51
|
Ubiquitination and SUMOylation in the chronic inflammatory tumor microenvironment. Biochim Biophys Acta Rev Cancer 2018; 1870:165-175. [DOI: 10.1016/j.bbcan.2018.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/10/2018] [Accepted: 08/15/2018] [Indexed: 12/28/2022]
|
52
|
Abstract
The transcription factor NF-κB is a critical regulator of immune and inflammatory responses. In mammals, the NF-κB/Rel family comprises five members: p50, p52, p65 (Rel-A), c-Rel, and Rel-B proteins, which form homo- or heterodimers and remain as an inactive complex with the inhibitory molecules called IκB proteins in resting cells. Two distinct NF-κB signaling pathways have been described: 1) the canonical pathway primarily activated by pathogens and inflammatory mediators, and 2) the noncanonical pathway mostly activated by developmental cues. The most abundant form of NF-κB activated by pathologic stimuli via the canonical pathway is the p65:p50 heterodimer. Disproportionate increase in activated p65 and subsequent transactivation of effector molecules is integral to the pathogenesis of many chronic diseases such as the rheumatoid arthritis, inflammatory bowel disease, multiple sclerosis, and even neurodegenerative pathologies. Hence, the NF-κB p65 signaling pathway has been a pivotal point for intense drug discovery and development. This review begins with an overview of p65-mediated signaling followed by discussion of strategies that directly target NF-κB p65 in the context of chronic inflammation.
Collapse
Affiliation(s)
- Sivagami Giridharan
- Department of Oral Medicine, Madha Dental College, Kundrathur, Chennai, TN, India
| | - Mythily Srinivasan
- Department of Oral Pathology, Medicine and Radiology, Indiana University School of Dentistry, Indiana University Purdue University at Indianapolis, Indianapolis, IN, USA,
- Provaidya LLC, Indianapolis, IN, USA,
| |
Collapse
|
53
|
Evans SM, Rodino KG, Adcox HE, Carlyon JA. Orientia tsutsugamushi uses two Ank effectors to modulate NF-κB p65 nuclear transport and inhibit NF-κB transcriptional activation. PLoS Pathog 2018; 14:e1007023. [PMID: 29734393 PMCID: PMC5957444 DOI: 10.1371/journal.ppat.1007023] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 05/17/2018] [Accepted: 04/11/2018] [Indexed: 01/05/2023] Open
Abstract
Orientia tsutsugamushi causes scrub typhus, a potentially fatal infection that threatens over one billion people. Nuclear translocation of the transcription factor, NF-κB, is the central initiating cellular event in the antimicrobial response. Here, we report that NF-κB p65 nuclear accumulation and NF-κB-dependent transcription are inhibited in O. tsutsugamushi infected HeLa cells and/or primary macrophages, even in the presence of TNFα. The bacterium modulates p65 subcellular localization by neither degrading it nor inhibiting IκBα degradation. Rather, it exploits host exportin 1 to mediate p65 nuclear export, as this phenomenon is leptomycin B-sensitive. O. tsutsugamushi antagonizes NF-κB-activated transcription even when exportin 1 is inhibited and NF-κB consequently remains in the nucleus. Two ankyrin repeat-containing effectors (Anks), Ank1 and Ank6, each of which possess a C-terminal F-box and exhibit 58.5% amino acid identity, are linked to the pathogen's ability to modulate NF-κB. When ectopically expressed, both translocate to the nucleus, abrogate NF-κB-activated transcription in an exportin 1-independent manner, and pronouncedly reduce TNFα-induced p65 nuclear levels by exportin 1-dependent means. Flag-tagged Ank 1 and Ank6 co-immunoprecipitate p65 and exportin 1. Both also bind importin β1, a host protein that is essential for the classical nuclear import pathway. Importazole, which blocks importin β1 activity, abrogates Ank1 and Ank6 nuclear translocation. The Ank1 and Ank6 regions that bind importin β1 also mediate their transport into the nucleus. Yet, these regions are distinct from those that bind p65/exportin 1. The Ank1 and Ank6 F-box and the region that lies between it and the ankyrin repeat domain are essential for blocking p65 nuclear accumulation. These data reveal a novel mechanism by which O. tsutsugamushi modulates the activity and nuclear transport of NF-κB p65 and identify the first microbial proteins that co-opt both importin β1 and exportin 1 to antagonize a critical arm of the antimicrobial response.
