Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Hum Mol Genet ; 33(11): 969-980, 2024 May 18.
Article En | MEDLINE | ID: mdl-38483349

RNA methylation of N6-methyladenosine (m6A) is emerging as a fundamental regulator of every aspect of RNA biology. RNA methylation directly impacts protein production to achieve quick modulation of dynamic biological processes. However, whether RNA methylation regulates mitochondrial function is not known, especially in neuronal cells which require a high energy supply and quick reactive responses. Here we show that m6A RNA methylation regulates mitochondrial function through promoting nuclear-encoded mitochondrial complex subunit RNA translation. Conditional genetic knockout of m6A RNA methyltransferase Mettl14 (Methyltransferase like 14) by Nestin-Cre together with metabolomic analysis reveals that Mettl14 knockout-induced m6A depletion significantly downregulates metabolites related to energy metabolism. Furthermore, transcriptome-wide RNA methylation profiling of wild type and Mettl14 knockout mouse brains by m6A-Seq shows enrichment of methylation on mitochondria-related RNA. Importantly, loss of m6A leads to a significant reduction in mitochondrial respiratory capacity and membrane potential. These functional defects are paralleled by the reduced expression of mitochondrial electron transport chain complexes, as well as decreased mitochondrial super-complex assembly and activity. Mechanistically, m6A depletion decreases the translational efficiency of methylated RNA encoding mitochondrial complex subunits through reducing their association with polysomes, while not affecting RNA stability. Together, these findings reveal a novel role for RNA methylation in regulating mitochondrial function. Given that mitochondrial dysfunction and RNA methylation have been increasingly implicate in neurodegenerative disorders, our findings not only provide insights into fundamental mechanisms regulating mitochondrial function, but also open up new avenues for understanding the pathogenesis of neurological diseases.


Adenosine , Methyltransferases , Mice, Knockout , Mitochondria , Animals , Mitochondria/metabolism , Mitochondria/genetics , Mice , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/genetics , RNA/genetics , RNA/metabolism , Humans , Protein Biosynthesis , Energy Metabolism/genetics , Neurons/metabolism , RNA Methylation
2.
Cancer Res Commun ; 3(5): 943-951, 2023 05.
Article En | MEDLINE | ID: mdl-37377894

Interferons (IFNs) are cytokines with potent antineoplastic and antiviral properties. IFNα has significant clinical activity in the treatment of myeloproliferative neoplasms (MPN), but the precise mechanisms by which it acts are not well understood. Here, we demonstrate that chromatin assembly factor 1 subunit B (CHAF1B), an Unc-51-like kinase 1 (ULK1)-interactive protein in the nuclear compartment of malignant cells, is overexpressed in patients with MPN. Remarkably, targeted silencing of CHAF1B enhances transcription of IFNα-stimulated genes and promotes IFNα-dependent antineoplastic responses in primary MPN progenitor cells. Taken together, our findings indicate that CHAF1B is a promising newly identified therapeutic target in MPN and that CHAF1B inhibition in combination with IFNα therapy might offer a novel strategy for treating patients with MPN. Significance: Our findings raise the potential for clinical development of drugs targeting CHAF1B to enhance IFN antitumor responses in the treatment of patients with MPN and should have important clinical translational implications for the treatment of MPN and possibly in other malignancies.


Bone Marrow Neoplasms , Myeloproliferative Disorders , Neoplasms , Humans , Myeloproliferative Disorders/drug therapy , Interferon-alpha/pharmacology , Chromatin Assembly Factor-1/genetics
3.
Mol Cancer Res ; 21(4): 332-344, 2023 04 01.
Article En | MEDLINE | ID: mdl-36573964

