1
|
Cusack R, Bos LD, Povoa P, Martin-Loeches I. Endothelial dysfunction triggers acute respiratory distress syndrome in patients with sepsis: a narrative review. Front Med (Lausanne) 2023; 10:1203827. [PMID: 37332755 PMCID: PMC10272540 DOI: 10.3389/fmed.2023.1203827] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/16/2023] [Indexed: 06/20/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a severe organ failure occurring mainly in critically ill patients as a result of different types of insults such as sepsis, trauma or aspiration. Sepsis is the main cause of ARDS, and it contributes to a high mortality and resources consumption both in hospital setting and in the community. ARDS develops mainly an acute respiratory failure with severe and often refractory hypoxemia. ARDS also has long term implications and sequelae. Endothelial damage plays an important role in the pathogenesis of ARDS. Understanding the mechanisms of ARDS presents opportunities for novel diagnostic and therapeutic targets. Biochemical signals can be used in concert to identify and classify patients into ARDS phenotypes allowing earlier effective treatment with personalised therapies. This is a narrative review where we aimed to flesh out the pathogenetic mechanisms and heterogeneity of ARDS. We examine the links between endothelium damage and its contribution to organ failure. We have also investigated future strategies for treatment with a special emphasis in endothelial damage.
Collapse
Affiliation(s)
- Rachael Cusack
- Department of Intensive Care, St. James’s Hospital, Dublin, Ireland
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Lieuwe D. Bos
- Intensive Care, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Pedro Povoa
- NOVA Medical School, CHRC, New University of Lisbon, Lisbon, Portugal
- Center for Clinical Epidemiology and Research Unit of Clinical Epidemiology, OUH Odense University Hospital, Odense, Denmark
- Department of Intensive Care, Hospital de São Francisco Xavier, CHLO, Lisbon, Portugal
| | - Ignacio Martin-Loeches
- Department of Intensive Care, St. James’s Hospital, Dublin, Ireland
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
2
|
Neha, Parvez S. Emerging therapeutics agents and recent advances in drug repurposing for Alzheimer's disease. Ageing Res Rev 2023; 85:101815. [PMID: 36529440 DOI: 10.1016/j.arr.2022.101815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is a multivariate and diversified disease and affects the most sensitive areas of the brain, the cerebral cortex, and the hippocampus. AD is a progressive age-related neurodegenerative disease most often associated with memory deficits and cognition that get more worsen over time. The central theory on the pathophysiological hallmark features of AD is characterized by the accumulation of amyloid β (Aβ) peptides, also associated with tau proteins (τ) dysfunctioning which leads to distorted microtubular structure, affects the cholinergic system, and mitochondrial biogenesis. This review emphasizes how simple it is to find novel treatments for AD and focuses on several recently developed medications through repurposing that can speed up traditional drug development.
Collapse
Affiliation(s)
- Neha
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
3
|
Li Y, Sun Z, Zhu H, Sun Y, Shteyman DB, Markx S, Leong KW, Xu B, Fu BM. Inhibition of Abl Kinase by Imatinib Can Rescue the Compromised Barrier Function of 22q11.2DS Patient-iPSC-Derived Blood-Brain Barriers. Cells 2023; 12:422. [PMID: 36766762 PMCID: PMC9913366 DOI: 10.3390/cells12030422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/06/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
We have previously established that the integrity of the induced blood-brain barrier (iBBB) formed by brain microvascular endothelial cells derived from the iPSC of 22q11.2 DS (22q11.2 Deletion Syndrome, also called DiGeorge Syndrome) patients is compromised. We tested the possibility that the haploinsufficiency of CRKL, a gene within the 22q11.2 DS deletion region, contributes to the deficit. The CRKL is a major substrate of the Abl tyrosine kinase, and the Abl/CRKL signaling pathway is critical for endothelial barrier functions. Imatinib, an FDA-approved drug, inhibits Abl kinase and has been used to treat various disorders involving vascular leakages. To test if imatinib can restore the compromised iBBB, we treated the patient's iBBB with imatinib. After treatment, both trans-endothelial electrical resistance and solute permeability returned to comparable levels of the control iBBB. Correspondingly, changes in tight junctions and endothelial glycocalyx of the iBBB were also restored. Western blotting showed that imatinib increased the level of active forms of the CRKL protein. A transcriptome study revealed that imatinib up-regulated genes in the signaling pathways responsible for the protein modification process and down-regulated those for cell cycling. The KEGG pathway analysis further suggested that imatinib improved the gene expression of the CRKL signaling pathway and tight junctions, which agrees with our expectations and the observations at protein levels. Our results indicate that the 22q11.2DS iBBB is at least partially caused by the haploinsufficiency of CRKL, which can be rescued by imatinib via its effects on the Abl/CRKL signaling pathway. Our findings uncover a novel disease mechanism associated with 22q11.2DS.
Collapse
Affiliation(s)
- Yunfei Li
- Department of Biomedical Engineering, The City College of the City University of New York, New York, NY 10031, USA
| | - Zhixiong Sun
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Huixiang Zhu
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Yan Sun
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - David B. Shteyman
- Department of Biomedical Engineering, The City College of the City University of New York, New York, NY 10031, USA
| | - Sander Markx
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Bin Xu
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Bingmei M. Fu
- Department of Biomedical Engineering, The City College of the City University of New York, New York, NY 10031, USA
| |
Collapse
|
4
|
Fakhari S, Jamzad M, Nouri A, Arab-Salmanabadi S, Falaki F. A novel polyamidoamine dendrimer based nano-carrier for oral delivery of imatinib. JOURNAL OF POLYMER RESEARCH 2022. [DOI: 10.1007/s10965-022-03359-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
5
|
Morini M, Gentilini F, Turba ME, Gobbo F, Mandrioli L, Bettini G. Mutational Analysis of c-KIT and PDGFRA in Canine Gastrointestinal Stromal Tumors (GISTs). Vet Sci 2022; 9:vetsci9070376. [PMID: 35878393 PMCID: PMC9323380 DOI: 10.3390/vetsci9070376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the canine gastrointestinal tract and are diagnosed by the immunohistochemical expression of the receptor tyrosine kinase (RTK) KIT. Activating mutations of the proto-oncogenes c-KIT and PDGFRA drive GIST oncogenesis and are used to predict the response to RTK-inhibitors in human oncology. Currently, the frequency and significance of these mutations in canine GIST have not been adequately explored. Therefore, we investigated the mutational status of c-KIT (exons 9, 11 and 13) and PDGFRA (exons 12 and 18) genes by PCR followed by fragment analysis for c-KIT deletions and PCR followed by screening with DHPLC and direct sequencing confirmation for single nucleotide variations in 17 formalin-fixed paraffin-embedded canine GISTs confirmed by KIT immunopositivity. c-KIT mutations were detected in 47% of cases, with a mutation detection rate significantly higher (p = 0.0004, Fisher’s exact test) and always involving exon 11. A PDGFRA gene mutation (exon 18) was identified in one case. Even if follow-up data were not available for all cases, four cases with documented abdominal metastases displayed c-KIT mutations. These data confirm that c-KIT exon 11 mutations occur frequently in canine GISTs, and identify the presence of a PDGFRA mutation similar to human GISTs. This study also suggests a potential association of c-KIT mutation with more aggressive biological behavior.
Collapse
Affiliation(s)
- Maria Morini
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell’Emilia, 40064 Bologna, Italy; (F.G.); (F.G.); (L.M.); (G.B.)
- Correspondence:
| | - Fabio Gentilini
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell’Emilia, 40064 Bologna, Italy; (F.G.); (F.G.); (L.M.); (G.B.)
| | | | - Francesca Gobbo
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell’Emilia, 40064 Bologna, Italy; (F.G.); (F.G.); (L.M.); (G.B.)
| | - Luciana Mandrioli
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell’Emilia, 40064 Bologna, Italy; (F.G.); (F.G.); (L.M.); (G.B.)
| | - Giuliano Bettini
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell’Emilia, 40064 Bologna, Italy; (F.G.); (F.G.); (L.M.); (G.B.)
| |
Collapse
|
6
|
Isabelle C, Boles A, Chakravarti N, Porcu P, Brammer J, Mishra A. Cytokines in the Pathogenesis of Large Granular Lymphocytic Leukemia. Front Oncol 2022; 12:849917. [PMID: 35359386 PMCID: PMC8960188 DOI: 10.3389/fonc.2022.849917] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/08/2022] [Indexed: 12/25/2022] Open
Abstract
Large granular lymphocytic leukemia (LGLL) is a lymphoproliferative disorder of older adults characterized by the clonal expansion of cytotoxic T/natural killer cells due to constitutive pro-survival signaling. In recent years, it has become clear that cytokines and their receptors are aberrantly expressed in LGLL cells. The exact initiation process of LGLL is unknown, although several cytokine-driven mechanisms have emerged. Elevated levels of several cytokines, including interleukin-15 (IL-15) and platelet-derived growth factor (PDGF), have been described in LGLL patients. Evidence from humans and animal models has shown that cytokines may also contribute to the co-occurrence of a wide range of autoimmune diseases seen in patients with LGLL. The goal of this review is to provide a comprehensive analysis of the link between cytokines and pro-survival signaling in LGLL and to discuss the various strategies and research approaches that are being utilized to study this link. This review will also highlight the importance of cytokine-targeted therapeutics in the treatment of LGLL.
Collapse
Affiliation(s)
- Colleen Isabelle
- Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Amy Boles
- Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Nitin Chakravarti
- Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Pierluigi Porcu
- Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Jonathan Brammer
- Division of Hematology, The Ohio State University, Columbus, OH, United States
| | - Anjali Mishra
- Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Philadelphia, PA, United States
- *Correspondence: Anjali Mishra,
| |
Collapse
|
7
|
Kenda M, Avsec D, Zore T, Kogovšek E, Pečar Fonović U, Kos J, Bozovičar K, Bratkovič T, Karas Kuželički N, Žegura B, Filipič M, Sollner Dolenc M. Effects of tyrosine kinase inhibitors on androgen, estrogen α, glucocorticoid and thyroid receptors. Toxicol Appl Pharmacol 2022; 434:115818. [PMID: 34890638 DOI: 10.1016/j.taap.2021.115818] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/24/2021] [Accepted: 12/02/2021] [Indexed: 11/25/2022]
Abstract
Modern anticancer therapies favor a targeted approach. Tyrosine kinase inhibitors (TKIs) are drugs that target molecular pathways involved in various types of malignancies. Although TKIs are safe and well tolerated, they remain not completely selective; e.g., endocrine-mediated adverse events have been observed with their use. In the present study, the effects of seven TKIs were determined on the activities of androgen receptor, estrogen receptor α (ERα), glucocorticoid receptor and thyroid receptor in vitro using stably transfected cell lines expressing firefly luciferase reporter gene: AR-EcoScreen, hERα-HeLa9903, MDA-kb2, and GH3.TRE-Luc cells, respectively. Antiandrogenic activity was seen for erlotinib, estrogenic activity for imatinib, antiestrogenic activity for dasatinib, erlotinib, nilotinib, regorafenib and sorafenib, glucocorticoid activity for erlotinib and ibrutinib, antiglucocorticoid activity for regorafenib and sorafenib, and antithyroid activity for ibrutinib. Additionally, synergism was seen for 1-5 μM dasatinib and 500 nM hydrocortisone combination for glucocorticoid activity in MDA-kb2 cells. The estrogenic activity of imatinib was confirmed as mediated through ERα, and interference of the TKIs with the reporter gene assays was ruled out in a cell-lysate-based firefly luciferase enzyme inhibition assay. Imatinib in combination with 4-hydroxytamoxifen showed concentration-dependent effects on the metabolic activity of ERα-expressing AN3CA, MCF-7, and SKOV3 cells, and on cell proliferation and adhesion of MCF-7 cells. These findings contribute to the understanding of the endocrine effects of TKIs, in terms of toxicity and effectiveness, and define the need to further evaluate the endocrine disrupting activities of TKIs to safeguard human and environmental health.
