Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Invest Dermatol ; 144(7): 1579-1589.e8, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38219917

ABSTRACT

Cutaneous T-cell lymphomas are mature lymphoid neoplasias resulting from the malignant transformation of skin-resident T-cells. A distinctive clinical feature of cutaneous T-cell lymphomas is their sensitivity to treatment with histone deacetylase inhibitors. However, responses to histone deacetylase inhibitor therapy are universally transient and noncurative, highlighting the need for effective and durable drug combinations. In this study, we demonstrate that the combination of romidepsin, a selective class I histone deacetylase inhibitor, with afatinib, an EGFR family inhibitor, induces strongly synergistic antitumor effects in cutaneous T-cell lymphoma models in vitro and in vivo through abrogation of Jak-signal transducer and activator of transcription signaling. These results support a previously unrecognized potential role for histone deacetylase inhibitor plus afatinib combination in the treatment of cutaneous T-cell lymphomas.


Subject(s)
Afatinib , Depsipeptides , Drug Synergism , Lymphoma, T-Cell, Cutaneous , Signal Transduction , Skin Neoplasms , Depsipeptides/pharmacology , Depsipeptides/administration & dosage , Lymphoma, T-Cell, Cutaneous/drug therapy , Lymphoma, T-Cell, Cutaneous/pathology , Humans , Animals , Mice , Afatinib/pharmacology , Signal Transduction/drug effects , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Cell Line, Tumor , Janus Kinases/metabolism , Janus Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use
2.
Nat Commun ; 14(1): 1328, 2023 03 10.
Article in English | MEDLINE | ID: mdl-36899004

ABSTRACT

The TINCR (Terminal differentiation-Induced Non-Coding RNA) gene is selectively expressed in epithelium tissues and is involved in the control of human epidermal differentiation and wound healing. Despite its initial report as a long non-coding RNA, the TINCR locus codes for a highly conserved ubiquitin-like microprotein associated with keratinocyte differentiation. Here we report the identification of TINCR as a tumor suppressor in squamous cell carcinoma (SCC). TINCR is upregulated by UV-induced DNA damage in a TP53-dependent manner in human keratinocytes. Decreased TINCR protein expression is prevalently found in skin and head and neck squamous cell tumors and TINCR expression suppresses the growth of SCC cells in vitro and in vivo. Consistently, Tincr knockout mice show accelerated tumor development following UVB skin carcinogenesis and increased penetrance of invasive SCCs. Finally, genetic analyses identify loss-of-function mutations and deletions encompassing the TINCR gene in SCC clinical samples supporting a tumor suppressor role in human cancer. Altogether, these results demonstrate a role for TINCR as protein coding tumor suppressor gene recurrently lost in squamous cell carcinomas.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , RNA, Long Noncoding , Animals , Mice , Humans , Ubiquitin/metabolism , Carcinoma, Squamous Cell/genetics , Genes, Tumor Suppressor , Keratinocytes/metabolism , Head and Neck Neoplasms/genetics , RNA, Long Noncoding/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Micropeptides
3.
Cancer Discov ; 12(11): 2646-2665, 2022 11 02.
Article in English | MEDLINE | ID: mdl-35984649

ABSTRACT

Low-intensity maintenance therapy with 6-mercaptopurine (6-MP) limits the occurrence of acute lymphoblastic leukemia (ALL) relapse and is central to the success of multiagent chemotherapy protocols. Activating mutations in the 5'-nucleotidase cytosolic II (NT5C2) gene drive resistance to 6-MP in over 35% of early relapse ALL cases. Here we identify CRCD2 as a first-in-class small-molecule NT5C2 nucleotidase inhibitor broadly active against leukemias bearing highly prevalent relapse-associated mutant forms of NT5C2 in vitro and in vivo. Importantly, CRCD2 treatment also enhanced the cytotoxic activity of 6-MP in NT5C2 wild-type leukemias, leading to the identification of NT5C2 Ser502 phosphorylation as a novel NT5C2-mediated mechanism of 6-MP resistance in this disease. These results uncover an unanticipated role of nongenetic NT5C2 activation as a driver of 6-MP resistance in ALL and demonstrate the potential of NT5C2 inhibitor therapy for enhancing the efficacy of thiopurine maintenance therapy and overcoming resistance at relapse. SIGNIFICANCE: Relapse-associated NT5C2 mutations directly contribute to relapse in ALL by driving resistance to chemotherapy with 6-MP. Pharmacologic inhibition of NT5C2 with CRCD2, a first-in-class nucleotidase inhibitor, enhances the cytotoxic effects of 6-MP and effectively reverses thiopurine resistance mediated by genetic and nongenetic mechanisms of NT5C2 activation in ALL. This article is highlighted in the In This Issue feature, p. 2483.