Immune checkpoint inhibitors (ICI) have transformed the treatment of melanoma. However, the majority of patients have primary or acquired resistance to ICIs, limiting durable responses and patient survival. IFNγ signaling and the expression of IFNγ-stimulated genes correlate with either response or resistance to ICIs, in a context-dependent manner. While IFNγ-inducible immunostimulatory genes are required for response to ICIs, chronic IFNγ signaling induces the expression of immunosuppressive genes, promoting resistance to these therapies. Here, we show that high levels of Unc-51 like kinase 1 (ULK1) correlate with poor survival in patients with melanoma and overexpression of ULK1 in melanoma cells enhances IFNγ-induced expression of immunosuppressive genes, with minimal effects on the expression of immunostimulatory genes. In contrast, genetic or pharmacologic inhibition of ULK1 reduces expression of IFNγ-induced immunosuppressive genes. ULK1 binds IRF1 in the nuclear compartment of melanoma cells, controlling its binding to the programmed death-ligand 1 promoter region. In addition, pharmacologic inhibition of ULK1 in combination with anti-programmed cell death protein 1 therapy further reduces melanoma tumor growth in vivo. Our data suggest that targeting ULK1 represses IFNγ-dependent immunosuppression. These findings support the combination of ULK1 drug-targeted inhibition with ICIs for the treatment of patients with melanoma to improve response rates and patient outcomes. IMPLICATIONS: This study identifies ULK1, activated downstream of IFNγ signaling, as a druggable target to overcome resistance mechanisms to ICI therapy in metastatic melanoma.


Immune Checkpoint Inhibitors , Melanoma , Humans , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/drug therapy , Melanoma/genetics , Interferon-gamma/pharmacology , Immunosuppression Therapy , Immune Tolerance , Autophagy-Related Protein-1 Homolog/genetics , Intracellular Signaling Peptides and Proteins/genetics
4.
Cancer Res Commun ; 2(9): 966-978, 2022 09.
Article En | MEDLINE | ID: mdl-36382088

Glioblastoma (GBM) is an aggressive and incurable brain tumor in nearly all instances, whose disease progression is driven in part by the glioma stem cell (GSC) subpopulation. Here, we explored the effects of Schlafen family member 11 (SLFN11) in the molecular, cellular and tumor biology of GBM. CRISPR/Cas9 mediated knockout (KO) of SLFN11 inhibited GBM cell proliferation and neurosphere growth and was associated with reduced expression of progenitor/stem cell marker genes, such as NES, SOX2 and CD44. Loss of SLFN11 stimulated expression of NF-κB target genes, consistent with a negative regulatory role for SLFN11 on the NF-κB pathway. Further, our studies identify p21 as a direct transcriptional target of NF-κB2 in GBM whose expression was stimulated by loss of SLFN11. Genetic disruption of SLFN11 blocked GBM growth and significantly extended survival in an orthotopic patient-derived xenograft model. Together, our results identify SLFN11 as a novel component of signaling pathways that contribute to GBM and GSC with implications for future diagnostic and therapeutic strategies.


Glioblastoma , Glioma , Humans , Glioblastoma/genetics , NF-kappa B/genetics , Cell Line, Tumor , Signal Transduction/genetics , Nuclear Proteins/metabolism
5.
Nat Commun ; 13(1): 1750, 2022 04 01.
Article En | MEDLINE | ID: mdl-35365653

Interferons (IFNs) are key initiators and effectors of the immune response against malignant cells and also directly inhibit tumor growth. IFNα is highly effective in the treatment of myeloproliferative neoplasms (MPNs), but the mechanisms of action are unclear and it remains unknown why some patients respond to IFNα and others do not. Here, we identify and characterize a pathway involving PKCδ-dependent phosphorylation of ULK1 on serine residues 341 and 495, required for subsequent activation of p38 MAPK. We show that this pathway is essential for IFN-suppressive effects on primary malignant erythroid precursors from MPN patients, and that increased levels of ULK1 and p38 MAPK correlate with clinical response to IFNα therapy in these patients. We also demonstrate that IFNα treatment induces cleavage/activation of the ULK1-interacting ROCK1/2 proteins in vitro and in vivo, triggering a negative feedback loop that suppresses IFN responses. Overexpression of ROCK1/2 is seen in MPN patients and their genetic or pharmacological inhibition enhances IFN-anti-neoplastic responses in malignant erythroid precursors from MPN patients. These findings suggest the clinical potential of pharmacological inhibition of ROCK1/2 in combination with IFN-therapy for the treatment of MPNs.


