Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Cancers (Basel) ; 16(5)2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38473215

ABSTRACT

Identifying the molecular mechanisms underlying radioresistance is a priority for the treatment of RMS, a myogenic tumor accounting for approximately 50% of all pediatric soft tissue sarcomas. We found that irradiation (IR) transiently increased phosphorylation of Akt1, Src, and Cav1 in human RD and RH30 lines. Synthetic inhibition of Akt1 and Src phosphorylation increased ROS levels in all RMS lines, promoting cellular radiosensitization. Accordingly, the elevated activation of the Akt1/Src/Cav1 pathway, as detected in two RD lines characterized by overexpression of a myristoylated Akt1 form (myrAkt1) or Cav1 (RDCav1), was correlated with reduced levels of ROS, higher expression of catalase, and increased radioresistance. We found that treatment with cholesterol-lowering drugs such as lovastatin and simvastatin promoted cell apoptosis in all RMS lines by reducing Akt1 and Cav1 levels and increasing intracellular ROS levels. Combining statins with IR significantly increased DNA damage and cell apoptosis as assessed by γ histone 2AX (γH2AX) staining and FACS analysis. Furthermore, in combination with the chemotherapeutic agent actinomycin D, statins were effective in reducing cell survival through increased apoptosis. Taken together, our findings suggest that the molecularly linked signature formed by Akt1, Src, Cav1, and catalase may represent a prognostic determinant for identifying subgroups of RMS patients with higher probability of recurrence after radiotherapy. Furthermore, statin-induced oxidative stress could represent a treatment option to improve the success of radiotherapy.

2.
Biochim Biophys Acta Rev Cancer ; 1878(6): 189019, 2023 11.
Article in English | MEDLINE | ID: mdl-37951482

ABSTRACT

Endometrial carcinoma (EC) defines a heterogeneous group of neoplastic diseases originating from the transformation of endometrial cells that constitute the internal lining of the uterus. To date several molecular targets have been analysed to describe the natural course of the disease, claudins being among these. Claudins are the main components of tight junctions (TJs), and their main functions are ascribed to the compartmentalization of tissues and cell-cell communication by means of intracellular ions diffusion: these features are typical of epithelial cells. Their overexpression, mis-localization or loss contribute to the malignancy of EC cells. This review collected all available data regarding the expression, regulation and claudin-related signaling pathways to provide a comprehensive view on the influence of claudin in EC progression. Further, the translational potential of claudin differential expression was explored, indicating that their role in personalized medicine could also contribute to EC therapy besides their employment for diagnosis and prognosis.


Subject(s)
Claudins , Endometrial Neoplasms , Female , Humans , Claudins/genetics , Claudins/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Epithelial Cells/metabolism , Tight Junctions/metabolism , Tight Junctions/pathology , Signal Transduction
3.
Biochim Biophys Acta Gen Subj ; 1867(12): 130470, 2023 12.
Article in English | MEDLINE | ID: mdl-37778450

ABSTRACT

The activation loop (A-loop) of kinases, a key regulatory region, is recurrently mutated in several kinase proteins in cancer resulting in dysregulated kinase activity and response to kinase inhibitors. FGFR1 receptor tyrosine kinase represents an important oncogene and therapeutic target for solid and hematological tumors. Here we investigate the biochemical and molecular effects of D647N mutation lying in the A-loop of FGFR1. When expressed in normal and tumoral in vitro cell models, FGFR1D647N is phosphorylated also in the absence of ligands, and this is accompanied by the activation of intracellular signaling. The expression of FGFR1D647N significantly increases single and collective migration of cancer cells in vitro and in vivo, when compared to FGFR1WT. FGFR1D647N expression exacerbates the aggressiveness of cancer cells, increasing their invasiveness in vitro and augmenting their pro-angiogenic capacity in vivo. Remarkably, the D647N mutation significantly increases the sensitivity of FGFR1 to the ATP-competitive inhibitor Erdafitinib suggesting the possibility that this mutation could become a specific target for the development of new inhibitors. Although further efforts are warranted for an exhaustive description of the activation mechanisms, for the identification of more specific inhibitors and for confirming the clinical significance of mutated FGFR1D647N, overall our data demonstrate that the D647N substitution of FGFR1 is a novel pro-oncogenic activating mutation of the receptor that, when found in cancer patients, may anticipate good response to erdafitinib treatment.