Collapse
Affiliation(s)
- Sean M. Evans
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, United States of America
| | - Kyle G. Rodino
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, United States of America
| | - Haley E. Adcox
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, United States of America
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, United States of America
| |
Collapse
|
54
|
Gandhi UH, Senapedis W, Baloglu E, Unger TJ, Chari A, Vogl D, Cornell RF. Clinical Implications of Targeting XPO1-mediated Nuclear Export in Multiple Myeloma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2018; 18:335-345. [PMID: 29610030 DOI: 10.1016/j.clml.2018.03.003] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 02/26/2018] [Accepted: 03/08/2018] [Indexed: 12/30/2022]
Abstract
Multiple myeloma (MM) is a malignancy of plasma cells that is typically chronic, and relapse is common. Current therapeutic strategies include combination and sequential treatments with corticosteroids, alkylating agents, proteasomal inhibitors, immunomodulators, and monoclonal antibodies. These drugs prolong survival but ultimately become ineffective. Exportin 1 (XPO1), a nuclear export protein, is overexpressed in MM cells, and knockdown studies have suggested that XPO1 is essential for MM cell survival. Selective inhibitor of nuclear export (SINE) compounds are novel, orally bioavailable class of agents that specifically inhibit XPO1. Selinexor (KPT-330) is the first-in-human SINE compound. Early phase clinical trials have established the safety profile of this agent and have shown promising efficacy in combination with low-dose dexamethasone and other anti-MM agents. The combination of selinexor and dexamethasone has demonstrated activity in "penta-refractory" MM, (ie, MM refractory to the 5 most active anti-MM agents currently used in treatment). We have reviewed the available data on the molecular implications of XPO1 inhibition in MM. We also reviewed the pertinent early phase clinical data with SINE compounds and discuss management strategies for common toxicities encountered with use of selinexor.
Collapse
Affiliation(s)
- Ujjawal H Gandhi
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN
| | | | | | | | - Ajai Chari
- Division of Hematology and Oncology, Mount Sinai Hospital, New York, NY
| | - Dan Vogl
- Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA
| | - Robert F Cornell
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
55
|
Miyasato Y, Yoshizawa T, Sato Y, Nakagawa T, Miyasato Y, Kakizoe Y, Kuwabara T, Adachi M, Ianni A, Braun T, Komohara Y, Mukoyama M, Yamagata K. Sirtuin 7 Deficiency Ameliorates Cisplatin-induced Acute Kidney Injury Through Regulation of the Inflammatory Response. Sci Rep 2018; 8:5927. [PMID: 29651144 PMCID: PMC5897539 DOI: 10.1038/s41598-018-24257-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 03/29/2018] [Indexed: 12/31/2022] Open
Abstract
Cisplatin-induced acute kidney injury (AKI) has been recognized as one of cisplatin’s serious side effects, limiting its use in cancer therapy. Sirtuin 1 (SIRT1) and SIRT3 play protective roles against cisplatin-induced kidney injury. However, the role of SIRT7 in cisplatin-induced kidney injury is not yet known. In this study, we found that Sirt7 knockout (KO) mice were resistant to cisplatin-induced AKI. Furthermore, our studies identified that loss of SIRT7 decreases the expression of tumor necrosis factor-α (TNF-α) by regulating the nuclear expression of the transcription factor nuclear factor kappa B. It has been reported that cisplatin-induced nephrotoxicity is mediated by TNF-α. Our results indicate that SIRT7 plays an important role in cisplatin-induced AKI and suggest the possibility of SIRT7 as a novel therapeutic target for cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Yoshikazu Miyasato
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan.,Department of Nephrology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Tatsuya Yoshizawa
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Yoshifumi Sato
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Terumasa Nakagawa
- Department of Nephrology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Yuko Miyasato
- Department of Cell Pathology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Yutaka Kakizoe
- Department of Nephrology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Takashige Kuwabara
- Department of Nephrology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Masataka Adachi
- Department of Nephrology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Alessandro Ianni
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Yoshihiro Komohara
- Department of Cell Pathology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Masashi Mukoyama
- Department of Nephrology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Kazuya Yamagata
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan.
| |
Collapse
|
56
|
Robichaux WG, Cheng X. Intracellular cAMP Sensor EPAC: Physiology, Pathophysiology, and Therapeutics Development. Physiol Rev 2018; 98:919-1053. [PMID: 29537337 PMCID: PMC6050347 DOI: 10.1152/physrev.00025.2017] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 12/13/2022] Open
Abstract
This review focuses on one family of the known cAMP receptors, the exchange proteins directly activated by cAMP (EPACs), also known as the cAMP-regulated guanine nucleotide exchange factors (cAMP-GEFs). Although EPAC proteins are fairly new additions to the growing list of cAMP effectors, and relatively "young" in the cAMP discovery timeline, the significance of an EPAC presence in different cell systems is extraordinary. The study of EPACs has considerably expanded the diversity and adaptive nature of cAMP signaling associated with numerous physiological and pathophysiological responses. This review comprehensively covers EPAC protein functions at the molecular, cellular, physiological, and pathophysiological levels; and in turn, the applications of employing EPAC-based biosensors as detection tools for dissecting cAMP signaling and the implications for targeting EPAC proteins for therapeutic development are also discussed.
