicity. Here we report the isolation of stem-like cells from two canine OSA cultures, characterized by self-renewal, evaluated by sphere formation ability, differential marker expression, and in vitro proliferation when cultured in a medium containing EGF and bFGF. Current therapies for OSA increased survival time, but prognosis remains poor, due to the development of drug resistance and metastases. Chemotherapy shrinks the tumor mass but CSCs remain unaffected, leading to tumor recurrence. Metformin, a drug for type 2 diabetes, has been shown to possess antitumor properties affecting CSC survival in different human and animal cancers. Here we show that metformin has a significant antiproliferative effect on canine OSA stem-like cells, validating this in vitro model for further pre-clinical drug evaluations. In conclusion, our results demonstrate the feasibility of obtaining CSC-enriched cultures from primary canine OSA cells as a promising model for biological and pharmacological studies of canine and human OSAs.


Subject(s)
Dog Diseases/metabolism , Neoplastic Stem Cells/physiology , Osteosarcoma/veterinary , Animals , Biomarkers , Cell Line, Tumor , Cell Proliferation , Cell Survival , Dogs , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/cytology
14.
Oncotarget ; 7(25): 38638-38657, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27229535

ABSTRACT

Prion protein (PrPC) is a cell surface glycoprotein whose misfolding is responsible for prion diseases. Although its physiological role is not completely defined, several lines of evidence propose that PrPC is involved in self-renewal, pluripotency gene expression, proliferation and differentiation of neural stem cells. Moreover, PrPC regulates different biological functions in human tumors, including glioblastoma (GBM). We analyzed the role of PrPC in GBM cell pathogenicity focusing on tumor-initiating cells (TICs, or cancer stem cells, CSCs), the subpopulation responsible for development, progression and recurrence of most malignancies. Analyzing four GBM CSC-enriched cultures, we show that PrPC expression is directly correlated with the proliferation rate of the cells. To better define its role in CSC biology, we knocked-down PrPC expression in two of these GBM-derived CSC cultures by specific lentiviral-delivered shRNAs. We provide evidence that CSC proliferation rate, spherogenesis and in vivo tumorigenicity are significantly inhibited in PrPC down-regulated cells. Moreover, PrPC down-regulation caused loss of expression of the stemness and self-renewal markers (NANOG, Sox2) and the activation of differentiation pathways (i.e. increased GFAP expression). Our results suggest that PrPC controls the stemness properties of human GBM CSCs and that its down-regulation induces the acquisition of a more differentiated and less oncogenic phenotype.


Subject(s)
Glioblastoma/genetics , Neoplastic Stem Cells/metabolism , Prion Proteins/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Glioblastoma/pathology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/pathology , Transfection
15.
Curr Drug Targets ; 17(7): 824-49, 2016.
Article in English | MEDLINE | ID: mdl-26240051

ABSTRACT

Malignant pleural mesothelioma (MPM) is one of the deadliest and most heterogeneous tumors, highly refractory to multimodal therapeutic approach, including surgery, chemo- and radiotherapy. Preclinical and clinical studies exploring the efficacy of drugs targeting tyrosine kinases, angiogenesis and histone deacetylases, did not fulfil the expected clinical benefits. Thus, novel molecular targets should be identified from a definite knowledge of the unique biology and most relevant transduction pathways of MPM cells. Cancer stem cells (CSCs) are a subset of malignant precursors responsible for initiation, progression, resistance to cytotoxic drugs, recurrence and metastatic diffusion of tumor cells. CSCs are putative driving factors for MPM development and contribute to its clinical and biological heterogeneity; hence, targeted eradication of CSCs represents an ineludible goal to counteract MPM aggressiveness. In this context, innovative preclinical models could be exploited to identify novel intracellular pathway inhibitors able to target CSC viability. Novel drug targets have been identified among key factors responsible for the oncogenic transformation of mesothelial cells, often directly induced by asbestos. These include mitogenic and anti-apoptotic signaling that may also be activated by autocrine and paracrine cytokine pathways controlling cell plasticity. Both signaling pathways affecting proto-oncogene and transcription factor expression, or genetic and epigenetic alterations, such as mutations in cell cycle genes and silencing of tumor suppressor genes, represent promising disease-specific targets. In this review we describe current knowledge of MPM cell biology, focusing on potential targets to be tested in pharmacological studies, and highlighting results and challenges of clinical translation.