Subject(s)
Receptor, Fibroblast Growth Factor, Type 1 , Signal Transduction , Humans , Ligands , Cell Line, Tumor , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Phosphorylation , Mutation
4.
Int J Mol Sci ; 24(8)2023 Apr 12.
Article in English | MEDLINE | ID: mdl-37108275

ABSTRACT

In vertebrates, two homologous heterotetrameric AP1 complexes regulate the intracellular protein sorting via vesicles. AP-1 complexes are ubiquitously expressed and are composed of four different subunits: γ, ß1, µ1 and σ1. Two different complexes are present in eukaryotic cells, AP1G1 (contains γ1 subunit) and AP1G2 (contains γ2 subunit); both are indispensable for development. One additional tissue-specific isoform exists for µ1A, the polarized epithelial cells specific to µ1B; two additional tissue-specific isoforms exist for σ1A: σ1B and σ1C. Both AP1 complexes fulfil specific functions at the trans-Golgi network and endosomes. The use of different animal models demonstrated their crucial role in the development of multicellular organisms and the specification of neuronal and epithelial cells. Ap1g1 (γ1) knockout mice cease development at the blastocyst stage, while Ap1m1 (µ1A) knockouts cease during mid-organogenesis. A growing number of human diseases have been associated with mutations in genes encoding for the subunits of adaptor protein complexes. Recently, a new class of neurocutaneous and neurometabolic disorders affecting intracellular vesicular traffic have been referred to as adaptinopathies. To better understand the functional role of AP1G1 in adaptinopathies, we generated a zebrafish ap1g1 knockout using CRISPR/Cas9 genome editing. Zebrafish ap1g1 knockout embryos cease their development at the blastula stage. Interestingly, heterozygous females and males have reduced fertility and showed morphological alterations in the brain, gonads and intestinal epithelium. An analysis of mRNA profiles of different marker proteins and altered tissue morphologies revealed dysregulated cadherin-mediated cell adhesion. These data demonstrate that the zebrafish model organism enables us to study the molecular details of adaptinopathies and thus also develop treatment strategies.


Subject(s)
Neurodevelopmental Disorders , Transcription Factor AP-1 , Zebrafish Proteins , Zebrafish , Animals , Female , Humans , Male , Mice , Endosomes/metabolism , Epithelial Cells/metabolism , Protein Isoforms/metabolism , trans-Golgi Network/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Neurodevelopmental Disorders/genetics , Transcription Factor AP-1/metabolism , Zebrafish Proteins/metabolism
5.
Methods Mol Biol ; 2572: 181-189, 2023.
Article in English | MEDLINE | ID: mdl-36161417

ABSTRACT

Embryonic stem cells give rise to teratomas when injected in vivo in experimental animal models. The characterization, the manipulation, and the breaking off of this specific characteristic are doubtlessly the last frontier for the applications of stem cells in translational medicine. Moreover, the urgency to adapt to new scientific demands drives the researcher to find alternative and faster models for testing the teratogenic properties of embryonic stem cells. Here, we compare the emerging model of the chick embryo chorioallantoic membrane (CAM) to the murine model, which represents the gold standard procedure for teratogenesis.