Collapse
Affiliation(s)
- Maša Kenda
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | - Damjan Avsec
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | - Taja Zore
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | - Eva Kogovšek
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia
| | - Urša Pečar Fonović
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | - Janko Kos
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | - Krištof Bozovičar
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | - Tomaž Bratkovič
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | | | - Bojana Žegura
- National Institute of Biology, Department of Genetic Toxicology and Cancer Biology, Večna pot 111, SI-1000 Ljubljana, Slovenia.
| | - Metka Filipič
- National Institute of Biology, Department of Genetic Toxicology and Cancer Biology, Večna pot 111, SI-1000 Ljubljana, Slovenia.
| | - Marija Sollner Dolenc
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
8
|
Gage MC, Thippeswamy T. Inhibitors of Src Family Kinases, Inducible Nitric Oxide Synthase, and NADPH Oxidase as Potential CNS Drug Targets for Neurological Diseases. CNS Drugs 2021; 35:1-20. [PMID: 33515429 PMCID: PMC7893831 DOI: 10.1007/s40263-020-00787-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 12/21/2022]
Abstract
Neurological diseases share common neuroinflammatory and oxidative stress pathways. Both phenotypic and molecular changes in microglia, astrocytes, and neurons contribute to the progression of disease and present potential targets for disease modification. Src family kinases (SFKs) are present in both neurons and glial cells and are upregulated following neurological insults in both human and animal models. In neurons, SFKs interact with post-synaptic protein domains to mediate hyperexcitability and neurotoxicity. SFKs are upstream of signaling cascades that lead to the modulation of neurotransmitter receptors and the transcription of pro-inflammatory cytokines as well as producers of free radicals through the activation of glia. Inducible nitric oxide synthase (iNOS/NOS-II) and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2), the major mediators of reactive nitrogen/oxygen species (RNS/ROS) production in the brain, are also upregulated along with the pro-inflammatory cytokines following neurological insult and contribute to disease progression. Persistent neuronal hyperexcitability, RNS/ROS, and cytokines can exacerbate neurodegeneration, a common pathognomonic feature of the most prevalent neurological disorders such as Alzheimer's disease, Parkinson's disease, and epilepsy. Using a wide variety of preclinical disease models, inhibitors of the SFK-iNOS-NOX2 signaling axis have been tested to cure or modify disease progression. In this review, we discuss the SFK-iNOS-NOX2 signaling pathway and their inhibitors as potential CNS targets for major neurological diseases.
Collapse
Affiliation(s)
- Meghan C. Gage
- Iowa State University, Ames, Iowa, United States of America
| | | |
Collapse
|
9
|
Kumari S, Carmona AV, Tiwari AK, Trippier PC. Amide Bond Bioisosteres: Strategies, Synthesis, and Successes. J Med Chem 2020; 63:12290-12358. [PMID: 32686940 DOI: 10.1021/acs.jmedchem.0c00530] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The amide functional group plays a key role in the composition of biomolecules, including many clinically approved drugs. Bioisosterism is widely employed in the rational modification of lead compounds, being used to increase potency, enhance selectivity, improve pharmacokinetic properties, eliminate toxicity, and acquire novel chemical space to secure intellectual property. The introduction of a bioisostere leads to structural changes in molecular size, shape, electronic distribution, polarity, pKa, dipole or polarizability, which can be either favorable or detrimental to biological activity. This approach has opened up new avenues in drug design and development resulting in more efficient drug candidates introduced onto the market as well as in the clinical pipeline. Herein, we review the strategic decisions in selecting an amide bioisostere (the why), synthetic routes to each (the how), and success stories of each bioisostere (the implementation) to provide a comprehensive overview of this important toolbox for medicinal chemists.
Collapse
Affiliation(s)
- Shikha Kumari
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Angelica V Carmona
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, Ohio 43614, United States
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States.,Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States.,UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
10
|
Advances in pathogenic mechanisms and management of radiation-induced fibrosis. Biomed Pharmacother 2020; 121:109560. [DOI: 10.1016/j.biopha.2019.109560] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 10/04/2019] [Accepted: 10/17/2019] [Indexed: 12/12/2022] Open
|
11
|
Kumar M, Kulshrestha R, Singh N, Jaggi AS. Expanding spectrum of anticancer drug, imatinib, in the disorders affecting brain and spinal cord. Pharmacol Res 2019; 143:86-96. [PMID: 30902661 DOI: 10.1016/j.phrs.2019.03.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/07/2019] [Accepted: 03/17/2019] [Indexed: 02/07/2023]
Abstract
Imatinib is a tyrosine kinase inhibitor and is used as a first line drug in the treatment of Philadelphia-chromosome-positive chronic myeloid leukaemia and gastrointestinal stromal tumors. Being tyrosine kinase inhibitor, imatinib modulates the activities of Abelson gene (c-Abl), Abelson related gene (ARG), platelet-derived growth factor receptor (PDGFR), FMS-like tyrosine kinase 3 (FLT3), lymphocyte-specific protein (Lck), mitogen activated protein kinase (MAPK), amyloid precursor protein intracellular domain (AICD), α-synuclein and the stem-cell factor receptor (c-kit). Studies have shown the role of imatinib in modulating the pathophysiological state of a number of disorders affecting brain and spinal cord such as Alzheimer's disease, Parkinson's disease, stroke, multiple sclerosis and spinal cord injury. The present review discusses the role of imatinib in the above described disorders and the possible mechanisms involved in these diseases.
Collapse
Affiliation(s)
- Manish Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | | | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India.
| |
Collapse
|
12
|
Aiello FB, Guszczynski T, Li W, Hixon JA, Jiang Q, Hodge DL, Massignan T, Di Lisio C, Merchant A, Procopio AD, Bonetto V, Durum SK. IL-7-induced phosphorylation of the adaptor Crk-like and other targets. Cell Signal 2018; 47:131-141. [PMID: 29581031 DOI: 10.1016/j.cellsig.2018.03.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 03/20/2018] [Indexed: 12/16/2022]
Abstract
IL-7 is required for T cell differentiation and mature T cell homeostasis and promotes pro-B cell proliferation and survival. Tyrosine phosphorylation plays a central role in IL-7 signaling. We identified by two-dimensional electrophoresis followed by anti-phosphotyrosine immunoblotting and mass spectrometry sixteen tyrosine phosphorylated proteins from the IL-7-dependent cell line D1. IL-7 stimulation induced the phosphorylation of the proteins STI1, ATIC and hnRNPH, involved in pathways related to survival, proliferation and gene expression, respectively, and increased the phosphorylation of CrkL, a member of a family of adaptors including the highly homologous Crk isoforms CrkII and CrkI, important in multiple signaling pathways. We observed an increased phosphorylation of CrkL in murine pro-B cells and in murine and human T cells. In addition, IL-7 increased the association of CrkL with the transcription factor Stat5, essential for IL-7 pro-survival activity. The selective tyrosine kinase inhibitor Imatinib. counteracted the IL-7 pro-survival effect in D1 cells and decreased CrkL phosphorylation. These data suggested that CrkL could play a pro-survival role in IL-7-mediated signaling. We observed that pro-B cells also expressed, in addition to CrkL, the Crk isoforms CrkII and CrkI and therefore utilized pro-B cells conditionally deficient in all three to evaluate the role of these proteins. The observation that the IL-7 pro-survival effect was reduced in Crk/CrkL conditionally-deficient pro-B cells further pointed to a pro-survival role of these adaptors. To further evaluate the role of these proteins, gene expression studies were performed in Crk/CrkL conditionally-deficient pro-B cells. IL-7 decreased the transcription of the receptor LAIR1, which inhibits B cell proliferation, in a Crk/CrkL-dependent manner, suggesting that the Crk family of proteins may promote pro-B cell proliferation. Our data contribute to the understanding of IL-7 signaling and suggest the involvement of Crk family proteins in pathways promoting survival and proliferation.
Collapse
Affiliation(s)
- Francesca B Aiello
- Cancer and Inflammation Program, CCR, NCI, NIH, Bldg 560, Frederick, MD 21702, USA.
| | - Tad Guszczynski
- Molecular Targets Laboratory, FCRDC, Bldg 560, Frederick, MD 21702, USA.
| | - Wenqing Li
- Cancer and Inflammation Program, CCR, NCI, NIH, Bldg 560, Frederick, MD 21702, USA.
| | - Julie A Hixon
- Cancer and Inflammation Program, CCR, NCI, NIH, Bldg 560, Frederick, MD 21702, USA.
| | - Qiong Jiang
- Cancer and Inflammation Program, CCR, NCI, NIH, Bldg 560, Frederick, MD 21702, USA.
| | - Deborah L Hodge
- Laboratory of Experimental Medicine, FCRDC, Bldg 560, Frederick, MD 21702, USA.
| | - Tania Massignan
- Dulbecco Telethon Institute, IRCCS-Istituto di Ricerche Farmacologiche M. Negri, via La Masa 19, 20156 Milano, Italy
| | - Chiara Di Lisio
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, via dei Vestini, 66013 Chieti, Italy.
| | - Anand Merchant
- Center for Cancer Research, NIH, Bethesda, MD 20892, USA.
| | - Antonio D Procopio
- Department of Clinical and Medical Sciences, Marche Polytechnic University, via Tronto 10, 60100 Ancona, Italy.
| | - Valentina Bonetto
- Dulbecco Telethon Institute, IRCCS-Istituto di Ricerche Farmacologiche M. Negri, via La Masa 19, 20156 Milano, Italy.