Collapse
Affiliation(s)
- William G Robichaux
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center , Houston, Texas
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center , Houston, Texas
| |
Collapse
|
57
|
Hu M, Cheng YX, Yang X, Yu J, Huang J, Hong L. Dysregulation of CD44v6 may lead to recurrent spontaneous abortion by inhibiting the proliferation and migration of trophoblast cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:2072-2079. [PMID: 31938314 PMCID: PMC6958214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 02/05/2018] [Indexed: 06/10/2023]
Abstract
In the establishment and continuation of a successful pregnancy, the proliferation, migration, and invasion of trophoblast cells play essential roles. Impaired trophoblast function has been reported to be involved in recurrent spontaneous abortion (RSA) but the potential mechanisms are still unknown. CD44 variant domain 6 (CD44v6) is a transmembrane glycoprotein that has been known for decades to be expressed in the placenta. In this study, we investigated the effect of CD44v6 on proliferation and migration of trophoblast cells. Our results show that CD44v6 expression in the villi is lower in RSA patients than in women with normal pregnancies. Furthermore, downregulation of CD44v6 leads to a reduction in proliferation and migration in the human trophoblast cell line HTR-8/SVneo, along with decreased expression and nuclear translocation of NF-κB. These results suggest that decreased expression of CD44v6 may cause miscarriage by downregulating trophoblast cell proliferation via the NF-κB pathway.
Collapse
Affiliation(s)
- Min Hu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan UniversityWuhan, Hubei, PR China
| | - Yan-Xiang Cheng
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan UniversityWuhan, Hubei, PR China
| | - Xiao Yang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan UniversityWuhan, Hubei, PR China
| | - Jia Yu
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan UniversityWuhan, Hubei, PR China
| | - Jinling Huang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan UniversityWuhan, Hubei, PR China
| | - Li Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan UniversityWuhan, Hubei, PR China
| |
Collapse
|
58
|
PORDEL S, NEMATI K, KARIMI MH, DOROUDCHI M. NF-κB1 Rs28362491 Mutant Allele Frequencies along the Silk Road and Beyond. IRANIAN JOURNAL OF PUBLIC HEALTH 2018; 47:397-406. [PMID: 29845028 PMCID: PMC5971177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND In the human evolutionary history, Single Nucleotide Polymorphism (SNP) frequencies are valuable in terms of finding connections between different populations. Due to the pronounced role of the immune system in combating pathogens and environmental stressors, polymorphisms in the immune genes are subject to selection pressure of the diseases as well. The functional polymorphisms in NF-κB1 promoter (-94 ins/del) are associated with different diseases; therefore, we aimed to establish the frequencies of NF-κB1 rs28362491 alleles in a population of Southwestern Iranians in comparison with the world populations. METHODS We assessed the polymorphism of -94 ATTG ins/del (rs28362491) in 201 Iranian healthy blood donors from Fars Province, central Iran in a one year period between 2015 and 2016 by PCR-RFLP method using DNA extracted from peripheral blood mononuclear cells. RESULTS The frequency of ins/ins homozygote genotype was found to be 46.97%. The frequency of heterozygote individuals was 42.42% and the percentage of del/del homozygote genotype was 10.61%. We observed a genetic similarity based on the genotype frequencies of NF-κB1 -94 ins/del ATTG polymorphism between our sample of Iranians with American Jewish, Turkish, American non-Jewish, Chinese-Uyghurs and Germans. CONCLUSION The results confirmed genetic interrelation of Iranians with some ancient neighbors and their admixture with countries along the Silk Road. We suggest that mapping the distribution of NF-κB1-94 ATTG ins/del along with HLA genes may help to better define the relations between human populations and design population-specific vaccines for pathogens with a high rate of variation.
Collapse
Affiliation(s)
- Safoora PORDEL
- Dept. of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kazem NEMATI
- Dept. of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mehrnoosh DOROUDCHI
- Dept. of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran,Corresponding Author:
| |
Collapse
|
59
|
Regulation of inflammatory responses by dynamic subcellular localization of RNA-binding protein Arid5a. Proc Natl Acad Sci U S A 2018; 115:E1214-E1220. [PMID: 29358370 PMCID: PMC5819453 DOI: 10.1073/pnas.1719921115] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Adenine-thymine (AT)-rich interactive domain 5a (Arid5a) is an RNA-binding protein found in the cytoplasm and nucleus of normally growing cells. Although Arid5a is known to play an important role in immune regulation, whether and how Arid5a subcellular localization impacts immune regulation has remained unclear. In this study, we generated Arid5a transgenic (TG) mice to address this question. While ectopic Arid5a overexpression did not affect expression of inflammatory cytokines under unstimulated conditions, significantly higher levels of inflammatory cytokines, such as IL-6, were produced in response to lipopolysaccharide (LPS) stimulation. Consistent with this, TG mice were more sensitive to LPS treatment than wild-type mice. We also found that Arid5a is imported into the nucleus via a classical importin-α/β1-mediated pathway. On stimulation, nuclear Arid5a levels were decreased, while there was a concomitant increase in cytoplasmic Arid5a. Arid5a is associated with up-frameshift protein 1, and its nuclear export is regulated by a nuclear export receptor, chromosomal region maintenance 1. Taken together, these data indicate that Arid5a is a dynamic protein that translocates to the cytoplasm from the nucleus so as to properly exert its dual function in mRNA stabilization and transcriptional regulation during inflammatory conditions.