Subject(s)
Teratoma , Animals , Chick Embryo , Chorioallantoic Membrane , Embryonic Stem Cells , Mice
6.
Cytokine Growth Factor Rev ; 69: 51-60, 2023 02.
Article in English | MEDLINE | ID: mdl-36155165

ABSTRACT

Gremlin-1 is a secreted bone morphogenetic protein (BMP) antagonist playing a pivotal role in the regulation of tissue formation and embryonic development. Since its first identification in 1997, gremlin-1 has been shown to be a multifunctional factor involved in wound healing, inflammation, cancer and tissue fibrosis. Among others, the activity of gremlin-1 is mediated by its interaction with BMPs or with membrane receptors such as the vascular endothelial growth factor receptor 2 (VEGFR2) or heparan sulfate proteoglycans (HSPGs). Growing evidence has highlighted a central role of gremlin-1 in the homeostasis of the adipose tissue (AT). Of note, gremlin-1 is involved in AT dysfunction during type 2 diabetes, obesity and non-alcoholic fatty liver disease (NAFLD) metabolic disorders. In this review we discuss recent findings on gremlin-1 involvement in AT biology, with particular attention to its role in metabolic diseases, to highlight its potential as a prognostic marker and therapeutic target.


Subject(s)
Diabetes Mellitus, Type 2 , Vascular Endothelial Growth Factor A , Humans , Bone Morphogenetic Proteins
7.
Cytokine Growth Factor Rev ; 69: 61-72, 2023 02.
Article in English | MEDLINE | ID: mdl-35953434

ABSTRACT

Adipose tissue (AT) is a highly active and plastic endocrine organ. It secretes numerous soluble molecules known as adipokines, which act locally to AT control the remodel and homeostasis or exert pleiotropic functions in different peripheral organs. Aberrant production or loss of certain adipokines contributes to AT dysfunction associated with metabolic disorders, including obesity. The AT plasticity is strictly related to tissue vascularization. Angiogenesis supports the AT expansion, while regression of blood vessels is associated with AT hypoxia, which in turn mediates tissue inflammation, fibrosis and metabolic dysfunction. Several adipokines can regulate endothelial cell functions and are endowed with either pro- or anti-angiogenic properties. Here we address the role of adipokines in the regulation of angiogenesis. A better understanding of the link between adipokines and angiogenesis will open the way for novel therapeutic approaches to treat obesity and metabolic diseases.


Subject(s)
Adipokines , Adipose Tissue , Metabolic Diseases , Humans , Adipokines/metabolism , Adipose Tissue/blood supply , Adipose Tissue/metabolism , Inflammation/metabolism , Metabolic Diseases/metabolism , Obesity/metabolism , Neovascularization, Physiologic/physiology
8.
Int J Mol Sci ; 23(17)2022 Sep 01.
Article in English | MEDLINE | ID: mdl-36077366

ABSTRACT

Cancer is a set of diseases characterized by several hallmark properties, such as increased angiogenesis, proliferation, invasion, and metastasis. The increased angiogenic activity constantly supplies the tumors with nutrients and a plethora of cytokines to ensure cell survival. Along these cytokines is a newly discovered protein, called irisin, which is released into the circulation after physical exercise. Irisin is the product of fibronectin type III domain-containing protein 5 (FNDC5) proteolytic cleavage. Recently it has been the topic of investigation in several types of cancer. In this study, we conducted a systematic review and meta-analysis to investigate its implication in different types of cancer. Our results suggest that irisin expression is decreased in cancer patients, thus it can be used as a valid biomarker for the diagnosis of several types of cancer. In addition, our results indicate that irisin may have an important role in tumor progression and metastasis since it is involved in multiple signaling pathways that promote cell proliferation and migration.


Subject(s)
Fibronectins , Neoplasms , Cytokines , Exercise , Fibronectins/metabolism , Humans , Transcription Factors
9.
Cells ; 11(18)2022 09 14.
Article in English | MEDLINE | ID: mdl-36139434