Subject(s)
Antineoplastic Agents , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Mercaptopurine/pharmacology , Mercaptopurine/therapeutic use , 5'-Nucleotidase/genetics , 5'-Nucleotidase/therapeutic use , Drug Resistance, Neoplasm/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Antineoplastic Agents/therapeutic use , Recurrence
4.
Cells ; 10(4)2021 03 25.
Article in English | MEDLINE | ID: mdl-33806062

ABSTRACT

The timing of centrosome separation and the distance moved apart influence the formation of the bipolar spindle, affecting chromosome stability. Epidermal growth factor receptor (EGFR) signaling induces early centrosome separation through downstream G protein-coupled receptor kinase GRK2, which phosphorylates the Hippo pathway component MST2 (Mammalian STE20-like protein kinase 2), in turn allowing NIMA kinase Nek2A activation for centrosomal linker disassembly. However, the mechanisms that counterbalance centrosome disjunction and separation remain poorly understood. We unveil that timely degradation of GRK2 by the E3 ligase Mdm2 limits centrosome separation in the G2. Both knockout expression and catalytic inhibition of Mdm2 result in GRK2 accumulation and enhanced centrosome separation before mitosis onset. Phosphorylation of GRK2 on residue S670 enables a complex pattern of non-K48-linked polyubiquitin chains assembled by Mdm2, which correlate with kinase protein degradation. Remarkably, GRK2-S670A protein fails to phosphorylate MST2 despite overcoming Mdm2-dependent degradation, which results in defective centrosome separation, shorter spindles, and abnormal chromosome congression. Conversely, extra levels of wild-type kinase in the G2 cause increased inter-centrosome distances with longer spindles, also converging in congression issues. Our findings show that the signals enabling activity of the GRK2/MST2/Nek2A axis for separation also switches on Mdm2 degradation of GRK2 to ensure accurate centrosome dynamics and proper mitotic spindle functionality.


Subject(s)
Centrosome/metabolism , Chromosome Segregation , Down-Regulation , G-Protein-Coupled Receptor Kinase 2/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Animals , G2 Phase , HEK293 Cells , HeLa Cells , Humans , Mice, Knockout , Phosphorylation , Phosphoserine/metabolism , Proteolysis , Spindle Apparatus/metabolism , Ubiquitination
5.
Nat Cancer ; 2(1): 98-113, 2021 01.
Article in English | MEDLINE | ID: mdl-33928261

ABSTRACT

Angioimmunoblastic T cell lymphoma (AITL) and peripheral T cell lymphoma not-otherwise-specified (PTCL, NOS) have poor prognosis and lack driver actionable targets for directed therapies in most cases. Here we identify FYN-TRAF3IP2 as a recurrent oncogenic gene fusion in AITL and PTCL, NOS tumors. Mechanistically, we show that FYN-TRAF3IP2 leads to aberrant NF-κB signaling downstream of T cell receptor activation. Consistent with a driver oncogenic role, FYN-TRAF3IP2 expression in hematopoietic progenitors induces NF-κB-driven T cell transformation in mice and cooperates with loss of the Tet2 tumor suppressor in PTCL development. Moreover, abrogation of NF-κB signaling in FYN-TRAF3IP2-induced tumors with IκB kinase inhibitors delivers strong anti-lymphoma effects in vitro and in vivo. These results demonstrate an oncogenic and pharmacologically targetable role for FYN-TRAF3IP2 in PTCLs and call for the clinical testing of anti-NF-κB targeted therapies in these diseases.