Myeloproliferative Disorders , Neoplasms , Antiviral Agents/therapeutic use , Feedback , Humans , Interferon-alpha/pharmacology , Interferon-alpha/therapeutic use , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Neoplasms/drug therapy , Signal Transduction , rho-Associated Kinases/metabolism
6.
Oncogene ; 40(18): 3273-3286, 2021 05.
Article En | MEDLINE | ID: mdl-33846574

We provide evidence that a member of the human Schlafen (SLFN) family of proteins, SLFN5, is overexpressed in human pancreatic ductal adenocarcinoma (PDAC). Targeted deletion of SLFN5 results in decreased PDAC cell proliferation and suppresses PDAC tumorigenesis in in vivo PDAC models. Importantly, high expression levels of SLFN5 correlate with worse outcomes in PDAC patients, implicating SLFN5 in the pathophysiology of PDAC that leads to poor outcomes. Our studies establish novel regulatory effects of SLFN5 on cell cycle progression through binding/blocking of the transcriptional repressor E2F7, promoting transcription of key genes that stimulate S phase progression. Together, our studies suggest an essential role for SLFN5 in PDAC and support the potential for developing new therapeutic approaches for the treatment of pancreatic cancer through SLFN5 targeting.


Pancreatic Neoplasms , Carcinogenesis , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Pancreatic Neoplasms
7.
Blood Cancer J ; 11(3): 48, 2021 03 03.
Article En | MEDLINE | ID: mdl-33658483

Internal tandem duplication (-ITD) mutations of Fms-like tyrosine kinase 3 (FLT3) provide growth and pro-survival signals in the context of established driver mutations in FLT3 mutant acute myeloid leukemia (AML). Maternal embryonic leucine zipper kinase (MELK) is an aberrantly expressed gene identified as a target in AML. The MELK inhibitor OTS167 induces cell death in AML including cells with FLT3 mutations, yet the role of MELK and mechanisms of OTS167 function are not understood. OTS167 alone or in combination with tyrosine kinase inhibitors (TKIs) were used to investigate the effect of OTS167 on FLT3 signaling and expression in human FLT3 mutant AML cell lines and primary cells. We describe a mechanism whereby OTS167 blocks FLT3 expression by blocking FLT3 translation and inhibiting phosphorylation of eukaryotic initiation factor 4E-binding protein 1 (4E-BP1) and eukaryotic translation initiation factor 4B (eIF4B). OTS167 in combination with TKIs results in synergistic induction of FLT3 mutant cell death in FLT3 mutant cell lines and prolonged survival in a FLT3 mutant AML xenograft mouse model. Our findings suggest signaling through MELK is necessary for the translation and expression of FLT3-ITD, and blocking MELK with OTS167 represents a viable therapeutic strategy for patients with FLT3 mutant AML.


Leukemia, Myeloid, Acute/drug therapy , Naphthyridines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , fms-Like Tyrosine Kinase 3/genetics , Animals , Cell Line, Tumor , Drug Synergism , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Mutation/drug effects , Naphthyridines/pharmacology , Protein Biosynthesis/drug effects , Protein Kinase Inhibitors/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors
8.
Sci Rep ; 10(1): 21873, 2020 12 14.
Article En | MEDLINE | ID: mdl-33318517

Glioblastoma (GBM) is the most common and lethal primary intrinsic tumour of the adult brain and evidence indicates disease progression is driven by glioma stem cells (GSCs). Extensive advances in the molecular characterization of GBM allowed classification into proneural, mesenchymal and classical subtypes, and have raised expectations these insights may predict response to targeted therapies. We utilized GBM neurospheres that display GSC characteristics and found activation of the PI3K/AKT pathway in sphere-forming cells. The PI3Kα selective inhibitor alpelisib blocked PI3K/AKT activation and inhibited spheroid growth, suggesting an essential role for the PI3Kα catalytic isoform. p110α expression was highest in the proneural subtype and this was associated with increased phosphorylation of AKT. Further, employing the GBM BioDP, we found co-expression of PIK3CA with the neuronal stem/progenitor marker NES was associated with poor prognosis in PN GBM patients, indicating a unique role for PI3Kα in PN GSCs. Alpelisib inhibited GSC neurosphere growth and these effects were more pronounced in GSCs of the PN subtype. The antineoplastic effects of alpelisib were substantially enhanced when combined with pharmacologic mTOR inhibition. These findings identify the alpha catalytic PI3K isoform as a unique therapeutic target in proneural GBM and suggest that pharmacological mTOR inhibition may sensitize GSCs to selective PI3Kα inhibition.