ABSTRACT

In pediatric rhabdomyosarcoma (RMS), elevated Akt signaling is associated with increased malignancy. Here, we report that expression of a constitutively active, myristoylated form of Akt1 (myrAkt1) in human RMS RD cells led to hyperactivation of the mammalian target of rapamycin (mTOR)/70-kDa ribosomal protein S6 kinase (p70S6K) pathway, resulting in the loss of both MyoD and myogenic capacity, and an increase of Ki67 expression due to high cell mitosis. MyrAkt1 signaling increased migratory and invasive cell traits, as detected by wound healing, zymography, and xenograft zebrafish assays, and promoted repair of DNA damage after radiotherapy and doxorubicin treatments, as revealed by nuclear detection of phosphorylated H2A histone family member X (γH2AX) through activation of DNA-dependent protein kinase (DNA-PK). Treatment with synthetic inhibitors of phosphatidylinositol-3-kinase (PI3K) and Akt was sufficient to completely revert the aggressive cell phenotype, while the mTOR inhibitor rapamycin failed to block cell dissemination. Furthermore, we found that pronounced Akt1 signaling increased the susceptibility to cell apoptosis after treatments with 2-deoxy-D-glucose (2-DG) and lovastatin, enzymatic inhibitors of hexokinase, and 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR), especially in combination with radiotherapy and doxorubicin. In conclusion, these data suggest that restriction of glucose metabolism and the mevalonate pathway, in combination with standard therapy, may increase therapy success in RMS tumors characterized by a dysregulated Akt signaling.


Subject(s)
Proto-Oncogene Proteins c-akt , Rhabdomyosarcoma, Embryonal , Animals , Child , DNA Repair , DNA-Activated Protein Kinase/genetics , Deoxyglucose , Doxorubicin/pharmacology , Glucose , Glycolysis , Hexokinase/metabolism , Histones/metabolism , Humans , Ki-67 Antigen/metabolism , Lovastatin , MTOR Inhibitors , Mevalonic Acid , Oxidoreductases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositols , Proto-Oncogene Proteins c-akt/metabolism , Rhabdomyosarcoma, Embryonal/drug therapy , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Zebrafish/genetics
11.
Int J Mol Sci ; 23(3)2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35163075

ABSTRACT

Gremlin-1 is a secreted cystine-knot protein that acts as an antagonist of bone morphogenetic proteins (BMPs), and as a ligand of heparin and the vascular endothelial growth factor receptor 2 (VEGFR2), thus regulating several physiological and pathological processes, including embryonic development, tissue fibrosis and cancer. Gremlin-1 exerts all these biological activities only in its homodimeric form. Here, we propose a multi-step approach for the expression and purification of homodimeric, fully active, histidine-tagged recombinant gremlin-1, using mammalian HEK293T cells. Ion metal affinity chromatography (IMAC) of crude supernatant followed by heparin-affinity chromatography enables obtaining a highly pure recombinant dimeric gremlin-1 protein, exhibiting both BMP antagonist and potent VEGFR2 agonist activities.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Chromatography, Affinity/methods , Intercellular Signaling Peptides and Proteins/metabolism , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor Receptor-2/agonists , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/isolation & purification , Recombinant Proteins/genetics
12.
Biochim Biophys Acta Gen Subj ; 1866(4): 130085, 2022 04.
Article in English | MEDLINE | ID: mdl-35016977

ABSTRACT

BACKGROUND: Adipose tissue plays a pivotal role in the development and progression of the metabolic syndrome which along with its complications is an epidemic of the 21st century. Irisin is an adipo-myokine secreted mainly by skeletal muscle and targeting, among others, adipose tissue. In brown adipose tissue it upregulates uncoupling protein-1 (UCP1) which is responsible for mitochondrial non-shivering thermogenesis. METHODS: Here we analyzed the effects of irisin on the metabolic activity of 3T3-L1 derived adipocytes through a mitochondrial flux assay. We also assessed the effects of irisin on the intracellular signaling through Western Blot. Finally, the gene expression of ucp1 and lipolytic genes was examined through RT-qPCR. RESULTS: Irisin affects mitochondrial respiration and lipolysis in a time-dependent manner through the regulation of PI3K-AKT pathway. Irisin also induces the expression of UCP1 and the regulation of NF-κB, and CREB and ERK pathways. CONCLUSION: Our data supports the role of irisin in the induction of non-shivering thermogenesis, the regulation of energy expenditure and lipolysis in adipocytes. GENERAL SIGNIFICANCE: Irisin may be an attractive therapeutic target in the treatment of obesity and related metabolic disorders.