Collapse
|
60
|
Pickens JA, Tripp RA. Verdinexor Targeting of CRM1 is a Promising Therapeutic Approach against RSV and Influenza Viruses. Viruses 2018; 10:E48. [PMID: 29361733 PMCID: PMC5795461 DOI: 10.3390/v10010048] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 01/15/2018] [Accepted: 01/17/2018] [Indexed: 12/11/2022] Open
Abstract
Two primary causes of respiratory tract infections are respiratory syncytial virus (RSV) and influenza viruses, both of which remain major public health concerns. There are a limited number of antiviral drugs available for the treatment of RSV and influenza, each having limited effectiveness and each driving selective pressure for the emergence of drug-resistant viruses. Novel broad-spectrum antivirals are needed to circumvent problems with current disease intervention strategies, while improving the cytokine-induced immunopathology associated with RSV and influenza infections. In this review, we examine the use of Verdinexor (KPT-335, a novel orally bioavailable drug that functions as a selective inhibitor of nuclear export, SINE), as an antiviral with multifaceted therapeutic potential. KPT-335 works to (1) block CRM1 (i.e., Chromosome Region Maintenance 1; exportin 1 or XPO1) mediated export of viral proteins critical for RSV and influenza pathogenesis; and (2) repress nuclear factor κB (NF-κB) activation, thus reducing cytokine production and eliminating virus-associated immunopathology. The repurposing of SINE compounds as antivirals shows promise not only against RSV and influenza virus but also against other viruses that exploit the nucleus as part of their viral life cycle.
Collapse
Affiliation(s)
- Jennifer A Pickens
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA.
| | - Ralph A Tripp
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
61
|
McKenna S, Butler B, Jatana L, Ghosh S, Wright CJ. Inhibition of IκBβ/NFκB signaling prevents LPS-induced IL1β expression without increasing apoptosis in the developing mouse lung. Pediatr Res 2017; 82:1064-1072. [PMID: 28753596 PMCID: PMC5761659 DOI: 10.1038/pr.2017.182] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 07/05/2017] [Indexed: 01/09/2023]
Abstract
BackgroundThe pro-inflammatory consequences of IL1β expression contribute to the pathogenesis of bronchopulmonary dysplasia. Selectively targeting Lipopolysaccharide (LPS)-induced IκBβ/NFκB signaling attenuates IL1β mRNA expression in macrophages. Whether targeting IκBβ/NFκB signaling affects the anti-apoptotic gene expression, a known consequence of global LPS-induced NFκB inhibition, is unknown.MethodsMacrophages (RAW 264.7, bone marrow-derived macrophage) were assessed for LPS-induced IL1β mRNA/protein expression, anti-apoptotic gene expression, cell viability (trypan blue exclusion), and activation of apoptosis (caspase-3 and PARP cleavage) following pharmacologic and genetic attenuation of IκBβ/NFκB signaling. Expressions of IL1β and anti-apoptotic genes were assessed in endotoxemic newborn mice (P0) with intact (WT), absent (IκBβ KO), and attenuated (IκBβ overexpressing) IκBβ/NFκB signaling.ResultsIn cultured macrophages, pharmacologic and genetic inhibition of LPS-induced IκBβ/NFκB signaling significantly attenuated IL1β mRNA and protein expression. Importantly, targeting IκBβ/NFκB signaling did not attenuate LPS-induced expression of anti-apoptotic genes or result in cell death. In endotoxemic neonatal mice, targeting LPS-induced IκBβ/NFκB signaling significantly attenuated pulmonary IL1β expression without affecting the anti-apoptotic gene expression.ConclusionTargeting IκBβ/NFκB signaling prevents LPS-induced IL1β expression without inducing apoptosis in cultured macrophages and in the lungs of endotoxemic newborn mice. Inhibiting this pathway may prevent inflammatory injury without affecting the protective role of NFκB activity in the developing lung.
Collapse
Affiliation(s)
- Sarah McKenna
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045
| | - Brittany Butler
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045
| | - Laurie Jatana
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045
| | - Sankar Ghosh
- Department of Microbiology & Immunology, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Clyde J. Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045
| |
Collapse
|
62
|
Abstract
Cancer stem cells can generate tumors from only a small number of cells, whereas differentiated cancer cells cannot. The prominent feature of cancer stem cells is its ability to self-renew and differentiate into multiple types of cancer cells. Cancer stem cells have several distinct tumorigenic abilities, including stem cell signal transduction, tumorigenicity, metastasis, and resistance to anticancer drugs, which are regulated by genetic or epigenetic changes. Like normal adult stem cells involved in various developmental processes and tissue homeostasis, cancer stem cells maintain their self-renewal capacity by activating multiple stem cell signaling pathways and inhibiting differentiation signaling pathways during cancer initiation and progression. Recently, many studies have focused on targeting cancer stem cells to eradicate malignancies by regulating stem cell signaling pathways, and products of some of these strategies are in preclinical and clinical trials. In this review, we describe the crucial features of cancer stem cells related to tumor relapse and drug resistance, as well as the new therapeutic strategy to target cancer stem cells named "differentiation therapy."