Brain Neoplasms , Glioma , Neoplastic Stem Cells , Phosphatidylinositol 3-Kinases/metabolism , TOR Serine-Threonine Kinases , Thiazoles/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Cell Line, Tumor , Glioma/drug therapy , Glioma/enzymology , Glioma/pathology , Humans , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-akt/metabolism , Spheroids, Cellular/enzymology , Spheroids, Cellular/pathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
9.
Front Public Health ; 8: 594789, 2020.
Article En | MEDLINE | ID: mdl-33520915

Low radiation doses can affect and modulate cell responses to various stress stimuli, resulting in perturbations leading to resistance or sensitivity to damage. To explore possible mechanisms taking place at an environmental radiation exposure, we set-up twin biological models, one growing in a low radiation environment (LRE) laboratory at the Gran Sasso National Laboratory, and one growing in a reference radiation environment (RRE) laboratory at the Italian National Health Institute (Istituto Superiore di Sanità, ISS). Studies were performed on pKZ1 A11 mouse hybridoma cells, which are derived from the pKZ1 transgenic mouse model used to study the effects of low dose radiation, and focused on the analysis of cellular/molecular end-points, such as proliferation and expression of key proteins involved in stress response, apoptosis, and autophagy. Cells cultured up to 4 weeks in LRE showed no significant differences in proliferation rate compared to cells cultured in RRE. However, caspase-3 activation and PARP1 cleavage were observed in cells entering to an overgrowth state in RRE, indicating a triggering of apoptosis due to growth-stress conditions. Notably, in LRE conditions, cells responded to growth stress by switching toward autophagy. Interestingly, autophagic signaling induced by overgrowth in LRE correlated with activation of p53. Finally, the gamma component of environmental radiation did not significantly influence these biological responses since cells grown in LRE either in incubators with or without an iron shield did not modify their responses. Overall, in vitro data presented here suggest the hypothesis that environmental radiation contributes to the development and maintenance of balance and defense response in organisms.


Apoptosis , Autophagy , Animals , Gamma Rays , Italy , Mice , Signal Transduction
10.
Cancer Immunol Res ; 7(12): 1928-1943, 2019 12.
Article En | MEDLINE | ID: mdl-31530559

The potent immunosuppression induced by glioblastoma (GBM) is one of the primary obstacles to finding effective immunotherapies. One hallmark of the GBM-associated immunosuppressive landscape is the massive infiltration of myeloid-derived suppressor cells (MDSC) and, to a lesser extent, regulatory T cells (Treg) within the tumor microenvironment. Here, we showed that regulatory B cells (Breg) are a prominent feature of the GBM microenvironment in both preclinical models and clinical samples. Forty percent of GBM patients (n = 60) scored positive for B-cell tumor infiltration. Human and mouse GBM-associated Bregs were characterized by immunosuppressive activity toward activated CD8+ T cells, the overexpression of inhibitory molecules PD-L1 and CD155, and production of immunosuppressive cytokines TGFß and IL10. Local delivery of B cell-depleting anti-CD20 immunotherapy improved overall survival of animals (IgG vs. anti-CD20 mean survival: 18.5 vs. 33 days, P = 0.0001), suggesting a potential role of Bregs in GBM progression. We unveiled that GBM-associated MDSCs promoted regulatory B-cell function by delivering microvesicles transporting membrane-bound PD-L1, able to be up-taken by tumoral B cells. The transfer of functional PD-L1 via microvesicles conferred Bregs the potential to suppress CD8+ T-cell activation and acquisition of an effector phenotype. This work uncovered the role of B cells in GBM physiopathology and provides a mechanism by which the GBM microenvironment controls B cell-mediated immunosuppression.See related Spotlight on p. 1902.