Subject(s)
Fibronectins , Lipolysis , Thermogenesis , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Animals , Fibronectins/genetics , Mice , Phosphatidylinositol 3-Kinases/metabolism , Thermogenesis/genetics
13.
Biochim Biophys Acta Mol Basis Dis ; 1868(2): 166313, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34826586

ABSTRACT

Fibroblast growth factor receptors (FGFRs) are recurrently altered by single nucleotide variants (SNVs) in many human cancers. The prevalence of SNVs in FGFRs depends on the cancer type. In some tumors, such as the urothelial carcinoma, mutations of FGFRs occur at very high frequency (up to 60%). Many characterized mutations occur in the extracellular or transmembrane domains, while fewer known mutations are found in the kinase domain. In this study, we performed a bioinformatics analysis to identify novel putative cancer driver or therapeutically actionable mutations of the kinase domain of FGFRs. To pinpoint those mutations that may be clinically relevant, we exploited the recurrence of alterations on analogous amino acid residues within the kinase domain (PK_Tyr_Ser-Thr) of different kinases as a predictor of functional impact. By exploiting MutationAligner and LowMACA bioinformatics resources, we highlighted novel uncharacterized mutations of FGFRs which recur in other protein kinases. By revealing unanticipated correspondence with known variants, we were able to infer their functional effects, as alterations clustering on similar residues in analogous proteins have a high probability to elicit similar effects. As FGFRs represent an important class of oncogenes and drug targets, our study opens the way for further studies to validate their driver and/or actionable nature and, in the long term, for a more efficacious application of precision oncology.


Subject(s)
Carcinogenesis/pathology , Mutation , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Fibroblast Growth Factor/genetics , Amino Acid Sequence , Carcinogenesis/genetics , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Phosphorylation , Protein Domains
14.
Biochim Biophys Acta Rev Cancer ; 1876(2): 188614, 2021 12.
Article in English | MEDLINE | ID: mdl-34403770

ABSTRACT

The tremendous number of cancer variants that can be detected by NGS analyses has required the development of computational approaches to prioritize mutations on the basis of their biological and clinical significance. Standard strategies take a gene-centric approach to the problem, allowing exclusively the identification of highly frequent variants. On the contrary, protein domain (PD)-based approaches allow to identify functionally relevant low frequency variants by searching for mutations that recur on analogous residues across homologous proteins (i.e. containing the same PD). Such approaches enable to transfer information about the effects and druggability from one known mutation to unknown ones. Here we describe how PD-based strategies work, and discuss how they could be exploited for mutation prioritization. The principle that mutations clustered on specific residues of PDs have the same functional consequences and are therapeutically actionable in a similar manner could help the choice of patient-specific targeted drugs, eventually improving the management of cancer patients.


Subject(s)
Genetic Variation/genetics , Neoplasms/genetics , Protein Domains/genetics , Humans
16.
Blood ; 138(18): 1705-1720, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34077955

ABSTRACT

Alterations in KRAS have been identified as the most recurring somatic variants in the multiple myeloma (MM) mutational landscape. Combining DNA and RNA sequencing, we studied 756 patients and observed KRAS as the most frequently mutated gene in patients at diagnosis; in addition, we demonstrated the persistence or de novo occurrence of the KRAS aberration at disease relapse. Small-molecule inhibitors targeting KRAS have been developed; however, they are selective for tumors carrying the KRASG12C mutation. Therefore, there is still a need to develop novel therapeutic approaches to target the KRAS mutational events found in other tumor types, including MM. We used AZD4785, a potent and selective antisense oligonucleotide that selectively targets and downregulates all KRAS isoforms, as a tool to dissect the functional sequelae secondary to KRAS silencing in MM within the context of the bone marrow niche and demonstrated its ability to significantly silence KRAS, leading to inhibition of MM tumor growth, both in vitro and in vivo, and confirming KRAS as a driver and therapeutic target in MM.