Subject(s)
Antineoplastic Agents/therapeutic use , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Neoplastic Stem Cells/drug effects , Pleural Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Clinical Trials as Topic , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/pathology , Mesothelioma/pathology , Mesothelioma, Malignant , Molecular Targeted Therapy , Neoplastic Stem Cells/pathology , Pleural Neoplasms/pathology , Proto-Oncogene Mas , Signal Transduction/drug effects
16.
Int J Endocrinol ; 2014: 753524, 2014.
Article in English | MEDLINE | ID: mdl-25484899

ABSTRACT

Chemokines are chemotactic regulators of immune surveillance in physiological and pathological conditions such as inflammation, infection, and cancer. Several chemokines and cognate receptors are constitutively expressed in the central nervous system, not only in glial and endothelial cells but also in neurons, controlling neurogenesis, neurite outgrowth, and axonal guidance during development. In particular, the chemokine CXCL12 and its receptors, CXCR4 and CXCR7, form a functional network that controls plasticity in different brain areas, influencing neurotransmission, neuromodulation, and cell migration, and the dysregulation of this chemokinergic axis is involved in several neurodegenerative, neuroinflammatory, and malignant diseases. CXCR4 primarily mediates the transduction of proliferative signals, while CXCR7 seems to be mainly responsible for scavenging CXCL12. Importantly, the multiple intracellular signalling generated by CXCL12 interaction with its receptors influences hypothalamic modulation of neuroendocrine functions, although a direct modulation of pituitary functioning via autocrine/paracrine mechanisms was also reported. Both CXCL12 and CXCR4 are constitutively overexpressed in pituitary adenomas and their signalling induces cell survival and proliferation, as well as hormonal hypersecretion. In this review we focus on the physiological and pathological functions of immune-related cyto- and chemokines, mainly focusing on the CXCL12/CXCR4-7 axis, and their role in pituitary tumorigenesis. Accordingly, we discuss the potential targeting of CXCR4 as novel pharmacological approach for pituitary adenomas.

17.
Biochim Biophys Acta ; 1833(6): 1511-26, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23485396

ABSTRACT

FE65 proteins constitute a family of adaptors which modulates the processing of amyloid precursor protein and the consequent amyloid ß production. Thus, they have been involved in the complex and partially unknown cascade of reactions at the base of Alzheimer's disease etiology. However, FE65 and FE65-like proteins may be linked to neurodegeneration through the regulation of cell cycle in post-mitotic neurons. In this work we disclose novel molecular mechanisms by which APBB2 can modulate APP processing. We show that APBB2 mRNA splicing, driven by the over-expression of a novel non-coding RNA named 45A, allow the generation of alternative protein forms endowed with differential effects on Aß production, cell cycle control, and DNA damage response. 45A overexpression also favors cell transformation and tumorigenesis leading to a marked increase of malignancy of neuroblastoma cells. Therefore, our results highlight a novel regulatory pathway of considerable interest linking APP processing with cell cycle regulation and DNA-surveillance systems, that may represent a molecular mechanism to induce neurodegeneration in post-mitotic neurons.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Alternative Splicing , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/genetics , Cell Cycle , Neuroblastoma/pathology , RNA, Small Nuclear/genetics , Adaptor Proteins, Signal Transducing/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloidosis/metabolism , Animals , Apoptosis , Blotting, Western , Cell Adhesion , Cell Movement , Cell Proliferation , Cells, Cultured , Fluorescent Antibody Technique , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Micronucleus Tests , Neuroblastoma/genetics , Neuroblastoma/metabolism , Protein Binding , Protein Processing, Post-Translational , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
18.
Int J Pept ; 2013: 926295, 2013.
Article in English | MEDLINE | ID: mdl-23476673