| | - Scott K Durum
- Cancer and Inflammation Program, CCR, NCI, NIH, Bldg 560, Frederick, MD 21702, USA.
| |
Collapse
|
13
|
Wang X, Bleher R, Wang L, Garcia JGN, Dudek SM, Shekhawat GS, Dravid VP. Imatinib Alters Agonists-mediated Cytoskeletal Biomechanics in Lung Endothelium. Sci Rep 2017; 7:14152. [PMID: 29075042 PMCID: PMC5658337 DOI: 10.1038/s41598-017-14722-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 10/16/2017] [Indexed: 01/11/2023] Open
Abstract
The endothelium serves as a size-selective barrier and tightly controls the fluid exchange from the circulation to the surrounding tissues. In this study, a multiplexed microscopy characterization is developed to study the spatio-temporal effects of Abl kinases on endothelial cytoskeletal structure using AFM, SEM, and immunofluorescence. Sphingosine 1-phosphate (S1P) produces significant endothelial barrier enhancement by means of peripheral actin rearrangement. However, Abl kinase inhibition by imatinib reduces rapid redistribution of the important cytoskeletal proteins to the periphery and their association with the cortical actin ring. Herein, it moderates the thickness of the cortical actin ring, and diminishes the increase in elastic modulus at the periphery and cytoplasm. These findings demonstrate that imatinib attenuates multiple cytoskeletal changes associated with S1P-mediated endothelial barrier enhancement and suggest a novel role for Abl kinases in mediating these S1P effects. These observations bridge the gap between molecule dynamics, structure complexity and function connectivity across varied length-scales to improve our understanding on human pulmonary endothelial barrier regulation. Moreover, our study suggests a framework for understanding form-function relationships in other biomechanical subsystems, wherein complex hierarchical organization programmed from the molecular scale to the cellular and tissue levels has an intimate relationship to the overall physiological function.
Collapse
Affiliation(s)
- X Wang
- Tianjin Key Laboratory of the Design and Intelligent Control of the Advanced Mechatronical System, Tianjin University of Technology, Tianjin, China, 300384.,National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, China, 300384.,Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA, 60208
| | - R Bleher
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA, 60208
| | - L Wang
- Department of Medicine, University of Illinois, Chicago, IL, USA, 60612
| | - J G N Garcia
- Department of Medicine, University of Arizona, Tucson, AZ, USA, 85721
| | - S M Dudek
- Department of Medicine, University of Illinois, Chicago, IL, USA, 60612.
| | - G S Shekhawat
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA, 60208.
| | - V P Dravid
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA, 60208.
| |
Collapse
|
14
|
Koseła-Paterczyk H, Rutkowski P. Dermatofibrosarcoma protuberans and gastrointestinal stromal tumor as models for targeted therapy in soft tissue sarcomas. Expert Rev Anticancer Ther 2017; 17:1107-1116. [DOI: 10.1080/14737140.2017.1390431] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Hanna Koseła-Paterczyk
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute Oncology Center, Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute Oncology Center, Warsaw, Poland
| |
Collapse
|
15
|
Impact of germline and somatic missense variations on drug binding sites. THE PHARMACOGENOMICS JOURNAL 2016; 17:128-136. [PMID: 26810135 PMCID: PMC5380835 DOI: 10.1038/tpj.2015.97] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 11/02/2015] [Accepted: 11/13/2015] [Indexed: 11/10/2022]
Abstract
Advancements in next-generation sequencing (NGS) technologies are generating a vast amount of data. This exacerbates the current challenge of translating NGS data into actionable clinical interpretations. We have comprehensively combined germline and somatic nonsynonymous single-nucleotide variations (nsSNVs) that affect drug binding sites in order to investigate their prevalence. The integrated data thus generated in conjunction with exome or whole-genome sequencing can be used to identify patients who may not respond to a specific drug because of alterations in drug binding efficacy due to nsSNVs in the target protein's gene. To identify the nsSNVs that may affect drug binding, protein–drug complex structures were retrieved from Protein Data Bank (PDB) followed by identification of amino acids in the protein–drug binding sites using an occluded surface method. Then, the germline and somatic mutations were mapped to these amino acids to identify which of these alter protein–drug binding sites. Using this method we identified 12 993 amino acid–drug binding sites across 253 unique proteins bound to 235 unique drugs. The integration of amino acid–drug binding sites data with both germline and somatic nsSNVs data sets revealed 3133 nsSNVs affecting amino acid–drug binding sites. In addition, a comprehensive drug target discovery was conducted based on protein structure similarity and conservation of amino acid–drug binding sites. Using this method, 81 paralogs were identified that could serve as alternative drug targets. In addition, non-human mammalian proteins bound to drugs were used to identify 142 homologs in humans that can potentially bind to drugs. In the current protein–drug pairs that contain somatic mutations within their binding site, we identified 85 proteins with significant differential gene expression changes associated with specific cancer types. Information on protein–drug binding predicted drug target proteins and prevalence of both somatic and germline nsSNVs that disrupt these binding sites can provide valuable knowledge for personalized medicine treatment. A web portal is available where nsSNVs from individual patient can be checked by scanning against DrugVar to determine whether any of the SNVs affect the binding of any drug in the database.
Collapse
|
16
|
Dadrich M, Nicolay NH, Flechsig P, Bickelhaupt S, Hoeltgen L, Roeder F, Hauser K, Tietz A, Jenne J, Lopez R, Roehrich M, Wirkner U, Lahn M, Huber PE. Combined inhibition of TGFβ and PDGF signaling attenuates radiation-induced pulmonary fibrosis. Oncoimmunology 2015; 5:e1123366. [PMID: 27467922 PMCID: PMC4910723 DOI: 10.1080/2162402x.2015.1123366] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 11/11/2015] [Accepted: 11/18/2015] [Indexed: 11/25/2022] Open
Abstract
Background: Radiotherapy (RT) is a mainstay for the treatment of lung cancer, but the effective dose is often limited by the development of radiation-induced pneumonitis and pulmonary fibrosis. Transforming growth factor β (TGFβ) and platelet-derived growth factor (PDGF) play crucial roles in the development of these diseases, but the effects of dual growth factor inhibition on pulmonary fibrosis development remain unclear. Methods: C57BL/6 mice were treated with 20 Gy to the thorax to induce pulmonary fibrosis. PDGF receptor inhibitors SU9518 and SU14816 (imatinib) and TGFβ receptor inhibitor galunisertib were applied individually or in combinations after RT. Lung density and septal fibrosis were measured by high-resolution CT and MRI. Lung histology and gene expression analyses were performed and Osteopontin levels were studied. Results: Treatment with SU9518, SU14816 or galunisertib individually attenuated radiation-induced pulmonary inflammation and fibrosis and decreased radiological and histological signs of lung damage. Combining PDGF and TGFβ inhibitors showed to be feasible and safe in a mouse model, and dual inhibition significantly attenuated radiation-induced lung damage and extended mouse survival compared to blockage of either pathway alone. Gene expression analysis of irradiated lung tissue showed upregulation of PDGF and TGFβ-dependent signaling components by thoracic irradiation, and upregulation patterns show crosstalk between downstream mediators of the PDGF and TGFβ pathways. Conclusion: Combined small-molecule inhibition of PDGF and TGFβ signaling is a safe and effective treatment for radiation-induced pulmonary inflammation and fibrosis in mice and may offer a novel approach for treatment of fibrotic lung diseases in humans. Translational statement: RT is an effective treatment modality for cancer with limitations due to acute and chronic toxicities, where TGFβ and PDGF play a key role. Here, we show that a combined inhibition of TGFβ and PDGF signaling is more effective in attenuating radiation-induced lung damage compared to blocking either pathway alone. We used the TGFβ-receptor I inhibitor galunisertib, an effective anticancer compound in preclinical models and the PDGFR inhibitors imatinib and SU9518, a sunitinib analog. Our signaling data suggest that the reduction of TGFβ and PDGF signaling and the attenuation of SPP1 (Osteopontin) expression may be responsible for the observed benefits. With the clinical availability of similar compounds currently in phase-I/II trials as cancer therapeutics or already approved for certain cancers or idiopathic lung fibrosis (IPF), our study suggests that the combined application of small molecule inhibitors of TGFβ and PDGF signaling may offer a promising approach to treat radiation-associated toxicity in RT of lung cancer.
Collapse
Affiliation(s)
- Monika Dadrich
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF, Heidelberg, Germany; Department of Radiology, University Hospital Center, 400 INF, Heidelberg, Germany
| | - Nils H Nicolay
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF, Heidelberg, Germany; Department of Radiation Oncology, University Hospital Center, 400 INF, Heidelberg, Germany
| | - Paul Flechsig
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF, Heidelberg, Germany; Department of Radiology, University Hospital Center, 400 INF, Heidelberg, Germany
| | - Sebastian Bickelhaupt
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Line Hoeltgen
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Falk Roeder
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF, Heidelberg, Germany; Department of Radiation Oncology, University Hospital Center, 400 INF, Heidelberg, Germany; Department of Radiation Oncology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Kai Hauser
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Alexandra Tietz
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Jürgen Jenne
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Ramon Lopez
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Manuel Roehrich
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Ute Wirkner
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Michael Lahn
- Oncology Early Clinical Investigation, Lilly Research Laboratories, Indianapolis , IN, USA
| | - Peter E Huber
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF, Heidelberg, Germany; Department of Radiation Oncology, University Hospital Center, 400 INF, Heidelberg, Germany
| |
Collapse
|
17
|
Brümmendorf TH, Cortes JE, Khoury HJ, Kantarjian HM, Kim DW, Schafhausen P, Conlan MG, Shapiro M, Turnbull K, Leip E, Gambacorti-Passerini C, Lipton JH. Factors influencing long-term efficacy and tolerability of bosutinib in chronic phase chronic myeloid leukaemia resistant or intolerant to imatinib. Br J Haematol 2015; 172:97-110. [PMID: 26537529 PMCID: PMC4737299 DOI: 10.1111/bjh.13801] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 08/26/2015] [Accepted: 08/31/2015] [Indexed: 11/28/2022]
Abstract
The dual SRC/ABL1 tyrosine kinase inhibitor bosutinib is indicated for adults with Ph+ chronic myeloid leukaemia (CML) resistant/intolerant to prior therapy. This analysis of an ongoing phase 1/2 study (NCT00261846) assessed effects of baseline patient characteristics on long‐term efficacy and safety of bosutinib 500 mg/day in adults with imatinib (IM)‐resistant (IM‐R; n = 196)/IM‐intolerant (IM‐I; n = 90) chronic phase (CP) CML. Median treatment duration was 24·8 months (median follow‐up, 43·6 months). Cumulative major cytogenetic response (MCyR) rate [95% confidence interval (CI)], was 59% (53–65%); Kaplan‐Meier (KM) probability of maintaining MCyR at 4 years was 75% (66–81%). Cumulative incidence of on‐treatment progression/death at 4 years was 19% (95% CI, 15–24%); KM 2‐year overall survival was 91% (87–94%). Significant baseline predictors of both MCyR and complete cytogenetic response (newly attained/maintained from baseline) at 3 and 6 months included prior IM cytogenetic response, baseline MCyR, prior interferon therapy and <6 months duration from diagnosis to IM treatment initiation and no interferon treatment before IM. The most common adverse event (AE) was diarrhoea (86%). Baseline bosutinib‐sensitive BCR‐ABL1 mutation was the only significant predictor of grade 3/4 diarrhoea; no significant predictors were identified for liver‐related AEs. Bosutinib demonstrates durable efficacy and manageable toxicity in IM‐R/IM‐I CP‐CML patients.