Collapse
Affiliation(s)
- Xiong Jin
- 1 Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- 2 Institute of Animal Molecular Biotechnology, Korea University, Seoul, Republic of Korea
| | - Xun Jin
- 3 Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- 4 Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- 5 Institute of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hyunggee Kim
- 1 Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- 2 Institute of Animal Molecular Biotechnology, Korea University, Seoul, Republic of Korea
| |
Collapse
|
63
|
Identification of islet-enriched long non-coding RNAs contributing to β-cell failure in type 2 diabetes. Mol Metab 2017; 6:1407-1418. [PMID: 29107288 PMCID: PMC5681241 DOI: 10.1016/j.molmet.2017.08.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/10/2017] [Accepted: 08/14/2017] [Indexed: 11/21/2022] Open
Abstract
Objective Non-coding RNAs constitute a major fraction of the β-cell transcriptome. While the involvement of microRNAs is well established, the contribution of long non-coding RNAs (lncRNAs) in the regulation of β-cell functions and in diabetes development remains poorly understood. The aim of this study was to identify novel islet lncRNAs differently expressed in type 2 diabetes models and to investigate their role in β-cell failure and in the development of the disease. Methods Novel transcripts dysregulated in the islets of diet-induced obese mice were identified by high throughput RNA-sequencing coupled with de novo annotation. Changes in the level of the lncRNAs were assessed by real-time PCR. The functional role of the selected lncRNAs was determined by modifying their expression in MIN6 cells and primary islet cells. Results We identified about 1500 novel lncRNAs, a number of which were differentially expressed in obese mice. The expression of two lncRNAs highly enriched in β-cells, βlinc2, and βlinc3, correlated to body weight gain and glycemia levels in obese mice and was also modified in diabetic db/db mice. The expression of both lncRNAs was also modulated in vitro in isolated islet cells by glucolipotoxic conditions. Moreover, the expression of the human orthologue of βlinc3 was altered in the islets of type 2 diabetic patients and was associated to the BMI of the donors. Modulation of the level of βlinc2 and βlinc3 by overexpression or downregulation in MIN6 and mouse islet cells did not affect insulin secretion but increased β-cell apoptosis. Conclusions Taken together, the data show that lncRNAs are modulated in a model of obesity-associated type 2 diabetes and that variations in the expression of some of them may contribute to β-cell failure during the development of the disease. Mouse pancreatic islets express a large number of novel long non-coding RNAs. Many long non-coding RNAs are differentially expressed in the islets of obese mice. The level of two islet long non-coding RNAs correlates to body weight and glycemia. The expression of these islet long non-coding RNAs is altered in Type 2 diabetes. Altered expression of these long non-coding RNAs sensitise β-cells to apoptosis.
Collapse
|
64
|
Nair JS, Musi E, Schwartz GK. Selinexor (KPT-330) Induces Tumor Suppression through Nuclear Sequestration of IκB and Downregulation of Survivin. Clin Cancer Res 2017; 23:4301-4311. [PMID: 28314790 DOI: 10.1158/1078-0432.ccr-16-2632] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 11/14/2016] [Accepted: 03/10/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Selinexor, a small molecule that inhibits nuclear export protein XPO1, has demonstrated efficacy in solid tumors and hematologic malignancies with the evidence of clinical activity in sarcoma as a single agent. Treatment options available are very few, and hence the need to identify novel targets and strategic therapies is of utmost importance.Experimental Design: The mechanistic effects of selinexor in sarcomas as a monotherapy and in combination with proteasome inhibitor, carfilzomib, across a panel of cell lines in vitro and few in xenograft mouse models were investigated.Results: Selinexor induced IκB nuclear localization as a single agent, and the effect was enhanced by stabilization of IκB when pretreated with the proteasome inhibitor carfilzomib. This stabilization and retention of IκB in the nucleus resulted in inhibition of NFκB and transcriptional suppression of the critical antiapoptotic protein, survivin. Treatment of carfilzomib followed by selinexor caused selinexor-sensitive and selinexor-resistant cell lines to be more sensitive to selinexor as determined by an increase in apoptosis. This was successfully demonstrated in the MPNST xenograft model with enhanced tumor suppression.Conclusions: The subcellular distributions of IκB and NFκB are indicative of carcinogenesis. Inhibition of XPO1 results in intranuclear retention of IκB, which inhibits NFκB and thereby provides a novel mechanism for drug therapy in sarcoma. This effect can be further enhanced in relatively selinexor-resistant sarcoma cell lines by pretreatment with the proteasome inhibitor carfilzomib. Because of these results, a human clinical trial with selinexor in combination with a proteasome inhibitor is planned for the treatment of sarcoma. Clin Cancer Res; 23(15); 4301-11. ©2017 AACR.