Subject(s)
Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Mutation/drug effects , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Humans , Mice, SCID , Molecular Targeted Therapy , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Oligonucleotides, Antisense/therapeutic use , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
17.
Infect Immun ; 89(8): e0014121, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34031126

ABSTRACT

Some bacterial pathogens can manipulate the angiogenic response, suppressing or inducing it for their own ends. In humans, Bartonella henselae is associated with cat-scratch disease and vasculoproliferative disorders such as bacillary angiomatosis and bacillary peliosis. Although endothelial cells (ECs) support the pathogenesis of B. henselae, the mechanisms by which B. henselae induces EC activation are not completely clear, as well as the possible contributions of other cells recruited at the site of infection. Mesenchymal stromal cells (MSCs) are endowed with angiogenic potential and play a dual role in infections, exerting antimicrobial properties but also acting as a shelter for pathogens. Here, we delved into the role of MSCs as a reservoir of B. henselae and modulator of EC functions. B. henselae readily infected MSCs and survived in perinuclearly bound vacuoles for up to 8 days. Infection enhanced MSC proliferation and the expression of epidermal growth factor receptor (EGFR), Toll-like receptor 2 (TLR2), and nucleotide-binding oligomerization domain-containing protein 1 (NOD1), proteins that are involved in bacterial internalization and cytokine production. Secretome analysis revealed that infected MSCs secreted higher levels of the proangiogenic factors vascular endothelial growth factor (VEGF), fibroblast growth factor 7 (FGF-7), matrix metallopeptidase 9 (MMP-9), placental growth factor (PIGF), serpin E1, thrombospondin 1 (TSP-1), urokinase-type plasminogen activator (uPA), interleukin 6 (IL-6), platelet-derived growth factor D (PDGF-D), chemokine ligand 5 (CCL5), and C-X-C motif chemokine ligand 8 (CXCL8). Supernatants from B. henselae-infected MSCs increased the susceptibility of ECs to B. henselae infection and enhanced EC proliferation, invasion, and reorganization in tube-like structures. Altogether, these results indicate MSCs as a still underestimated niche for persistent B. henselae infection and reveal MSC-EC cross talk that may contribute to exacerbate bacterium-induced angiogenesis and granuloma formation.


Subject(s)
Angiomatosis, Bacillary/metabolism , Angiomatosis, Bacillary/microbiology , Bartonella henselae/physiology , Endothelial Cells/metabolism , Mesenchymal Stem Cells/metabolism , Neovascularization, Pathologic/metabolism , Angiomatosis, Bacillary/pathology , Biomarkers , Disease Susceptibility , Host-Pathogen Interactions , Humans
18.
Free Radic Biol Med ; 169: 294-303, 2021 06.
Article in English | MEDLINE | ID: mdl-33892112

ABSTRACT

Ferroptosis is a form of regulated cell death dependent on iron, reactive oxygen species and characterized by the accumulation of lipid peroxides. It can be experimentally initiated by chemicals, such as erastin and RSL3, that modulate GPX4 activity, the cellular antioxidant machinery that avert lipid peroxidation. The study aimed to investigate mitochondrial respiration and ferritin function as biomarkers of ferroptosis sensitivity of HepG2 and HA22T/VGH, two Hepatocellular Carcinoma (HCC) cell line models. Cell viability was determined by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyltetrazolium bromide (MTT) assay, labile iron levels were determined using Calcein-AM fluorescence microscopy, ferritin, glutathione and lipid peroxidation were assayed with commercially available kits. The Seahorse assay was used to investigate mitochondrial function in the cells. The study shows that highly differentiated HepG2 cells were more sensitive to RSL3-induced ferroptosis than the poorly differentiated HA22T/VGH (HCC) cell line (RSL3 IC50 0.07 µM in HepG2 vs 0.3 µM in HA22T/VGH). Interestingly, HepG2 exhibited higher mitochondrial respiration and lower glycolytic activity than HA22T/VGH and were more sensitive to RSL3-induced ferroptosis, indicating a mitochondrial-specific mechanism of action of RSL3. Interestingly, iron metabolism seems to be involved in this different sensitivity, specifically, the downregulation of H-ferritin (but not of L-subunit), makes HA22T/VGH more sensitive toward both RSL3-and iron-induced ferroptosis. Hence only the H-ferritin seems involved in the protection from this cell death process.