Collapse
Affiliation(s)
- Tim H Brümmendorf
- Universitätsklinikum RWTH Aachen, Aachen, Germany.,Department of Internal Medicine II, Hubertus Wald Tumorzentrum University Cancer Centre Hamburg, Hamburg, Germany
| | - Jorge E Cortes
- University of Texas MD, Anderson Cancer Center, Houston, TX, USA
| | - Hanna J Khoury
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | | | | | - Philippe Schafhausen
- Department of Internal Medicine II, Hubertus Wald Tumorzentrum University Cancer Centre Hamburg, Hamburg, Germany
| | | | | | | | | | | | - Jeff H Lipton
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
18
|
Motawi TMK, Sadik NAH, Fahim SA, Shouman SA. Combination of imatinib and clotrimazole enhances cell growth inhibition in T47D breast cancer cells. Chem Biol Interact 2015; 233:147-56. [PMID: 25863232 DOI: 10.1016/j.cbi.2015.03.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 03/13/2015] [Accepted: 03/25/2015] [Indexed: 01/19/2023]
Abstract
Imatinib mesylate (IM), a tyrosine kinase inhibitor, is used as targeted cancer therapy. However, mono-targeting by IM does not always achieve full tumor eradication and thus it is recommended to combine IM with other anticancer agents. Clotrimazole (CLT) is an antifungal azole derivative with promising anticancer effects due to inhibiting the activity of glycolytic enzymes. The present study aimed to evaluate the effect of combining CLT with IM on breast cancer cell line in an attempt to establish effective new combination. T47D human breast cancer cell line was treated with different concentrations of IM and/or CLT for 48 h. IM-CLT interaction was determined by isobologram equation and combination index. Cell viability was confirmed by measuring LDH activity. As indicators of glycolysis inhibition, the expression of hexokinase-2 (HK-2) and 6-phosphofructo-1-kinase (PFK-1) plus the activity of intracellular lactate dehydrogenase (LDH) and pyruvate kinase (PK) were determined. In addition, glucose consumption and adenosine triphosphate (ATP) production were measured. Moreover, nitric oxide (NO), vascular endothelial growth factor (VEGF) and hypoxia inducible factor-α (HIF-α) were also determined as they are modulators for glycolysis. This study demonstrated that IM or CLT synergistically inhibited cell growth in T47D as shown by combination and dose reduction indices. The combination of 15 μM IM and 20 μM CLT significantly decreased glucose consumption, activity of both PK and intracellular LDH, while increased leaked LDH, VEGF and NO in the medium compared to each drug alone. Furthermore the combination decreased gene expression of HK-2, PFK-1 and ATP content compared to the control. In conclusion, the synergistic effect of CLT on IM cytotoxicity in T47D cell line maybe mediated through inhibition of glycolysis and increasing both NO and VEGF. Further studies are required to confirm the efficiency and safety of this combination.
Collapse
Affiliation(s)
- Tarek M K Motawi
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nermin A H Sadik
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Sally A Fahim
- Biochemistry Department, Faculty of Pharmacy, Ahram Canadian University, Cairo, Egypt
| | - Samia A Shouman
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt.
| |
Collapse
|
19
|
Ollero M, Sahali D. Inhibition of the VEGF signalling pathway and glomerular disorders. Nephrol Dial Transplant 2014; 30:1449-55. [PMID: 25480873 DOI: 10.1093/ndt/gfu368] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/27/2014] [Indexed: 11/14/2022] Open
Abstract
Anti-cancer therapeutic approaches targeting the vascular endothelial growth factor (VEGF) ligand (anti-VEGF) or inhibiting its receptors (RTKI) have recently been developed. In spite of the promising results achieved, a serious drawback and dose-limiting side effect is the development, among others, of renal complications. This encompasses two glomerular pathological entities, namely minimal change/focal segmental glomerulosclerosis and thrombotic micro-angiopathy, involving two distinct cell types, podocytes and endothelial cells, respectively. The mechanisms that link anti-cancer therapy by RTKI to podocyte dysfunction and nephrotic level proteinuria are still poorly understood. Nevertheless, recent findings strongly suggest a central role of RelA, the master subunit of NF-κB and c-mip, an active player in podocyte disorders. RelA, which is up-regulated following anti-VEGF therapy, is inactivated by RTKI, leading to c-mip over-expression in the podocyte. This results in severe alterations in the architecture of podocyte actin cytoskeleton and subsequent severe proteinuria. Hence, clarifying the mechanisms linking c-mip and RelA as key pathogenic factors represents a critical goal in the understanding of different glomerulopathies. In the context of VEGF-targeted anti-cancer therapy, the study of these mechanisms along with the molecular cross-talk between podocyte and endothelial cell constitutes the basis for the emerging field of onconephrology.
Collapse
Affiliation(s)
- Mario Ollero
- INSERM, U955, Equipe 21, Créteil, France Université Paris-Est Créteil Val-de-Marne, Créteil, France
| | - Djillali Sahali
- INSERM, U955, Equipe 21, Créteil, France Université Paris-Est Créteil Val-de-Marne, Créteil, France AP-HP, Groupe Hospitalier Henri Mondor-Albert Chenevier, Service de Néphrologie, Créteil, France
| |
Collapse
|
20
|
Beckmann S, Long T, Scheld C, Geyer R, Caffrey CR, Grevelding CG. Serum albumin and α-1 acid glycoprotein impede the killing of Schistosoma mansoni by the tyrosine kinase inhibitor Imatinib. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2014; 4:287-95. [PMID: 25516839 PMCID: PMC4266805 DOI: 10.1016/j.ijpddr.2014.07.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The Abl tyrosine-kinase inhibitor Imatinib is toxic to S. mansoni in vitro but not in vivo in rodents. Blood components like serum albumin and alpha-1 acid glycoprotein (AGP) negated Imatinib’s toxicity in vitro. Erythromycin partially restored the toxicity of Imatinib in vitro. High levels of AGP upon infection make rodents poor models for examining some small molecule inhibitors.
In the search for new drugs and drug targets to treat the flatworm disease schistosomiasis, protein kinases (PKs) have come under particular scrutiny because of their essential roles in developmental and physiological processes in schistosome parasites. In this context the application of the anti-cancer Abl tyrosine kinase (TK) inhibitor Imatinib (Gleevec/Glivec; STI-571) to adult Schistosoma mansoni in vitro has indicated negative effects on diverse physiological processes including survival. Motivated by these in vitro findings, we performed in vivo experiments in rodent models of S. mansoni infection. Unexpectedly, Imatinib had no effect on worm burden or egg-production. We found that the blood components serum albumin (SA) and alpha-1 acid glycoprotein (AGP or orosomucoid) negated Imatinib’s deleterious effects on adult S. mansoni and schistosomula (post-infective larvae) in vitro. This negative effect was partially reversed by erythromycin. AGP synthesis can increase as a consequence of inflammatory processes or infection; in addition upon infection AGP levels are 6–8 times higher in mice compared to humans. Therefore, mice and probably other rodents are poor infection models for measuring the effects of Imatinib in vivo. Accordingly, we suggest the routine evaluation of the ability of AGP and SA to block in vitro anti-schistosomal effects of small molecules like Imatinib prior to laborious and expensive animal experiments.
Collapse
Affiliation(s)
- Svenja Beckmann
- BFS, Institute for Parasitology, Justus-Liebig-University, Giessen, Germany
| | - Thavy Long
- Center for Discovery and Innovation in Parasitic Diseases and the Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Christina Scheld
- BFS, Institute for Parasitology, Justus-Liebig-University, Giessen, Germany
| | - Rudolf Geyer
- Institute for Biochemistry, Justus-Liebig-University, Giessen, Germany
| | - Conor R Caffrey
- Center for Discovery and Innovation in Parasitic Diseases and the Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | | |
Collapse
|
21
|
Canonical and new generation anticancer drugs also target energy metabolism. Arch Toxicol 2014; 88:1327-50. [PMID: 24792321 DOI: 10.1007/s00204-014-1246-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 04/15/2014] [Indexed: 01/05/2023]
Abstract
Significant efforts have been made for the development of new anticancer drugs (protein kinase or proteasome inhibitors, monoclonal humanized antibodies) with presumably low or negligible side effects and high specificity. However, an in-depth analysis of the side effects of several currently used canonical (platin-based drugs, taxanes, anthracyclines, etoposides, antimetabolites) and new generation anticancer drugs as the first line of clinical treatment reveals significant perturbation of glycolysis and oxidative phosphorylation. Canonical and new generation drug side effects include decreased (1) intracellular ATP levels, (2) glycolytic/mitochondrial enzyme/transporter activities and/or (3) mitochondrial electrical membrane potentials. Furthermore, the anti-proliferative effects of these drugs are markedly attenuated in tumor rho (0) cells, in which functional mitochondria are absent; in addition, several anticancer drugs directly interact with isolated mitochondria affecting their functions. Therefore, several anticancer drugs also target the energy metabolism, and hence, the documented inhibitory effect of anticancer drugs on cancer growth should also be linked to the blocking of ATP supply pathways. These often overlooked effects of canonical and new generation anticancer drugs emphasize the role of energy metabolism in maintaining cancer cells viable and its targeting as a complementary and successful strategy for cancer treatment.