Subject(s)
Immunoblastic Lymphadenopathy , Lymphoma, T-Cell, Peripheral , Adaptor Proteins, Signal Transducing/genetics , Animals , Immunoblastic Lymphadenopathy/genetics , Lymphoma, T-Cell, Peripheral/genetics , Mice , NF-kappa B/genetics , Oncogenes , Signal Transduction
6.
Cancers (Basel) ; 12(5)2020 May 13.
Article in English | MEDLINE | ID: mdl-32413989

ABSTRACT

Adaptation to hypoxia is a common feature in solid tumors orchestrated by oxygen-dependent and independent upregulation of the hypoxia-inducible factor-1α (HIF-1α). We unveiled that G protein-coupled receptor kinase (GRK2), known to be overexpressed in certain tumors, fosters this hypoxic pathway via phosphorylation of the mRNA-binding protein HuR, a central HIF-1α modulator. GRK2-mediated HuR phosphorylation increases the total levels and cytoplasmic shuttling of HuR in response to hypoxia, and GRK2-phosphodefective HuR mutants show defective cytosolic accumulation and lower binding to HIF-1α mRNA in hypoxic Hela cells. Interestingly, enhanced GRK2 and HuR expression correlate in luminal breast cancer patients. GRK2 also promotes the HuR/HIF-1α axis and VEGF-C accumulation in normoxic MCF7 breast luminal cancer cells and is required for the induction of HuR/HIF1-α in response to adrenergic stress. Our results point to a relevant role of the GRK2/HuR/HIF-1α module in the adaptation of malignant cells to tumor microenvironment-related stresses.

7.
Nutrients ; 11(11)2019 Nov 16.
Article in English | MEDLINE | ID: mdl-31744117

ABSTRACT

Cancer mortality rates are undergoing a global downward trend; however, metastasis and relapse after surgery and adjuvant treatments still correlate with poor prognosis and represent the most significant challenges in the treatment of this disease. Advances in genomics, metabolomics, and proteomics are improving our understanding regarding cancer metabolic diversity, resulting in detailed classifications of tumors and raising the effectiveness of precision medicine. Likewise, the growing knowledge of interactions between nutrients and the expression of certain genes could lead to cancer therapies based on precision nutrition strategies. This review aims to identify the recent advances in the knowledge of the mechanistic role of bioactive phytochemicals in foodstuffs in tumor progression, metastasis, and chemo-resistance in order to assess their potential use in precision nutrition therapies targeting relapse in lung, breast, colon, and prostate cancer, and leukemia. A considerable number of bioactive phytochemicals in foodstuffs were identified in the literature with proven effects modulating tumor growth, progression, and metastasis. In addition, the use of foodstuffs in cancer, and specifically in relapse therapies, is being reinforced by the development of different formulations that significantly increase the therapeutic efficiency of these products. This can open the possibility for testing combinations of bioactive phytochemicals with cancer relapse treatments as a potential prevention strategy.


Subject(s)
Neoplasm Recurrence, Local/prevention & control , Neoplasms/therapy , Nutrition Therapy/methods , Phytochemicals/therapeutic use , Precision Medicine/methods , Breast Neoplasms/therapy , Colonic Neoplasms/therapy , Female , Humans , Leukemia/therapy , Lung Neoplasms/therapy , Male , Prostatic Neoplasms/therapy
8.
Cancer Discov ; 9(12): 1774-1791, 2019 12.
Article in English | MEDLINE | ID: mdl-31519704

ABSTRACT

Long-range enhancers govern the temporal and spatial control of gene expression; however, the mechanisms that regulate enhancer activity during normal and malignant development remain poorly understood. Here, we demonstrate a role for aberrant chromatin accessibility in the regulation of MYC expression in T-cell lymphoblastic leukemia (T-ALL). Central to this process, the NOTCH1-MYC enhancer (N-Me), a long-range T cell-specific MYC enhancer, shows dynamic changes in chromatin accessibility during T-cell specification and maturation and an aberrant high degree of chromatin accessibility in mouse and human T-ALL cells. Mechanistically, we demonstrate that GATA3-driven nucleosome eviction dynamically modulates N-Me enhancer activity and is strictly required for NOTCH1-induced T-ALL initiation and maintenance. These results directly implicate aberrant regulation of chromatin accessibility at oncogenic enhancers as a mechanism of leukemic transformation. SIGNIFICANCE: MYC is a major effector of NOTCH1 oncogenic programs in T-ALL. Here, we show a major role for GATA3-mediated enhancer nucleosome eviction as a driver of MYC expression and leukemic transformation. These results support the role of aberrant chromatin accessibility and consequent oncogenic MYC enhancer activation in NOTCH1-induced T-ALL.This article is highlighted in the In This Issue feature, p. 1631.