Collapse
Affiliation(s)
- Jayasree S Nair
- Columbia University Medical Center, Herbert Irving Comprehensive Cancer Center, New York, New York.
| | - Elgilda Musi
- Columbia University Medical Center, Herbert Irving Comprehensive Cancer Center, New York, New York
| | - Gary K Schwartz
- Columbia University Medical Center, Herbert Irving Comprehensive Cancer Center, New York, New York
| |
Collapse
|
65
|
Muz B, Azab F, de la Puente P, Landesman Y, Azab AK. Selinexor Overcomes Hypoxia-Induced Drug Resistance in Multiple Myeloma. Transl Oncol 2017; 10:632-640. [PMID: 28668761 PMCID: PMC5496204 DOI: 10.1016/j.tranon.2017.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/20/2017] [Accepted: 04/24/2017] [Indexed: 01/07/2023] Open
Abstract
Increased levels of the nuclear export protein, exportin 1 (XPO1), were demonstrated in multiple myeloma (MM) patients. Targeting XPO1 with selinexor (the selective inhibitor of nuclear export; SINE compound KPT-330) demonstrates broad antitumor activity also in patient cells resistant to bortezomib; hence, it is a promising target in MM patients. Hypoxia is known to mediate tumor progression and drug resistance (including bortezomib resistance) in MM cells. In this study, we tested the effects of selinexor alone or in combination with bortezomib in normoxia and hypoxia on MM cell survival and apoptosis in vitro and in vivo. In vitro, selinexor alone decreased survival and increased apoptosis, resensitizing MM cells to bortezomib. In vivo, we examined the effects of selinexor alone on tumor initiation and tumor progression, as well as selinexor in combination with bortezomib, on tumor growth in a bortezomib-resistant MM xenograft mouse model. Selinexor, used as a single agent, delayed tumor initiation and tumor progression, prolonging mice survival. In bortezomib-resistant xenografts, selinexor overcame drug resistance, significantly decreasing tumor burden and extending mice survival when combined with bortezomib.
Collapse
Affiliation(s)
- Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, Saint Louis, MO 63108, USA
| | - Feda Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, Saint Louis, MO 63108, USA
| | - Pilar de la Puente
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, Saint Louis, MO 63108, USA
| | | | - Abdel Kareem Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, Saint Louis, MO 63108, USA.
| |
Collapse
|
66
|
Soung YH, Kashyap T, Nguyen T, Yadav G, Chang H, Landesman Y, Chung J. Selective Inhibitors of Nuclear Export (SINE) compounds block proliferation and migration of triple negative breast cancer cells by restoring expression of ARRDC3. Oncotarget 2017; 8:52935-52947. [PMID: 28881784 PMCID: PMC5581083 DOI: 10.18632/oncotarget.17987] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 05/06/2017] [Indexed: 11/25/2022] Open
Abstract
Arrestin-related domain-containing protein-3 (ARRDC3) is one of 6 mammalian arrestins, which suppresses metastasis by inducing degradation of phosphorylated β2-adrenergic receptor (β2 AR) and integrin β4 (ITG β4). Our previous studies demonstrated that expression of ARRDC3 is epigentically silenced in Triple Negative Breast Cancer (TNBC) cells, and the forced expression of ARRDC3 significantly reduced the invasive potential of TNBC cells. In the current study, we found that Selective Inhibitors of Nuclear Export (SINE) compounds (KPT-185 and selinexor (KPT-330)) restore ARRDC3 expression in TNBC cell lines (MDA-MB-231 and MDA-MB-468) at both the mRNA and protein level in a dose and time course dependent manner. SINE compounds inhibit the proliferation, pro-invasive migration and anchorage independent growth of the TNBC cells by restoring ARRDC3 expression. We found that ARRDC3 expression is lower in TNBC cell lines than those of luminal breast cancer cell lines, and inversely correlated with IC50s of selinexor. Analysis of tissue microarray confirmed that ARRDC3 expression in patient samples is significantly lower in the majority of TNBC tumors relative to normal tissue. In vivo, selinexor inhibited the tumor growth of MDA-MB-231 xenografts by nearly 100% compared with vehicle treated animals. Furthermore, immunohistochemical analysis of TNBC tumors from selinexor treated mice revealed increased ARRDC3 expression versus vehicle treated animals. Our results suggest that restoration of ARRDC3 expression is an important antineoplastic mechanism of SINE compounds in TNBC, and therefore selinexor could be an effective treatment option for breast tumors with down-regulated ARRDC3.