B-Lymphocytes/immunology , B7-H1 Antigen/immunology , Brain Neoplasms/immunology , Glioblastoma/immunology , Myeloid-Derived Suppressor Cells/immunology , Animals , Antigens, CD20/genetics , Antigens, CD20/immunology , Brain Neoplasms/genetics , Cell Line, Tumor , Glioblastoma/genetics , Humans , Immune Tolerance , Mice, Inbred C57BL , Mice, Knockout , Tumor Microenvironment/immunology
11.
Oncotarget ; 10(67): 7112-7121, 2019 Dec 24.
Article En | MEDLINE | ID: mdl-31903169

MAPK interacting kinase (MNK), a downstream effector of mitogen-activated protein kinase (MAPK) pathways, activates eukaryotic translation initiation factor 4E (eIF4E) and plays a key role in the mRNA translation of mitogenic and antiapoptotic genes in acute myeloid leukemia (AML) cells. We examined the antileukemic properties of a novel MNK inhibitor, SEL201. Our studies provide evidence that SEL201 suppresses eIF4E phosphorylation on Ser209 in AML cell lines and in primary patient-derived AML cells. Such effects lead to growth inhibitory effects and leukemic cell apoptosis, as well as suppression of leukemic progenitor colony formation. Combination of SEL201 with 5'-azacytidine or rapamycin results in synergistic inhibition of AML cell growth. Collectively, these results suggest that SEL201 has significant antileukemic activity and further underscore the relevance of the MNK pathway in leukemogenesis.

12.
Blood ; 133(11): 1171-1185, 2019 03 14.
Article En | MEDLINE | ID: mdl-30587525

Aberrant activation of mTOR signaling in acute myeloid leukemia (AML) results in a survival advantage that promotes the malignant phenotype. To improve our understanding of factors that contribute to mammalian target of rapamycin (mTOR) signaling activation and identify novel therapeutic targets, we searched for unique interactors of mTOR complexes through proteomics analyses. We identify cyclin dependent kinase 9 (CDK9) as a novel binding partner of the mTOR complex scaffold protein, mLST8. Our studies demonstrate that CDK9 is present in distinct mTOR-like (CTOR) complexes in the cytoplasm and nucleus. In the nucleus, CDK9 binds to RAPTOR and mLST8, forming CTORC1, to promote transcription of genes important for leukemogenesis. In the cytoplasm, CDK9 binds to RICTOR, SIN1, and mLST8, forming CTORC2, and controls messenger RNA (mRNA) translation through phosphorylation of LARP1 and rpS6. Pharmacological targeting of CTORC complexes results in suppression of growth of primitive human AML progenitors in vitro and elicits strong antileukemic responses in AML xenografts in vivo.


Carcinogenesis/drug effects , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , RNA, Messenger/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Biomarkers, Tumor/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Cytarabine/pharmacology , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Nude , Phosphorylation , Protein Biosynthesis , Proteome/analysis , RNA, Messenger/drug effects , RNA, Messenger/genetics , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Sci Signal ; 11(557)2018 11 20.
Article En | MEDLINE | ID: mdl-30459284

It is well established that activation of the transcription factor signal transducer and activator of transcription 1 (STAT1) is required for the interferon-γ (IFN-γ)-mediated antiviral response. Here, we found that IFN-γ receptor stimulation also activated Unc-51-like kinase 1 (ULK1), an initiator of Beclin-1-mediated autophagy. Furthermore, the interaction between ULK1 and the mitogen-activated protein kinase kinase kinase MLK3 (mixed lineage kinase 3) was necessary for MLK3 phosphorylation and downstream activation of the kinase ERK5. This autophagy-independent activity of ULK1 promoted the transcription of key antiviral IFN-stimulated genes (ISGs) and was essential for IFN-γ-dependent antiviral effects. These findings define a previously unknown IFN-γ pathway that appears to be a key element of the antiviral response.