Collapse
|
22
|
DrugPath: a database for academic investigators to match oncology molecular targets with drugs in development. Cancer Chemother Pharmacol 2014; 73:1089-93. [DOI: 10.1007/s00280-014-2433-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 02/27/2014] [Indexed: 10/25/2022]
|
23
|
Response to imatinib as a function of target kinase expression in recurrent glioblastoma. SPRINGERPLUS 2014; 3:111. [PMID: 25674429 PMCID: PMC4320134 DOI: 10.1186/2193-1801-3-111] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 02/03/2014] [Indexed: 12/30/2022]
Abstract
BACKGROUND Despite some progress in the treatment of glioblastoma, most patients experience tumor recurrence. Imatinib mesylate, a tyrosine kinase inhibitor of platelet derived growth factor receptor-alpha and -beta, c-fms, c-kit, abl and arg kinase (imatinib targets), has been shown to prevent tumor progression in early studies of recurrent gliomas, but has shown weak activity in randomized controlled trials. We studied the response to oral imatinib in 24 patients with recurrent glioblastoma who showed immunohistochemical expression of these imatinib targets in the initially resected tumor tissue. METHODS We offered oral imatinib, 400 mg once daily treatment to 24 recurrent glioblastoma patients whose initial biopsy showed presence of at least one imatinib inhibitable tyrosine kinase. RESULTS Six imatinib treated patients survived over one year. Twelve patients achieved at least tumor stabilisations from 2.6 months to 13.4 months. Median progression free survival was 3 months and median overall survival was 6 months. Imatinib was well tolerated. We found evidence, though not statistically significant, that arg kinase [Abl-2] immunopositivity had shorter survival [5 months] than the arg kinase immunonegative group [9 months]. CONCLUSIONS Responses to imatinib observed in this patient series where imatinib inhibitable tyrosine kinases were documented on the original biopsy are marginally better than that previously reported in imatinib treatment of unselected recurrent glioblastoma patients. We thus present a suggestion for defining a patient sub-population who might potentially benefit from imatinib.
Collapse
|
24
|
Blair BG, Bardelli A, Park BH. Somatic alterations as the basis for resistance to targeted therapies. J Pathol 2014; 232:244-54. [PMID: 24114654 DOI: 10.1002/path.4278] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 08/23/2013] [Accepted: 09/21/2013] [Indexed: 12/20/2022]
Abstract
Recent advances in genetics and genomics have revealed new genes and pathways that are somatically altered in human malignancies. This wealth of knowledge has translated into molecularly defined targets for therapy over the past two decades, serving as key examples that translation of laboratory findings can have great impact on the ability to treat patients with cancer. However, given the genetic instability and heterogeneity that are characteristic of all human cancers, drug resistance to virtually all therapies has emerged, posing further and future challenges for clinical oncology. Here we review the history of targeted therapies, including examples of genetically defined cancer targets and their approved therapies. We also discuss resistance mechanisms that have been uncovered, with an emphasis on somatic genetic alterations that lead to these phenotypes.
Collapse
Affiliation(s)
- Brian G Blair
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | | | | |
Collapse
|
25
|
Kunz M. Oncogenes in melanoma: an update. Eur J Cell Biol 2013; 93:1-10. [PMID: 24468268 DOI: 10.1016/j.ejcb.2013.12.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/16/2013] [Accepted: 12/17/2013] [Indexed: 12/13/2022] Open
Abstract
Melanoma is a highly aggressive tumour with poor prognosis in the metastatic stage. BRAF, NRAS, and KIT are three well-known oncogenes involved in melanoma pathogenesis. Targeting of mutated BRAF kinase has recently been shown to significantly improve overall survival of metastatic melanoma patients, underscoring the particular role of this oncogene in melanoma biology. However, recurrences regularly occur within several months, which supposedly involve further oncogenes. Moreover, oncogenic driver mutations have not been described for up to 30% of all melanomas. In order to obtain a more complete picture of the mutational landscape of melanoma, more recent studies used high-throughput DNA sequencing technologies. A number of new oncogene candidates such as MAPK1/2, ERBB4, GRIN2A, GRM3, RAC1, and PREX2 were identified. Their particular role in melanoma biology is currently under investigation. Evidence for the functional relevance of some of these new oncogene candidates has been provided in in vitro and in vivo experiments. However, these findings await further validation in clinical studies. This review provides an overview on well-known melanoma oncogenes and new oncogene candidates, based on recent high-throughput sequencing studies. The list of genes discussed herein is of course not complete but highlights some of the most significant of recent findings in this area. The new candidates may support more individualized treatment approaches for metastatic melanoma patients in the future.
Collapse
Affiliation(s)
- Manfred Kunz
- Department of Dermatology, Venereology and Allergology, University of Leipzig, 04103 Leipzig, Germany.
| |
Collapse
|
26
|
Affiliation(s)
- Mark R Wick
- Departments of Pathology, University of Virginia Health System, Charlottesville, VA.
| | | | | |
Collapse
|
27
|
Wang-Rosenke Y, Khadzhynov D, Loof T, Mika A, Kawachi H, Neumayer HH, Peters H. Tyrosine kinases inhibition by Imatinib slows progression in chronic anti-thy1 glomerulosclerosis of the rat. BMC Nephrol 2013; 14:223. [PMID: 24119229 PMCID: PMC3816310 DOI: 10.1186/1471-2369-14-223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 09/25/2013] [Indexed: 12/20/2022] Open
Abstract
Background Chronic progressive mesangioproliferative nephropathy represents a major cause of end-stage renal disease worldwide. Until now, effective approaches to stop or even slow its progression are limited. We tested the effects of an inhibitor of PDGF receptor, abl and c-kit tyrosine kinases, Imatinib, in a chronic progressive model of mesangioproliferative glomerulosclerosis. Methods Anti-thy1 glomerulosclerosis was induced by injection of anti-thy1 antibody into uninephrectomized Wistar rats. One week after disease induction, according to the degree of proteinuria, animals were stratified and assigned to chronic glomerulosclerosis (cGS) and cGS plus Imatinib (10 mg/kg body weight/day). In week 20, renoprotective actions of Imatinib were analyzed by a set of functional, histological and molecular biological parameters. Results Untreated cGS rats showed elevation of systolic blood pressure and marked progression in proteinuria, renal fibrosis, cell infiltration, cell proliferation and function lost. Administration of Imatinib went along significantly with lower systolic blood pressure (−10 mmHg) and proteinuria (−33%). Imatinib administration was paralled by significant reductions in tubulointerstitial accumulation of matrix proteins (−44%), collagen I deposition (−86%), expression of TGF-beta1 (−30%), production of fibronectin (−23%), myofibroblast differentiation (−87%), macrophage infiltration (−36%) and cell proliferation (−45%), respectively. In comparison with untreated cGS animals, Imatinib therapy lowered also blood creatinine (−41%) and blood urea concentrations (−36%) and improved creatinine clearance (+25%). Glomerular fibrotic changes were lowered moderately by Imatinib. Conclusions Therapy with Imatinib limits the progressive course of chronic anti-thy1 glomerulosclerosis towards tubulointerstitial fibrosis and renal insufficiency. This was paralleled by direct and indirect sign of TGF-β1 and PDGF inhibition. The findings suggest that the pharmacological principal of inhibition of tyrosine kinases with drugs such as Imatinib might serve as approach for limiting progression of human mesangioproliferative glomerulosclerosis.
Collapse
Affiliation(s)
- Yingrui Wang-Rosenke
- Department of Nephrology and Center of Cardiovascular Research, Campus Charité Mitte, Charité University Medicine Berlin, Charitéplatz 1, Berlin D-10117, Germany.
| | | | | | | | | | | | | |
Collapse
|
28
|
Izzedine H, Mangier M, Ory V, Zhang SY, Sendeyo K, Bouachi K, Audard V, Péchoux C, Soria JC, Massard C, Bahleda R, Bourry E, Khayat D, Baumelou A, Lang P, Ollero M, Pawlak A, Sahali D. Expression patterns of RelA and c-mip are associated with different glomerular diseases following anti-VEGF therapy. Kidney Int 2013; 85:457-70. [PMID: 24067439 DOI: 10.1038/ki.2013.344] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 05/25/2013] [Accepted: 07/12/2013] [Indexed: 12/14/2022]
Abstract
Renal toxicity constitutes a dose-limiting side effect of anticancer therapies targeting vascular endothelial growth factor (VEGF). In order to study this further, we followed up 29 patients receiving this treatment, who experienced proteinuria, hypertension, and/or renal insufficiency. Eight developed minimal change nephropathy/focal segmental glomerulopathy (MCN/FSG)-like lesions and 13 developed thrombotic microangiopathy (TMA). Patients receiving receptor tyrosine kinase inhibitors (RTKIs) mainly developed MCN/FSG-like lesions, whereas TMA complicated anti-VEGF therapy. There were no mutations in factor H, factor I, or membrane cofactor protein of the complement alternative pathway, while plasma ADAMTS13 activity persisted and anti-ADAMTS13 antibodies were undetectable in patients with TMA. Glomerular VEGF expression was undetectable in TMA and decreased in MCN/FSG. Glomeruli from patients with TMA displayed a high abundance of RelA in endothelial cells and in the podocyte nuclei, but c-mip was not detected. Conversely, MCN/FSG-like lesions exhibited a high abundance of c-mip, whereas RelA was scarcely detected. RelA binds in vivo to the c-mip promoter and prevents its transcriptional activation, whereas RelA knockdown releases c-mip activation. The RTKI sorafenib inhibited RelA activity, which then promoted c-mip expression. Thus, our results suggest that c-mip and RelA define two distinct types of renal damage associated with VEGF-targeted therapies.