Subject(s)
Carcinoma, Hepatocellular , Ferroptosis , Liver Neoplasms , Apoferritins/genetics , Carbolines , Cell Line , Humans , Mitochondria , Respiration
19.
Cancer Lett ; 507: 80-88, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33744390

ABSTRACT

Vascular endothelial growth factor receptor 2 (VEGFR2) activating mutations are emerging as important oncogenic driver events. Understanding the biological implications of such mutations may help to pinpoint novel therapeutic targets. Here we show that activated VEGFR2 via the pro-oncogenic R1051Q mutation induces relevant metabolic changes in melanoma cells. The expression of VEGFR2R1051Q leads to higher energy metabolism and ATP production compared to control cells expressing VEGFR2WT. Furthermore, activated VEGFR2R1051Q augments the dependence on glutamine (Gln) of melanoma cells, thus increasing Gln uptake and their sensitivity to Gln deprivation and to inhibitors of glutaminase, the enzyme initiating Gln metabolism by cells. Overall, these results highlight Gln addiction as a metabolic vulnerability of tumors harboring the activating VEGFR2R1051Q mutation and suggest novel therapeutic approaches for those patients harboring activating mutations of VEGFR2.


Subject(s)
Energy Metabolism , Gain of Function Mutation , Glutamine/metabolism , Melanoma/metabolism , Skin Neoplasms/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Adenosine Triphosphate/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Energy Metabolism/drug effects , Enzyme Inhibitors/pharmacology , Glutaminase/antagonists & inhibitors , Glutaminase/metabolism , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Vascular Endothelial Growth Factor Receptor-2/genetics
20.
Front Immunol ; 12: 611761, 2021.
Article in English | MEDLINE | ID: mdl-33679750

ABSTRACT

Misfolded proteins, inflammation, and vascular alterations are common pathological hallmarks of neurodegenerative diseases. Alpha-synuclein is a small synaptic protein that was identified as a major component of Lewy bodies and Lewy neurites in the brain of patients affected by Parkinson's disease (PD), Lewy body dementia (LBD), and other synucleinopathies. It is mainly involved in the regulation of synaptic vesicle trafficking but can also control mitochondrial/endoplasmic reticulum (ER) homeostasis, lysosome/phagosome function, and cytoskeleton organization. Recent evidence supports that the pathological forms of α-synuclein can also reduce the release of vasoactive and inflammatory mediators from endothelial cells (ECs) and modulates the expression of tight junction (TJ) proteins important for maintaining the blood-brain barrier (BBB). This hints that α-synuclein deposition can affect BBB integrity. Border associated macrophages (BAMs) are brain resident macrophages found in association with the vasculature (PVMs), meninges (MAMs), and choroid plexus (CPMs). Recent findings indicate that these cells play distinct roles in stroke and neurodegenerative disorders. Although many studies have addressed how α-synuclein may modulate microglia, its effect on BAMs has been scarcely investigated. This review aims at summarizing the main findings supporting how α-synuclein can affect ECs and/or BAMs function as well as their interplay and effect on other cells in the brain perivascular environment in physiological and pathological conditions. Gaps of knowledge and new perspectives on how this protein can contribute to neurodegeneration by inducing BBB homeostatic changes in different neurological conditions are highlighted.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Endothelial Cells/metabolism , alpha-Synuclein/metabolism , Animals , Biomarkers , Brain/pathology , Cell Communication , Central Nervous System/metabolism , Central Nervous System/pathology , Disease Susceptibility , Gene Expression Regulation , Humans , Microglia/immunology , Microglia/metabolism , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , alpha-Synuclein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...