Autophagy-Related Protein-1 Homolog/metabolism , Interferon-gamma/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Animals , Autophagy , Beclin-1/metabolism , Cell Line, Tumor , Class III Phosphatidylinositol 3-Kinases/metabolism , Cytokines/metabolism , Gene Expression Profiling , Humans , Immunity, Innate , Mice , Multigene Family , Phosphorylation , Protein Binding , Receptors, Interferon/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Transcription, Genetic , U937 Cells , Virus Diseases/metabolism , Interferon gamma Receptor , Mitogen-Activated Protein Kinase Kinase Kinase 11
14.
Mol Cell Biol ; 38(16)2018 08 15.
Article En | MEDLINE | ID: mdl-29866656

Although members of the Slfn family have been implicated in the regulation of type I interferon (IFN) responses, the mechanisms by which they mediate their effects remain unknown. In the present study, we provide evidence that targeted disruption of the Slfn2 gene leads to increased transcription of IFN-stimulated genes (ISGs) and enhanced type I IFN-mediated antiviral responses. We demonstrate that Slfn2 interacts with protein phosphatase 6 regulatory subunit 1 (PPP6R1), leading to reduced type I IFN-induced activation of nuclear factor kappa B (NF-κB) signaling, resulting in reduced expression of ISGs. Altogether, these data suggest a novel mechanism by which Slfn2 controls ISG expression and provide evidence for a critical role for Slfn2 in the regulation of IFN-mediated biological responses.


Cell Cycle Proteins/metabolism , Interferon Type I/metabolism , NF-kappa B/metabolism , Animals , Binding Sites/genetics , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cells, Cultured , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , NIH 3T3 Cells , Phosphoprotein Phosphatases/metabolism , Promoter Regions, Genetic , Signal Transduction
15.
Cancer Res ; 78(5): 1275-1292, 2018 03 01.
Article En | MEDLINE | ID: mdl-29279355

T-cell exclusion from the tumor microenvironment (TME) is a major barrier to overcoming immune escape. Here, we identify a myeloid-intrinsic mechanism governed by the NF-κB effector molecule GADD45ß that restricts tumor-associated inflammation and T-cell trafficking into tumors. In various models of solid cancers refractory to immunotherapies, including hepatocellular carcinoma and ovarian adenocarcinoma, Gadd45b inhibition in myeloid cells restored activation of proinflammatory tumor-associated macrophages (TAM) and intratumoral immune infiltration, thereby diminishing oncogenesis. Our results provide a basis to interpret clinical evidence that elevated expression of GADD45B confers poor clinical outcomes in most human cancers. Furthermore, they suggest a therapeutic target in GADD45ß for reprogramming TAM to overcome immunosuppression and T-cell exclusion from the TME.Significance: These findings define a myeloid-based immune checkpoint that restricts T-cell trafficking into tumors, with potentially important therapeutic implications to generally improve the efficacy of cancer immunotherapy. Cancer Res; 78(5); 1275-92. ©2017 AACR.


Antigens, Differentiation/metabolism , Antigens, Differentiation/physiology , Carcinoma, Hepatocellular/immunology , Immune Tolerance/immunology , Immunosuppression Therapy , Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Antigens, Differentiation/genetics , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Female , Humans , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasms/genetics , Neoplasms/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Cells, Cultured
16.
BMC Cancer ; 16: 3, 2016 Jan 05.
Article En | MEDLINE | ID: mdl-26728044

BACKGROUND: Hepatocellular carcinoma (HCC) is the most common malignant tumor of the liver. Non-alcoholic fatty liver disease (NAFLD) is a frequent chronic liver disorder in developed countries. NAFLD can progress through the more severe non alcoholic steatohepatitis (NASH), cirrhosis and, lastly, HCC. Genetic and epigenetic alterations of coding genes as well as deregulation of microRNAs (miRNAs) activity play a role in HCC development. In this study, the C57BL/6J mouse model was long term high-fat (HF) or low-fat (LF) diet fed, in order to analyze molecular mechanisms responsible for the hepatic damage progression. METHODS: Mice were HF or LF diet fed for different time points, then plasma and hepatic tissues were collected. Histological and clinical chemistry assays were performed to assess the progression of liver disease. MicroRNAs' differential expression was evaluated on pooled RNAs from tissues, and some miRNAs showing dysregulation were further analyzed at the individual level. RESULTS: Cholesterol, low and high density lipoproteins, triglycerides and alanine aminotransferase increase was detected in HF mice. Gross anatomical examination revealed hepatomegaly in HF livers, and histological analysis highlighted different degrees and levels of steatosis, inflammatory infiltrate and fibrosis in HF and LF animals, demonstrating the progression from NAFLD through NASH. Macroscopic nodules, showing typical neoplastic features, were observed in 20% of HF diet fed mice. Fifteen miRNAs differentially expressed in HF with respect to LF hepatic tissues during the progression of liver damage, and in tumors with respect to HF non tumor liver specimens were identified. Among them, miR-340-5p, miR-484, miR-574-3p, miR-720, whose expression was never described in NAFLD, NASH and HCC tissues, and miR-125a-5p and miR-182, which showed early and significant dysregulation in the sequential hepatic damage process. CONCLUSIONS: In this study, fifteen microRNAs which were modulated in hepatic tissues and in tumors during the transition NAFLD-NASH-HCC are reported. Besides some already described, new and early dysregulated miRNAs were identified. Functional analyses are needed to validate the results here obtained, and to better define the role of these molecules in the progression of the hepatic disease.


Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , MicroRNAs/biosynthesis , Non-alcoholic Fatty Liver Disease/genetics , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cholesterol/blood , Diet, High-Fat , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , Mice , MicroRNAs/genetics , Non-alcoholic Fatty Liver Disease/pathology , Triglycerides/blood
17.
Anticancer Agents Med Chem ; 16(1): 7-19, 2016.
Article En | MEDLINE | ID: mdl-26299662

MicroRNAs are small non-coding RNAs which regulate gene expression and silence a wide set of target genes. Aberrant miRNA expression has been described in cancer cells and is at least in part responsible of cancer initiation, development and progression. Due to their role, miRNAs have emerged as therapeutic targets or molecules suitable at the therapeutic level as well as markers of the response to chemo/radio/targeted therapy. Restoration or repression of miRNAs expression and activity shows high potential in managing cancer, and many studies on pre-clinical models have demonstrated the feasibility and efficacy of miRNA-based therapy. However, despite the exciting potential, some limitations, due to the degree of delivery and biodistribution or to possible side effects, need to be taken into consideration and solved in order to accomplish transition to clinical application. In this review we report and discuss the role of miRNAs in cancer, focusing on their use as therapeutic agents and their involvement in modulating/affecting the response to chemo/radio/targeted therapy in some of the most frequent solid tumors.


MicroRNAs/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Animals , Humans , MicroRNAs/genetics
18.
Curr Drug Targets ; 17(3): 311-20, 2016.
Article En | MEDLINE | ID: mdl-26343112

Rel/NF-κB transcription factors are key regulators of genes implicated in inflammatory and immune activation, cell growth and protection from apoptosis. Constitutive activation of NF-κB has been observed in several types of cancers. Recently, it has been shown that inflammation and cancer are molecularly linked by means of NF-κB. During prostate cancer progression, NF-κB promotes cell survival, tumor invasion, metastasis and chemoresistance. NF-κB constitutive activation has been frequently demonstrated in primary prostate cancers and it correlates with loss of androgen receptor expression and castration-resistant phenotypes. Indeed, inhibition of NF-κB pathway may reduce the oncogenic effects mediated by chronic inflammatory response. Therefore, NF-κB represents a hopeful target for the treatment of prostate cancer due to its role in oncogenesis and chemoresistance. Here, the current knowledge about the roles of NF-κB signaling pathway in prostate tumorigenesis is discussed, taking into consideration the potentiality and effectiveness of NF-κB inhibitors as therapeutic agents for prostate cancer.


Antineoplastic Agents/pharmacology , NF-kappa B/metabolism , Prostatic Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Cell Survival/drug effects , Clinical Trials as Topic , Drug Resistance, Neoplasm/drug effects , Humans , Male , Molecular Targeted Therapy , Prostatic Neoplasms/metabolism , Signal Transduction/drug effects , Treatment Outcome
19.
Arthritis Res Ther ; 17: 247, 2015 Sep 10.
Article En | MEDLINE | ID: mdl-26357964