Collapse
Affiliation(s)
- Hassan Izzedine
- Department of Nephrology, Pitie-Salpetriere Hospital, Paris, France
| | - Melanie Mangier
- 1] INSERM U 955, Equipe 21, Créteil, France [2] Université Paris-Est Creteil, Créteil, France
| | - Virginie Ory
- 1] INSERM U 955, Equipe 21, Créteil, France [2] Université Paris-Est Creteil, Créteil, France
| | - Shao-Yu Zhang
- 1] INSERM U 955, Equipe 21, Créteil, France [2] Université Paris-Est Creteil, Créteil, France
| | - Kelhia Sendeyo
- 1] INSERM U 955, Equipe 21, Créteil, France [2] Université Paris-Est Creteil, Créteil, France
| | - Khedidja Bouachi
- 1] INSERM U 955, Equipe 21, Créteil, France [2] Université Paris-Est Creteil, Créteil, France [3] Service de Néphrologie, AP-HP, Groupe Hospitalier Henri Mondor-Albert Chenevier, Créteil, France
| | - Vincent Audard
- 1] INSERM U 955, Equipe 21, Créteil, France [2] Université Paris-Est Creteil, Créteil, France [3] Service de Néphrologie, AP-HP, Groupe Hospitalier Henri Mondor-Albert Chenevier, Créteil, France
| | - Christine Péchoux
- INRA, UR1196 Génomique et Physiologie de la Lactation, Plateforme MIMA2, Jouy-en-Josas, France
| | | | | | | | - Edward Bourry
- Department of Nephrology, Pitie-Salpetriere Hospital, Paris, France
| | - David Khayat
- Department of Medical Oncology, Pitie-Salpetriere Hospital, Paris, France
| | - Alain Baumelou
- Department of Nephrology, Pitie-Salpetriere Hospital, Paris, France
| | - Philippe Lang
- 1] INSERM U 955, Equipe 21, Créteil, France [2] Université Paris-Est Creteil, Créteil, France [3] Service de Néphrologie, AP-HP, Groupe Hospitalier Henri Mondor-Albert Chenevier, Créteil, France
| | - Mario Ollero
- 1] INSERM U 955, Equipe 21, Créteil, France [2] Université Paris-Est Creteil, Créteil, France
| | - Andre Pawlak
- 1] INSERM U 955, Equipe 21, Créteil, France [2] Université Paris-Est Creteil, Créteil, France
| | - Djillali Sahali
- 1] INSERM U 955, Equipe 21, Créteil, France [2] Université Paris-Est Creteil, Créteil, France [3] Service de Néphrologie, AP-HP, Groupe Hospitalier Henri Mondor-Albert Chenevier, Créteil, France
| |
Collapse
|
29
|
Gur S, Sikka SC, Abdel-Mageed AB, Abd Elmageed ZY, Rezk B, Pankey E, Kadowitz PJ, Hellstrom WJ. Imatinib Mesylate (Gleevec) Induces Human Corpus Cavernosum Relaxation by Inhibiting Receptor Tyrosine Kinases (RTKs): Identification of New RTK Targets. Urology 2013; 82:745.e11-6. [DOI: 10.1016/j.urology.2013.04.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 04/10/2013] [Accepted: 04/19/2013] [Indexed: 12/23/2022]
|
30
|
Abstract
PURPOSE OF REVIEW Anaplastic thyroid cancer is the most aggressive solid tumor known to humans. Even when found in a localized form, the prognosis is grave. For metastatic disease, there has been little effect on survival using traditional chemotherapy. RECENT FINDINGS Over a five-decade interval, there has been little progress in the treatment of this malignancy. However, targeted agents represent a new mode of treatment, and some studies have shown encouraging preclinical results. Combretastatin has shown activity in phase 1 and phase II trials; although the registration phase III study failed to meet its accrual goals, it did appear to show some benefit, especially in younger patients. SUMMARY Combinations of this compound and other targeted agents may prove to be a breakthrough in an otherwise untreatable cancer.
Collapse
|
31
|
Leblanc F, Zhang D, Liu X, Loughran TP. Large granular lymphocyte leukemia: from dysregulated pathways to therapeutic targets. Future Oncol 2013; 8:787-801. [PMID: 22830400 DOI: 10.2217/fon.12.75] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Large granular lymphocyte (LGL) leukemia is a clonal lymphoproliferative disorder of cytotoxic lymphocytes characterized by an expansion of CD3(+) cytotoxic T lymphocytes or CD3(-) natural killer cells. Patients present with various cytopenias including neutropenia, anemia and thrombocytopenia. In addition, there is an association of T-cell large granular lymphocytic leukemia with rheumatoid arthritis. It is believed that LGL leukemia begins as an antigen-driven immune response with subsequent constitutive activation of cytotoxic T lymphocytes or natural killer cells through PDGF and IL-15 contributing to their survival. Consequently, this leads to a dysregulation of apoptosis and dysfunction of the activation-induced cell death pathway. Treatment of LGL leukemia is based on a low-dose immunosuppressive regimen using methotrexate or cyclophosphamide. However, no standard of therapy has been established, as large prospective trials have not been conducted. In addition, some patients are refractory to treatment. The lack of a curative therapy for LGL leukemia means that new treatment options are needed. Insight into the various dysregulated signaling pathways in LGL leukemia may provide novel therapeutic treatment modalities.
Collapse
Affiliation(s)
- Francis Leblanc
- Penn State Hershey Cancer Institute, Experimental Therapeutics, Room 4427, 500 University Drive, PO Box 850, Hershey, PA 17033-0850, USA
| | | | | | | |
Collapse
|
32
|
McCloskey KD. Bladder interstitial cells: an updated review of current knowledge. Acta Physiol (Oxf) 2013; 207:7-15. [PMID: 23034074 DOI: 10.1111/apha.12009] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 01/22/2012] [Accepted: 09/10/2012] [Indexed: 01/12/2023]
Abstract
The field of bladder research has been energized by the study of novel interstitial cells (IC) over the last decade. Several subgroups of IC are located within the bladder wall and make structural interactions with nerves and smooth muscle, indicating integration with intercellular communication and key physiological functions. Significant progress has been made in the study of bladder ICs' cellular markers, ion channels and receptor expression, electrical and calcium signalling, yet their specific functions in normal bladder filling and emptying remain elusive. There is increasing evidence that the distribution of IC is altered in bladder pathophysiologies suggesting that changes in IC may be linked with the development of bladder dysfunction. This article summarizes the current state of the art of our knowledge of IC in normal bladder and reviews the literature on IC in dysfunctional bladder.
Collapse
Affiliation(s)
- K. D. McCloskey
- Centre for Cancer Research and Cell Biology; Queen's University Belfast; Belfast; Northern Ireland; UK
| |
Collapse
|
33
|
Wierstra I. FOXM1 (Forkhead box M1) in tumorigenesis: overexpression in human cancer, implication in tumorigenesis, oncogenic functions, tumor-suppressive properties, and target of anticancer therapy. Adv Cancer Res 2013; 119:191-419. [PMID: 23870513 DOI: 10.1016/b978-0-12-407190-2.00016-2] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
FOXM1 (Forkhead box M1) is a typical proliferation-associated transcription factor and is also intimately involved in tumorigenesis. FOXM1 stimulates cell proliferation and cell cycle progression by promoting the entry into S-phase and M-phase. Additionally, FOXM1 is required for proper execution of mitosis. In accordance with its role in stimulation of cell proliferation, FOXM1 exhibits a proliferation-specific expression pattern and its expression is regulated by proliferation and anti-proliferation signals as well as by proto-oncoproteins and tumor suppressors. Since these factors are often mutated, overexpressed, or lost in human cancer, the normal control of the foxm1 expression by them provides the basis for deregulated FOXM1 expression in tumors. Accordingly, FOXM1 is overexpressed in many types of human cancer. FOXM1 is intimately involved in tumorigenesis, because it contributes to oncogenic transformation and participates in tumor initiation, growth, and progression, including positive effects on angiogenesis, migration, invasion, epithelial-mesenchymal transition, metastasis, recruitment of tumor-associated macrophages, tumor-associated lung inflammation, self-renewal capacity of cancer cells, prevention of premature cellular senescence, and chemotherapeutic drug resistance. However, in the context of urethane-induced lung tumorigenesis, FOXM1 has an unexpected tumor suppressor role in endothelial cells because it limits pulmonary inflammation and canonical Wnt signaling in epithelial lung cells, thereby restricting carcinogenesis. Accordingly, FOXM1 plays a role in homologous recombination repair of DNA double-strand breaks and maintenance of genomic stability, that is, prevention of polyploidy and aneuploidy. The implication of FOXM1 in tumorigenesis makes it an attractive target for anticancer therapy, and several antitumor drugs have been reported to decrease FOXM1 expression.
Collapse
|
34
|
Moeller MJ, Smeets B. Novel target in the treatment of RPGN: the activated parietal cell. Nephrol Dial Transplant 2012; 28:489-92. [PMID: 23243041 DOI: 10.1093/ndt/gfs566] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Iyoda et al. have provided strong experimental evidence for beneficial effects of PDGF signalling inhibition in two seemingly unrelated glomerular diseases: rapidly progressive glomerulonephritis (RPGN) in the present study and focal and segmental glomerulosclerosis (FSGS) in a previous study. Novel insights into the pathogenesis of these two diseases have unravelled a common cellular mechanism: activation of parietal epithelial cells (PECs). In addition, recent studies have shown that PDGF signalling is sufficient to mediate the PEC activation and formation of cellular crescents, the hallmark of RPGN. In this comment, we make an attempt to assemble the pieces of the puzzle arguing that the activated PECs might play a significant role and could represent a target for novel treatment strategies for RPGN and FSGS.
Collapse
Affiliation(s)
- Marcus J Moeller
- University Hospital of the Aachen University of Technology, Aachen, Germany.
| | | |
Collapse
|
35
|
Kunz M, Dannemann M, Kelso J. High-throughput sequencing of the melanoma genome. Exp Dermatol 2012; 22:10-7. [PMID: 23174022 DOI: 10.1111/exd.12054] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2012] [Indexed: 12/16/2022]
Abstract
Next-generation sequencing technologies are now common for whole-genome, whole-exome and whole-transcriptome sequencing (RNA-seq) of tumors to identify point mutations, structural or copy number alterations and changes in gene expression. A substantial number of studies have already been performed for melanoma. One study analysed eight melanoma cell lines with RNA-Seq technology and identified 11 novel melanoma gene fusions. Whole-exome sequencing of seven melanoma cell lines identified overlapping gain of function mutations in MAP2K1 (MEK1) and MAP2K2 (MEK2) genes. Integrative sequencing of cutaneous melanoma metastases using different sequencing platforms revealed a new somatic point mutation in HRAS and a structural rearrangement affecting CDKN2C (a CDK4 inhibitor). These latter sequencing-based discoveries may be used to motivate the inclusion of the affected patients into clinical trials with specific signalling pathway inhibitors. Taken together, we are at the beginning of an era with new sequencing technologies providing a more comprehensive view of cancer mutational landscapes and hereby a better understanding of their pathogenesis. This will also open interesting perspectives for new treatment approaches and clinical trial designs.
Collapse
Affiliation(s)
- Manfred Kunz
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Leipzig, Germany.
| | | | | |
Collapse
|
36
|
Verstraete K, Savvides SN. Extracellular assembly and activation principles of oncogenic class III receptor tyrosine kinases. Nat Rev Cancer 2012; 12:753-66. [PMID: 23076159 DOI: 10.1038/nrc3371] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Intracellular signalling cascades initiated by class III receptor tyrosine kinases (RTK-IIIs) and their cytokine ligands contribute to haematopoiesis and mesenchymal tissue development. They are also implicated in a wide range of inflammatory disorders and cancers. Recent snapshots of RTK-III ectodomains in complex with cognate cytokines have revealed timely insights into the structural determinants of RTK-III activation, evolution and pathology. Importantly, candidate 'driver' and 'passenger' mutations that have been identified in RTK-IIIs can now be collectively mapped for the first time to structural scaffolds of the corresponding RTK-III ectodomains. Such insights will generate a renewed interest in dissecting the mechanistic effects of such mutations and their therapeutic relevance.