Collapse
Affiliation(s)
- Young Hwa Soung
- Department of Pathology, Stony Brook Medicine, Stony Brook, NY 11794, USA
| | | | - Thalia Nguyen
- University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Garima Yadav
- Department of Pathology, Stony Brook Medicine, Stony Brook, NY 11794, USA
| | - Hua Chang
- Karyopharm Therapeutics, Inc. Newton, MA 02459, USA
| | | | - Jun Chung
- Department of Pathology, Stony Brook Medicine, Stony Brook, NY 11794, USA
| |
Collapse
|
67
|
Time-lapse imaging of p65 and IκBα translocation kinetics following Ca 2+-induced neuronal injury reveals biphasic translocation kinetics in surviving neurons. Mol Cell Neurosci 2017; 80:148-158. [PMID: 28238890 DOI: 10.1016/j.mcn.2017.02.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 02/01/2017] [Accepted: 02/21/2017] [Indexed: 12/19/2022] Open
Abstract
The transcription factor nuclear factor-κB (NF-κB) regulates neuronal differentiation, plasticity and survival. It is well established that excitatory neurotransmitters such as glutamate control NF-κB activity. Glutamate receptor overactivation is also involved in ischemic- and seizure-induced neuronal injury and neurodegeneration. However, little is known at the single cell-level how NF-κB signaling relates to neuronal survival during excitotoxic injury. We found that silencing of p65/NF-κB delayed N-methyl-d-aspartate (NMDA)-induced excitotoxic injury in hippocampal neurons, suggesting a functional role of p65 in excitotoxicity. Time-lapse imaging of p65 and its inhibitor IκBα using GFP and Cerulean fusion proteins revealed specific patterns of excitotoxic NF-κB activation. Nuclear translocation of p65 began on average 8±3min following 15min of NMDA treatment and was observed in up to two thirds of hippocampal neurons. Nuclear translocation of IκBα preceded that of p65 suggesting independent translocation processes. In surviving neurons, the onset of p65 nuclear export correlated with mitochondrial membrane potential recovery. Dying neurons exhibited persistent nuclear accumulation of p65-eGFP until plasma membrane permeabilization. Our data demonstrate an important role for p65 activation kinetics in neuronal cell death decisions following excitotoxic injury.
Collapse
|
68
|
Horie K, Ma J, Umezawa K. Inhibition of canonical NF-κB nuclear localization by (-)-DHMEQ via impairment of DNA binding. Oncol Res 2017; 22:105-15. [PMID: 25706397 PMCID: PMC7838434 DOI: 10.3727/096504014x14146137738628] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
We previously discovered (−)-DHMEQ as a selective inhibitor of NF-κB, and it was shown to suppress many cancer and inflammation models in animals. (−)-DHMEQ directly binds to NF-κB components to inhibit DNA binding, and moreover, it often inhibits nuclear translocation of NF-κB. The mechanism of inhibiting nuclear translocation has been elucidated for RelB, a main noncanonical NF-κB component. However, it was not elucidated for p65, a main canonical NF-κB component. In the present research, we studied how (−)-DHMEQ inhibits nuclear localization of p65. First, (−)-DHMEQ inhibited p65 nuclear accumulation in adult T-cell leukemia MT-2 cells in which canonical p65 is constitutively activated. But there was no change in the stability and importin-α3 affinity of p65. Then, we prepared a p65 mutant protein with Arg35Ala and Tyr36Ala (AA) mutations having no DNA-binding ability in HeLa cells. The p65 AA mutant showed reduced nuclear localization without changing the stability and importin affinity. Taken together, the mechanism of inhibition is different between RelB and p65, and inhibition of p65 nuclear localization is likely to be due to the inhibition of DNA binding changing the equilibrium between the nuclear and cytoplasmic amounts of p65.
Collapse
Affiliation(s)
- Kana Horie
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | | | | |
Collapse
|
69
|
Tyler PM, Servos MM, de Vries RC, Klebanov B, Kashyap T, Sacham S, Landesman Y, Dougan M, Dougan SK. Clinical Dosing Regimen of Selinexor Maintains Normal Immune Homeostasis and T-cell Effector Function in Mice: Implications for Combination with Immunotherapy. Mol Cancer Ther 2017; 16:428-439. [PMID: 28148714 DOI: 10.1158/1535-7163.mct-16-0496] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/03/2016] [Accepted: 12/27/2016] [Indexed: 12/18/2022]
Abstract
Selinexor (KPT-330) is a first-in-class nuclear transport inhibitor currently in clinical trials as an anticancer agent. To determine how selinexor might affect antitumor immunity, we analyzed immune homeostasis in mice treated with selinexor and found disruptions in T-cell development, a progressive loss of CD8 T cells, and increases in inflammatory monocytes. Antibody production in response to immunization was mostly normal. Precursor populations in bone marrow and thymus were unaffected by selinexor, suggesting that normal immune homeostasis could recover. We found that a high dose of selinexor given once per week preserved nearly normal immune functioning, whereas a lower dose given 3 times per week did not restore immune homeostasis. Both naïve and effector CD8 T cells cultured in vitro showed impaired activation in the presence of selinexor. These experiments suggest that nuclear exportins are required for T-cell development and function. We determined the minimum concentration of selinexor required to block T-cell activation and showed that T-cell-inhibitory effects of selinexor occur at levels above 100 nmol/L, corresponding to the first 24 hours post-oral dosing. In a model of implantable melanoma, selinexor treatment at 10 mg/kg with a 4-day drug holiday led to intratumoral IFNγ+, granzyme B+ cytotoxic CD8 T cells that were comparable with vehicle-treated mice. Overall, selinexor treatment leads to transient inhibition of T-cell activation, but clinically relevant once and twice weekly dosing schedules that incorporate sufficient drug holidays allow for normal CD8 T-cell functioning and development of antitumor immunity. Mol Cancer Ther; 16(3); 428-39. ©2017 AACRSee related article by Farren et al., p. 417.