INTRODUCTION: Systemic sclerosis (SSc) is a complex and not fully understood autoimmune disease associated with fibrosis of multiple organs. The main effector cells, the myofibroblasts, are collagen-producing cells derived from the activation of resting fibroblasts. This process is regulated by a complex repertoire of profibrotic cytokines, and among them transforming growth factor beta (TGF-ß) and endothelin-1 (ET-1) play a major role. In this paper we show that TGF-ß and ET-1 receptors co-operate in myofibroblast activation, and macitentan, an ET-1 receptor antagonist binding ET-1 receptors, might interfere with both TGF-ß and ET-1 pathways, preventing myofibroblast differentiation. METHODS: Fibroblasts isolated from healthy controls and SSc patients were treated with TGF-ß and ET-1 and successively analyzed for alpha smooth muscle actin (α-SMA) and collagen (Col1A1) expression and for the Sma and Mad Related (SMAD) phosphorylation. We further tested the ability of macitentan to interfere with these process. Furthermore, we silenced ET-1 and endothelin-1 receptor A expression and evaluated the formation of an ET-1/TGF-ß receptor complex by immunoprecitation assay. RESULTS: We showed myofibroblast activation in SSc fibroblasts assessing the expression of α-SMA and Col1A1, after stimulation with TGF-ß and ET-1. Macitentan interfered with both ET-1- and TGF-ß-induced fibroblast activation. To explain this unexpected inhibitory effect of macitentan on TGF-ß activity, we silenced ET-1 expression on SSc fibroblasts and co-immunoprecipitated these two receptors, showing the formation of an ET-1/TGF-ß receptor complex. CONCLUSIONS: During SSc, ET-1 produced by activated endothelia contributes to myofibroblast activation using TGF-ß machinery via an ET-1/TGF-ß receptor complex. Macitentan interferes with the profibrotic action of TGF-ß, blocking the ET-1 receptor portion of the ET-1/TGF-ß receptor complex.


Fibroblasts/drug effects , Pyrimidines/pharmacology , Receptor, Endothelin A/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Sulfonamides/pharmacology , Transforming Growth Factor beta1/pharmacology , Actins/metabolism , Adult , Blotting, Western , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Dermis/pathology , Endothelin A Receptor Antagonists/pharmacology , Endothelin-1/pharmacology , Female , Fibroblasts/metabolism , Humans , Male , Middle Aged , Multiprotein Complexes/metabolism , Phosphorylation/drug effects , RNA Interference , Receptor, Endothelin A/genetics , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Signal Transduction/drug effects , Smad Proteins/metabolism , Up-Regulation/drug effects , Young Adult
20.
Biomed Res Int ; 2014: 380398, 2014.
Article En | MEDLINE | ID: mdl-25045667

Prostate cancer is the most common noncutaneous cancer among men in the United States. A genetic contribution to prostate cancer risk has been documented, but knowledge of the molecular mechanisms involved in prostate cancer initiation is still not well understood. Loss of heterozygosity (LOH) of chromosomal regions is crucial in tumor progression. In human prostate cancer, several chromosomal regions demonstrating a high frequency of LOH have been previously identified. KCTD11 (REN) is a tumor suppressor gene mapping on human chromosome 17p13.2, whose expression is frequently lost in human medulloblastoma and in several other cancer types. KCTD11 acts as a negative regulator of the Hedgehog (Hh) signaling. Here, we demonstrated that KCTD11 LOH is a common genetic lesion in human prostate adenocarcinoma. Indeed, nuclear KCTD11 protein expression is strongly reduced in primary prostate cancer, and this event correlated with overexpression of proteins acting into the Hedgehog pathway. Low levels of KCTD11 mRNA have been also observed in prostatic cancer cells, and ectopic overexpression of KCTD11 led to growth arrest. Our study demonstrates and supports that KCTD11, as well as negatively regulated downstream effectors belonging to Hh signaling, plays a role in prostate cancer pathogenesis. This could be suitable to characterize new diagnostic and therapeutic markers.


Adenocarcinoma/genetics , Genes, Tumor Suppressor , Potassium Channels/biosynthesis , Prostatic Neoplasms/genetics , Adenocarcinoma/pathology , Cell Cycle Proteins , Chromosome Deletion , Gene Expression Regulation, Neoplastic , Humans , Loss of Heterozygosity , Male , Potassium Channels/genetics , Prostatic Neoplasms/pathology , Signal Transduction/genetics , Transferases
...