Collapse
Affiliation(s)
- Kenneth Verstraete
- Unit for Structural Biology, Laboratory for Protein Biochemistry and Biomolecular Engineering, Ghent University, K.L. Ledeganckstraat 35, 9000 Ghent, Belgium.
| | | |
Collapse
|
37
|
Aman J, van Bezu J, Damanafshan A, Huveneers S, Eringa EC, Vogel SM, Groeneveld ABJ, Vonk Noordegraaf A, van Hinsbergh VWM, van Nieuw Amerongen GP. Effective treatment of edema and endothelial barrier dysfunction with imatinib. Circulation 2012; 126:2728-38. [PMID: 23099479 DOI: 10.1161/circulationaha.112.134304] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Tissue edema and endothelial barrier dysfunction as observed in sepsis and acute lung injury carry high morbidity and mortality, but currently lack specific therapy. In a recent case report, we described fast resolution of pulmonary edema on treatment with the tyrosine kinase inhibitor imatinib through an unknown mechanism. Here, we explored the effect of imatinib on endothelial barrier dysfunction and edema formation. METHODS AND RESULTS We evaluated the effect of imatinib on endothelial barrier function in vitro and in vivo. In human macro- and microvascular endothelial monolayers, imatinib attenuated endothelial barrier dysfunction induced by thrombin and histamine. Small interfering RNA knock-downs of the imatinib-sensitive kinases revealed that imatinib attenuates endothelial barrier dysfunction via inhibition of Abl-related gene kinase (Arg/Abl2), a previously unknown mediator of endothelial barrier dysfunction. Indeed, Arg was activated by endothelial stimulation with thrombin, histamine, and vascular endothelial growth factor. Imatinib limited Arg-mediated endothelial barrier dysfunction by enhancing Rac1 activity and enforcing adhesion of endothelial cells to the extracellular matrix. Using mouse models of vascular leakage as proof-of-concept, we found that pretreatment with imatinib protected against vascular endothelial growth factor-induced vascular leakage in the skin, and effectively prevented edema formation in the lungs. In a murine model of sepsis, imatinib treatment (6 hours and 18 hours after induction of sepsis) attenuated vascular leakage in the kidneys and the lungs (24 hours after induction of sepsis). CONCLUSIONS Thus, imatinib prevents endothelial barrier dysfunction and edema formation via inhibition of Arg. These findings identify imatinib as a promising approach to permeability edema and indicate Arg as novel target for edema treatment.
Collapse
Affiliation(s)
- Jurjan Aman
- Department of Physiology, VU University Medical Center, Van der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Imura M, Kojima Y, Kubota Y, Hamakawa T, Yasui T, Sasaki S, Hayashi Y, Kohri K. Regulation of cell proliferation through a KIT-mediated mechanism in benign prostatic hyperplasia. Prostate 2012; 72:1506-13. [PMID: 22314612 DOI: 10.1002/pros.22500] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Accepted: 01/11/2012] [Indexed: 11/09/2022]
Abstract
BACKGROUND We investigated the role of the KIT-mediated mechanism in benign prostatic hyperplasia (BPH), and discuss the pathophysiology of BPH and a candidate target of BPH medical therapy. METHODS We performed RT-PCR, Western blotting, and immunohistochemistry to examine the expression of KIT in the prostate using a human prostate stromal cell line (PrSC) and human prostate. To investigate the pathophysiological function of KIT, the effects of KIT ligand, stem cell factor (SCF), and imatinib mesylate on cell proliferation were investigated using PrSC. Additionally, we compared the expression level and distribution of KIT in normal prostate and BPH of humans to clarify the contribution of KIT to the pathogenesis of BPH. RESULTS KIT was expressed in PrSC and human prostate, indicating that these samples are suitable for examining the function of KIT. Immunohistochemical analysis demonstrated that KIT was localized in interstitial cells (ICs) of the stromal component in human prostate. Administration of imatinib mesylate dose-dependently inhibited cell proliferation of PrSC with downregulation of JAK2 and STAT1, which are the main pathways downstream of SCF/KIT signal. SCF promoted cell proliferation of PrSC with upregulation of JAK2 and STAT1. KIT expression and the number of KIT-positive ICs in BPH were found to be significantly larger than in normal prostate. CONCLUSIONS This is the first report to suggest that KIT regulates cell proliferation in the prostate and plays a significant role in the pathophysiology of BPH. Our study may lead to a greater understanding of the mechanism of BPH and provide a therapeutic target.
Collapse
Affiliation(s)
- Makoto Imura
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya, Japan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
The nuclear factor-κB (NF-κB) transcription factor family has been considered the central mediator of the inflammatory process and a key participant in innate and adaptive immune responses. Coincident with the molecular cloning of NF-κB/RelA and identification of its kinship to the v-Rel oncogene, it was anticipated that NF-κB itself would be involved in cancer development. Oncogenic activating mutations in NF-κB genes are rare and have been identified only in some lymphoid malignancies, while most NF-κB activating mutations in lymphoid malignancies occur in upstream signaling components that feed into NF-κB. NF-κB activation is also prevalent in carcinomas, in which NF-κB activation is mainly driven by inflammatory cytokines within the tumor microenvironment. Importantly, however, in all malignancies, NF-κB acts in a cell type-specific manner: activating survival genes within cancer cells and inflammation-promoting genes in components of the tumor microenvironment. Yet, the complex biological functions of NF-κB have made its therapeutic targeting a challenge.
Collapse
Affiliation(s)
- Joseph A DiDonato
- Cleveland Clinic Foundation, Department of Cell Biology, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, USA
| | | | | |
Collapse
|
40
|
van Steensel L, Paridaens D, van Meurs M, van Hagen PM, van den Bosch WA, Kuijpers RWAM, Drexhage HA, Hooijkaas H, Dik WA. Orbit-infiltrating mast cells, monocytes, and macrophages produce PDGF isoforms that orchestrate orbital fibroblast activation in Graves' ophthalmopathy. J Clin Endocrinol Metab 2012; 97:E400-8. [PMID: 22238384 DOI: 10.1210/jc.2011-2697] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE Platelet-derived growth factors (PDGF) are regulators of fibroblast activity that may be involved in the pathophysiology of Graves' ophthalmopathy (GO). We unraveled the expression and origin of PDGF family members in GO orbital tissue and investigated the effect of PDGF isoforms on IL-6 and hyaluronan production and proliferation by orbital fibroblasts. METHODS PDGF-A, PDGF-B, PDGF-C, PDGF-D, PDGF-Rα, and PDGF-Rβ expression was determined by real-time quantitative PCR and PDGF-A and PDGF-B protein expression was determined by Western blot in orbital tissues. Orbital tissues were immunohistochemically stained for PDGF-A and PDGF-B expression, together with stainings for T cells, monocytes, B cells, macrophages, and mast cells. Effects of PDGF-AA, PDGF-AB, and PDGF-BB on orbital fibroblast proliferation and IL-6 and hyaluronan production were examined. Finally, effects of PDGF-BB- and PDGF-AA-neutralizing antibodies on IL-6 and hyaluronan production in GO whole orbital tissue cultures were tested. RESULTS GO orbital tissue showed increased PDGF-A and PDGF-B mRNA and protein levels. Increased numbers of PDGF-A- and PDGF-B-positive monocytes, macrophages, and mast cells were present in GO orbital tissue. PDGF-BB stimulated proliferation and hyaluronan and IL-6 production by orbital fibroblasts the most, followed by PDGF-AB and PDGF-AA. Finally, in particular imatinib mesylate and PDGF-BB-neutralizing antibodies reduced IL-6 and hyaluronan production by whole orbital tissue cultures from GO patients. CONCLUSIONS In GO, mast cells, monocytes, and macrophages may activate orbital fibroblasts via secretion of especially PDGF-AB and PDGF-BB. Preclinical studies with whole orbital tissue cultures show that blocking PDGF-B chain containing isoforms can be a promising treatment for GO.
Collapse
Affiliation(s)
- L van Steensel
- Department of Immunology, Erasmus Medical Center, University Medical Center, and Rotterdam Eye Hospital, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Reardon DA, Desjardins A, Vredenburgh JJ, Herndon JE, Coan A, Gururangan S, Peters KB, McLendon R, Sathornsumetee S, Rich JN, Lipp ES, Janney D, Friedman HS. Phase II study of Gleevec plus hydroxyurea in adults with progressive or recurrent low-grade glioma. Cancer 2012; 118:4759-67. [PMID: 22371319 DOI: 10.1002/cncr.26541] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 05/26/2011] [Accepted: 06/21/2011] [Indexed: 11/06/2022]
Abstract
BACKGROUND We evaluated the efficacy of imatinib plus hydroxyurea in patients with progressive/recurrent low-grade glioma. METHODS A total of 64 patients with recurrent/progressive low-grade glioma were enrolled in this single-center study that stratified patients into astrocytoma and oligodendroglioma cohorts. All patients received 500 mg of hydroxyurea twice a day. Imatinib was administered at 400 mg per day for patients not on enzyme-inducing antiepileptic drugs (EIAEDs) and at 500 mg twice a day if on EIAEDs. The primary endpoint was progression-free survival at 12 months (PFS-12) and secondary endpoints were safety, median progression-free survival, and radiographic response rate. RESULTS Thirty-two patients were enrolled into each cohort. Eleven patients (17%) had before radiotherapy and 24 (38%) had received before chemotherapy. The median PFS and PFS-12 were 11 months and 39%, respectively. Outcome did not differ between the histologic cohorts. No patient achieved a radiographic response. The most common grade 3 or greater adverse events were neutropenia (11%), thrombocytopenia (3%), and diarrhea (3%). CONCLUSIONS Imatinib plus hydroxyurea was well tolerated among recurrent/progressive LGG patients but this regimen demonstrated negligible antitumor activity.
Collapse
Affiliation(s)
- David A Reardon
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Bai C, Liu X, Zheng JM, Qiu C, Zhu Y, Xu J, Zhao J, Ma D. A novel PDGFRA mutation in gastrointestinal stromal tumours, L839P, is sensitive to imatinib in vitro. Oncol Lett 2012; 3:1139-1143. [PMID: 22783407 DOI: 10.3892/ol.2012.599] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 01/23/2012] [Indexed: 12/16/2022] Open
Abstract
Evidence suggests that different types of mutation in gastrointestinal stromal tumours (GISTs) correlate with different response rates to imatinib (Glivec, STI571). The purpose of this study was to explore the sensitivity of the PDGFRA(L839P) mutant, a novel gain-of-function mutation isoform related to GISTs, to imatinib in vitro. The eukaryotic expression vectors pcDNA3.1-PDGFRA(Wild), pcDNA3.1-PDGFRA(D842V) and pcDNA3.1-PDGFRA(L839P) were constructed and transfected into Chinese hamster ovary (CHO) cells by liposome methods. The responses of cells with PDGFRA(Wild), PDGFRA(L839P) and PDGFRA(D842V) mutants to imatinib were determined by methyl thiazolyl tetrazolium (MTT) assay, western blotting and apoptosis assays. Reults of the MTT assay revealed that the growth rate of CHO(PDGFRA(L839P)) cells decreased to approximately 60% when exposed to 1 μM imatinib and to approximately 50% with 5 μM imatinib. However, the growth rate of CHO(PDGFRA(D842V)) cells did not significantly change with 5 μM imatinib. Western blot analysis indicated that 1 μM imatinib completely blocked the phosphorylation of PDGFRA(L839P), but did not affect PDGFRA(D842V) phosphorylation. Apoptosis analysis suggested that the percentage of apoptotic CHO(PDGFRA(L839P)) cells increased approximately 4-fold (from 5.90 to 25.2%) with 1 μM imatinib. Although the treatment of CHO(PDGFRA(D842V)) and CHO(PDGFRA(Wild)) cells with 5 μM imatinib resulted in a slight increase in the number of apoptotic cells, the percentage of apoptotic cells remained approximately 10% of the total population. Our findings showed that the PDGFRA gene mutation isoform L839P is sensitive to inhibition by imatinib. Screening for PDGFRA mutations in GISTs is essential to identify the response to treatment with imatinib.