Subject(s)
Enhancer Elements, Genetic , GATA3 Transcription Factor/metabolism , Leukemia, T-Cell/pathology , Nucleosomes/metabolism , Proto-Oncogene Proteins c-myc/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Jurkat Cells , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Mice , Neoplasm Transplantation , Receptor, Notch1/metabolism
9.
Semin Cancer Biol ; 48: 78-90, 2018 02.
Article in English | MEDLINE | ID: mdl-28473253

ABSTRACT

Increasing evidences point to G protein-coupled receptor kinases (GRKs), a subfamily of protein kinase A/G/C-like kinases, as relevant players in cancer progression, in a cell-type and tumor-specific way. Alterations in the expression and/or activity of particular GRKs have been identified in several types of tumors, and demonstrated to modulate the proliferation, survival or invasive properties of tumor cells by acting as integrating signaling nodes. GRKs are able to regulate the functionality of both G protein-coupled receptors (GPCR) and growth factor receptors and to directly control cytosolic, cytoskeletal or nuclear signaling components of pathways relevant for these processes. Furthermore, many chemokines as well as angiogenic and inflammatory factors present in the tumor microenvironment act through GPCR and other GRK-modulated signaling modules. Changes in the dosage of certain GRKs in the tumor stroma can alter tumor angiogenesis and the homing of immune cells, thus putting forward these kinases as potentially relevant modulators of the carcinoma-fibroblast-endothelial-immune cell network fostering tumor development and dissemination. A better understanding of the alterations in different GRK isoforms taking place during cancer development and metastasis in specific tumors and cell types and of its impact in signaling pathways would help to design novel therapeutic strategies.


Subject(s)
G-Protein-Coupled Receptor Kinases/physiology , Neoplasms/pathology , Animals , Carcinogenesis/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Tumor Microenvironment
10.
Mol Pharmacol ; 91(3): 220-228, 2017 03.
Article in English | MEDLINE | ID: mdl-27895163

ABSTRACT

Malignant features-such as sustained proliferation, refractoriness to growth suppressors, resistance to cell death or aberrant motility, and metastasis-can be triggered by a variety of mutations and signaling adaptations. Signaling nodes can act as cancer-associated factors by cooperating with oncogene-governed pathways or participating in compensatory transduction networks to strengthen tumor properties. G-protein-coupled receptor kinase 2 (GRK2) is arising as one of such nodes. Via its complex network of connections with other cellular proteins, GRK2 contributes to the modulation of basic cellular functions-such as cell proliferation, survival, or motility-and is involved in metabolic homeostasis, inflammation, or angiogenic processes. Moreover, altered GRK2 levels are starting to be reported in different tumoral contexts and shown to promote breast tumorigenesis or to trigger the tumoral angiogenic switch. The ability to modulate several of the hallmarks of cancer puts forward GRK2 as an oncomodifier, able to modulate carcinogenesis in a cell-type specific way.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , Neoplasms/enzymology , Animals , Cell Proliferation , Humans , Metabolic Networks and Pathways , Neoplasms/blood supply , Neoplasms/pathology , Signal Transduction , Tumor Microenvironment
11.
EBioMedicine ; 13: 132-145, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27720394