ABSTRACT

Peptide receptors involved in pathophysiological processes represent promising therapeutic targets. Neuropeptide somatostatin (SST) is produced by specialized cells in a large number of human organs and tissues. SST primarily acts as inhibitor of endocrine and exocrine secretion via the activation of five G-protein-coupled receptors, named sst1-5, while in central nervous system, SST acts as a neurotransmitter/neuromodulator, regulating locomotory and cognitive functions. Critical points of SST/SST receptor biology, such as signaling pathways of individual receptor subtypes, homo- and heterodimerization, trafficking, and cross-talk with growth factor receptors, have been extensively studied, although functions associated with several pathological conditions, including cancer, are still not completely unraveled. Importantly, SST exerts antiproliferative and antiangiogenic effects on cancer cells in vitro, and on experimental tumors in vivo. Moreover, SST agonists are clinically effective as antitumor agents for pituitary adenomas and gastro-pancreatic neuroendocrine tumors. However, SST receptors being expressed by tumor cells of various tumor histotypes, their pharmacological use is potentially extendible to other cancer types, although to date no significant results have been obtained. In this paper the most recent findings on the expression and functional roles of SST and SST receptors in tumor cells are discussed.

19.
Dis Model Mech ; 6(2): 424-33, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22996644

ABSTRACT

Recent studies indicated that sortilin-related receptor 1 (SORL1) is a risk gene for late-onset Alzheimer's disease (AD), although its role in the aetiology and/or progression of this disorder is not fully understood. Here, we report the finding of a non-coding (nc) RNA (hereafter referred to as 51A) that maps in antisense configuration to intron 1 of the SORL1 gene. 51A expression drives a splicing shift of SORL1 from the synthesis of the canonical long protein variant A to an alternatively spliced protein form. This process, resulting in a decreased synthesis of SORL1 variant A, is associated with impaired processing of amyloid precursor protein (APP), leading to increased Aß formation. Interestingly, we found that 51A is expressed in human brains, being frequently upregulated in cerebral cortices from individuals with Alzheimer's disease. Altogether, these findings document a novel ncRNA-dependent regulatory pathway that might have relevant implications in neurodegeneration.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Brain/pathology , Introns/genetics , LDL-Receptor Related Proteins/genetics , Membrane Transport Proteins/genetics , RNA, Untranslated/metabolism , Up-Regulation/genetics , Aged , Alternative Splicing/genetics , Alzheimer Disease/pathology , Base Sequence , Brain/metabolism , Cell Extracts , Cell Line , Cell Nucleus/metabolism , Female , Humans , LDL-Receptor Related Proteins/metabolism , Male , Membrane Transport Proteins/metabolism , Molecular Sequence Data , Postmortem Changes , RNA, Untranslated/genetics , Transcription, Genetic
20.
Int J Mol Sci ; 13(7): 8648-8669, 2012.
Article in English | MEDLINE | ID: mdl-22942726

ABSTRACT

In several neurodegenerative diseases, such as Parkinson, Alzheimer's, Huntington, and prion diseases, the deposition of aggregated misfolded proteins is believed to be responsible for the neurotoxicity that characterizes these diseases. Prion protein (PrP), the protein responsible of prion diseases, has been deeply studied for the peculiar feature of its misfolded oligomers that are able to propagate within affected brains, inducing the conversion of the natively folded PrP into the pathological conformation. In this review, we summarize the available experimental evidence concerning the relationship between aggregation status of misfolded PrP and neuronal death in the course of prion diseases. In particular, we describe the main findings resulting from the use of different synthetic (mainly PrP106-126) and recombinant PrP-derived peptides, as far as mechanisms of aggregation and amyloid formation, and how these different spatial conformations can affect neuronal death. In particular, most data support the involvement of non-fibrillar oligomers rather than actual amyloid fibers as the determinant of neuronal death.


Subject(s)
Prion Diseases/metabolism , Prions/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Humans , Mutation, Missense , Prion Diseases/genetics , Prion Diseases/transmission , Prions/genetics , Protein Aggregation, Pathological/genetics , Protein Stability , Protein Transport , Soil/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...