Collapse
Affiliation(s)
- Chenguang Bai
- Department of Pathology, Changhai Hospital, Shanghai, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
He M, Wei MJ. Reversing multidrug resistance by tyrosine kinase inhibitors. CHINESE JOURNAL OF CANCER 2012; 31:126-33. [PMID: 22237041 PMCID: PMC3777484 DOI: 10.5732/cjc.011.10315] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Recently, a large number of tyrosine kinase inhibitors (TKIs) have been developed as anticancer agents. These TKIs can specifically and selectively inhibit tumor cell growth and metastasis by targeting various tyrosine kinases and thereby interfering with cellular signaling pathways. The therapeutic potential of TKIs has been hindered by multidrug resistance (MDR), which is commonly caused by overexpression of ATP-binding cassette (ABC) membrane transporters. Interestingly, some TKIs have also been found to reverse MDR by directly inhibiting the function of ABC transporters and enhancing the efficacy of conventional chemotherapeutic drugs. In this review, we discuss ABC transporter-mediated MDR to TKIs and MDR reversal by TKIs.
Collapse
Affiliation(s)
- Miao He
- Department of Pharmacology, Pharmaceutical College of China Medical University, Shenyang, Liaoning 110001, P. R. China
| | | |
Collapse
|
44
|
Bilir A, Erguven M, Ermis E, Sencan M, Yazihan N. Combination of imatinib mesylate with lithium chloride and medroxyprogesterone acetate is highly active in Ishikawa endometrial carcinoma in vitro. J Gynecol Oncol 2011; 22:225-32. [PMID: 22247798 PMCID: PMC3254840 DOI: 10.3802/jgo.2011.22.4.225] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2011] [Revised: 05/11/2011] [Accepted: 06/17/2011] [Indexed: 01/22/2023] Open
Abstract
Objective The aim of the study was to investigate whether lithium chloride and medroxyprogesterone acetate can potentiate the cytotoxicity of imatinib mesylate in human endometrial cancer in vitro and the effect of midkine in these therapies. Methods Imatinib mesylate (50 µM), lithium chloride (100 µM), medroxyprogesterone acetate (200 µM) and their combination were applied to monolayer and three dimensional cultures of human Ishikawa endometrial cancer for 72 hours. The cell proliferation index, apoptotic index, caspase-3 and midkine levels, cell cycle distributions in monolayer cultures and cell ultrastructure in spheroid cultures were evaluated. Results were statistically analyzed using the Student's t-test. Results All drug applications inhibited cell proliferation (p<0.05), however the combination were the effective groups for 72 hours (p<0.05). Interestingly, although the loss of efficiency was seen higly seen every 24 hours at single applications, the inhibition rates of the combination groups were almost same for 72 hours. In concordance with these results, the apoptotic index, caspase-3 levels (p<0.05), cell morphology and ultrastructure damages were much higher in the combination groups. Imatinib mesylate induced S-phase arrest, however other groups induced G0+G1-phase arrest at 24 hours and all groups induced G0+G1 arrest at 72 hours (p<0.05). Imatinib mesylate and imatinib mesylate with medroxyprogesterone acetate induced highest decrease in midkine levels, respectively (p<0.05). Conclusion The present study showed that the combination of imatinib mesylate with lithium chloride and medroxyprogesterone acetate is highly active in Ishikawa endometrial carcinoma in vitro and the inhibition of midkine involved in their mechanism of action against endometrium defense.
Collapse
Affiliation(s)
- Ayhan Bilir
- Department of Histology and Embryology, Istanbul University Faculty of Medicine, Istanbul, Turkey
| | | | | | | | | |
Collapse
|
45
|
Ammoun S, Schmid MC, Triner J, Manley P, Hanemann CO. Nilotinib alone or in combination with selumetinib is a drug candidate for neurofibromatosis type 2. Neuro Oncol 2011; 13:759-66. [PMID: 21727212 DOI: 10.1093/neuonc/nor056] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Loss of the tumor suppressor merlin is a cause of frequent tumors of the nervous system, such as schwannomas, meningiomas, and ependymomas, which occur spontaneously or as part of neurofibromatosis type 2 (NF2). Because there is medical need for drug therapies for these tumors, our aim is to find therapeutic targets. We have studied the pathobiology of schwannomas, because they are the most common merlin-deficient tumors and are a model for all merlin-deficient tumors. With use of a human schwannoma in vitro model, we previously described strong overexpression/activation of platelet-derived growth factor receptor-β (PDGFR-β) leading to strong, long-lasting activation of extracellular-signal-regulated kinase (ERK1/2) and AKT and increased schwannoma growth, which we successfully inhibited using the PDGFR/Raf inhibitor sorafenib. However, the benign character of schwannomas may require long-term treatment; thus, drug tolerability is an issue. With the use of Western blotting, proliferation assays, viability assays, and a primary human schwannoma cell in vitro model, we tested the PDGFR/c-KIT inhibitors imatinib (Glivec(;) Novartis) and nilotinib (Tasigna(;) Novartis). Imatinib and nilotinib inhibited PDGF-DD-mediated ERK1/2 activation, basal and PDGF-DD-mediated activation of PDGFR-β and AKT, and schwannoma proliferation. Nilotinib is more potent than imatinib, exerting its maximal inhibitory effect at concentrations lower than steady-state trough plasma levels. In addition, nilotinib combined with the MEK1/2 inhibitor selumetinib (AZD6244) at low concentrations displayed stronger efficiency toward tumor growth inhibition, compared with nilotinib alone. We suggest that therapy with nilotinib or combinational therapy that simultaneously inhibits PDGFR and the downstream Raf/MEK1/2/ERK1/2 pathway could represent an effective treatment for schwannomas and other merlin-deficient tumors.
Collapse
Affiliation(s)
- Sylwia Ammoun
- Clinical Neurobiology, Peninsula College for Medicine and Dentistry, The John Bull Building, Tamar Science Park, Research Way, Plymouth PL6 8BU, United Kingdom
| | | | | | | | | |
Collapse
|
46
|
Erguven M, Bilir A, Yazihan N, Korkmaz S, Aktas E, Ovalioglu C, Dundar T, Seyithanoglu H. Imatinib mesylate decreases the cytotoxic effect of roscovitine on human glioblastoma cells in vitro and the role of midkine. Oncol Lett 2011; 3:200-208. [PMID: 22740881 DOI: 10.3892/ol.2011.434] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 08/31/2011] [Indexed: 01/16/2023] Open
Abstract
The purpose of the present study was to overcome resistance to imatinib (IM) by combining it with roscovitine (ROSC) and to investigate whether or not midkine (MK) had an effect on this combination in the treatment of glioblastoma (GBL). Human T98 GBL cells were used to evaluate the effects of IM (10 μM), ROSC (200 μM) and their combination on the cell proliferation index, apoptotic index, the apoptotic protein and anti-apoptotic protein levels, and ultrastructure. All applications decreased the cell proliferation index and increased the apoptotic index, but ROSC was the most efficient drug and the second most efficient drug was IM. Notably, ROSC increased anti-apoptotic proteins levels (PDGFR-α, AQP-4, hTERT), COX-1 activity and ribosome numbers. The effects of ROSC on hTERT, MK, AQP-4 and MRP-1 levels and COX-1 activity were reported for the first time. ROSC induced the highest increase in caspase-3 levels. Autophagy was not involved in the activity of ROSC in GBL spheroids. The combination of IM with ROSC showed an antagonist effect in the treatment of human GBL cells. The combination group decreased certain anti-apoptotic protein levels (PDGFR-α, EGFR, p-gp, MRP-1 and MK), cAMP levels, COX-1 activity and apoptotic protein levels (caspase-3). However, it induced the highest increase in hTERT levels and COX-2 activity. Ribosome numbers were much lower than those in the ROSC group and no autophagic vacuole was observed. In conclusion, more investigations are required to identify the key regulatory components that are responsible for this antagonism; however, the determination of this combination therapy as a failure therapy may be precautionary for oncologists in the treatment of GBL patients and potentially may contribute to the efficacy of new therapeutic regimens.
Collapse
Affiliation(s)
- Mine Erguven
- Faculty of Medicine, Department of Biochemistry, Yeni Yüzyıl University, Istanbul
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Imatinib Mesylate (Gleevec) as Protein-tyrosine Kinase Inhibitor Elicits Smooth Muscle Relaxation in Isolated Human Prostatic Tissue. Urology 2011; 78:968.e1-6. [DOI: 10.1016/j.urology.2011.06.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 06/14/2011] [Accepted: 06/14/2011] [Indexed: 11/21/2022]
|
48
|
Boersch C, Merkul E, Müller TJJ. Katalytische Synthesen N-heterocyclischer Inone und Indione durch In-situ-Aktivierung von Carbonsäuren mit Oxalylchlorid. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201103296] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
49
|
Boersch C, Merkul E, Müller TJJ. Catalytic Syntheses of N-Heterocyclic Ynones and Ynediones by In Situ Activation of Carboxylic Acids with Oxalyl Chloride. Angew Chem Int Ed Engl 2011; 50:10448-52. [DOI: 10.1002/anie.201103296] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Indexed: 11/05/2022]
|
50
|
Bengió RM, Riva ME, Moiraghi B, Lanari E, Milone J, Ventriglia V, Bullorsky E, Tezanos pinto MD, Murro H, Bianchini M, Larripa I. Clinical outcome of chronic myeloid leukemia imatinib-resistant patients: do BCR–ABL kinase domain mutations affect patient survival? First multicenter Argentinean study. Leuk Lymphoma 2011; 52:1720-6. [DOI: 10.3109/10428194.2011.578310] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|