ABSTRACT

In addition to oncogenic drivers, signaling nodes can critically modulate cancer-related cellular networks to strength tumor hallmarks. We identify G-protein-coupled receptor kinase 2 (GRK2) as a relevant player in breast cancer. GRK2 is up-regulated in breast cancer cell lines, in spontaneous tumors in mice, and in a proportion of invasive ductal carcinoma patients. Increased GRK2 functionality promotes the phosphorylation and activation of the Histone Deacetylase 6 (HDAC6) leading to de-acetylation of the Prolyl Isomerase Pin1, a central modulator of tumor progression, thereby enhancing its stability and functional interaction with key mitotic regulators. Interestingly, a correlation between GRK2 expression and Pin1 levels and de-acetylation status is detected in breast cancer patients. Activation of the HDAC6-Pin1 axis underlies the positive effects of GRK2 on promoting growth factor signaling, cellular proliferation and anchorage-independent growth in both luminal and basal breast cancer cells. Enhanced GRK2 levels promote tumor growth in mice, whereas GRK2 down-modulation sensitizes cells to therapeutic drugs and abrogates tumor formation. Our data suggest that GRK2 acts as an important onco-modulator by strengthening the functionality of key players in breast tumorigenesis such as HDAC6 and Pin1.


Subject(s)
Breast Neoplasms/metabolism , Cell Transformation, Neoplastic/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , Histone Deacetylases/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Signal Transduction , Acetylation , Animals , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Disease Models, Animal , Female , G-Protein-Coupled Receptor Kinase 2/genetics , Gene Expression , Histone Deacetylase 6 , Histone Deacetylases/genetics , Humans , Mice, Transgenic , Models, Biological , Prognosis , RNA Interference , RNA, Small Interfering/genetics , Tumor Burden
12.
Mol Cell Oncol ; 1(4): e969166, 2014.
Article in English | MEDLINE | ID: mdl-27308373

ABSTRACT

Downregulation of G protein-coupled receptor kinase 2 (GRK2) in endothelial cells has recently been identified as a relevant event in the tumoral angiogenic switch. Based on the effects of altering GRK2 dosage in cell and animal models, this kinase appears to act as a hub in key signaling pathways involved in vascular stabilization and remodeling. Accordingly, decreased GRK2 expression in endothelial cells accelerates tumor growth in mice by impairing the pericytes ensheathing the vessels, thereby promoting hypoxia and macrophage infiltration. These results raise new questions regarding the mechanisms by which transformed cells trigger the decrease in GRK2 observed in human breast cancer vessels and how GRK2 modulates the interactions between different cell types that occur in the tumor microenvironment.

13.
J Clin Invest ; 123(11): 4714-30, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24135140

ABSTRACT

Tumor vessel dysfunction is a pivotal event in cancer progression. Using an in vivo neovascularization model, we identified G protein-coupled receptor kinase 2 (GRK2) as a key angiogenesis regulator. An impaired angiogenic response involving immature vessels was observed in mice hemizygous for Grk2 or in animals with endothelium-specific Grk2 silencing. ECs isolated from these animals displayed intrinsic alterations in migration, TGF-ß signaling, and formation of tubular networks. Remarkably, an altered pattern of vessel growth and maturation was detected in postnatal retinas from endothelium-specific Grk2 knockout animals. Mouse embryos with systemic or endothelium-selective Grk2 ablation had marked vascular malformations involving impaired recruitment of mural cells. Moreover, decreased endothelial Grk2 dosage accelerated tumor growth in mice, along with reduced pericyte vessel coverage and enhanced macrophage infiltration, and this transformed environment promoted decreased GRK2 in ECs and human breast cancer vessels. Our study suggests that GRK2 downregulation is a relevant event in the tumoral angiogenic switch.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/physiology , Neovascularization, Pathologic , Neovascularization, Physiologic , Activin Receptors, Type I/physiology , Activin Receptors, Type II , Animals , Cell Movement , Cell Proliferation , Endothelial Cells/pathology , Endothelial Cells/physiology , Female , G-Protein-Coupled Receptor Kinase 2/deficiency , G-Protein-Coupled Receptor Kinase 2/genetics , Hemizygote , Humans , Melanoma, Experimental/blood supply , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Knockout , Neovascularization, Pathologic/genetics , Neovascularization, Physiologic/genetics , Pregnancy , Protein Serine-Threonine Kinases/physiology , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/physiology , Retinal Vessels/abnormalities , Retinal Vessels/embryology , Signal Transduction , Transforming Growth Factor beta1/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...