Collapse
Affiliation(s)
- Paul M Tyler
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Romy C de Vries
- Dana-Farber Cancer Institute, Boston, Massachusetts.,University of Amsterdam, Program in Biomedical Sciences, Amsterdam, the Netherlands
| | | | | | - Sharon Sacham
- Karyopharm Therapeutics, Inc., Newton, Massachusetts
| | | | | | | |
Collapse
|
70
|
Pan M, Zhang Q, Liu P, Huang J, Wang Y, Chen S. Inhibition of the nuclear export of p65 and IQCG in leukemogenesis by NUP98-IQCG. Front Med 2016; 10:410-419. [PMID: 27864780 DOI: 10.1007/s11684-016-0489-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 09/29/2016] [Indexed: 01/04/2023]
Abstract
NUP98 fuses with approximately 34 different partner genes via translocation in hematological malignancies. Transgenic or retrovirus-mediated bone marrow transplanted mouse models reveal the leukemogenesis of some NUP98-related fusion genes. We previously reported the fusion protein NUP98-IQ motif containing G (IQCG) in a myeloid/T lymphoid bi-phenoleukemia patient with t(3;11) and confirmed its leukemogenic ability. Herein, we demonstrated the association of NUP98-IQCG with CRM1, and found that NUP98-IQCG expression inhibits the CRM1-mediated nuclear export of p65 and enhances the transcriptional activity of nuclear factor-κB. Moreover, IQCG could be entrapped in the nucleus by NUP98-IQCG, and the fusion protein interacts with calmodulin via the IQ motif in a calcium-independent manner. Therefore, the inhibition of nuclear exports of p65 and IQCG might contribute to the leukemogenesis of NUP98-IQCG.
Collapse
Affiliation(s)
- Mengmeng Pan
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, 200025, China
| | - Qiyao Zhang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, 200025, China
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences and Graduate School, Chinese Academy of Sciences and SJTU School of Medicine, Shanghai, 200025, China
| | - Ping Liu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, 200025, China
| | - Jinyan Huang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, 200025, China
| | - Yueying Wang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, 200025, China.
| | - Saijuan Chen
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, 200025, China.
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences and Graduate School, Chinese Academy of Sciences and SJTU School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
71
|
Turner JG, Kashyap T, Dawson JL, Gomez J, Bauer AA, Grant S, Dai Y, Shain KH, Meads M, Landesman Y, Sullivan DM. XPO1 inhibitor combination therapy with bortezomib or carfilzomib induces nuclear localization of IκBα and overcomes acquired proteasome inhibitor resistance in human multiple myeloma. Oncotarget 2016; 7:78896-78909. [PMID: 27806331 PMCID: PMC5340237 DOI: 10.18632/oncotarget.12969] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/12/2016] [Indexed: 02/05/2023] Open
Abstract
Acquired proteasome-inhibitor (PI) resistance is a major obstacle in the treatment of multiple myeloma (MM). We investigated whether the clinical XPO1-inhibitor selinexor, when combined with bortezomib or carfilzomib, could overcome acquired resistance in MM. PI-resistant myeloma cell lines both in vitro and in vivo and refractory myeloma patient biopsies were treated with selinexor/bortezomib or carfilzomib and assayed for apoptosis. Mechanistic studies included NFκB pathway protein expression assays, immunofluorescence microscopy, ImageStream flow-cytometry, and proximity-ligation assays. IκBα knockdown and NFκB activity were measured in selinexor/bortezomib-treated MM cells. We found that selinexor restored sensitivity of PI-resistant MM to bortezomib and carfilzomib. Selinexor/bortezomib treatment inhibited PI-resistant MM tumor growth and increased survival in mice. Myeloma cells from PI-refractory MM patients were sensitized by selinexor to bortezomib and carfilzomib without affecting non-myeloma cells. Immunofluorescence microscopy, Western blot, and ImageStream analyses of MM cells showed increases in total and nuclear IκBα by selinexor/bortezomib. Proximity ligation found increased IκBα-NFκB complexes in treated MM cells. IκBα knockdown abrogated selinexor/bortezomib-induced cytotoxicity in MM cells. Selinexor/bortezomib treatment decreased NFκB transcriptional activity. Selinexor, when used with bortezomib or carfilzomib, has the potential to overcome PI drug resistance in MM. Sensitization may be due to inactivation of the NFκB pathway by IκBα.
Collapse
MESH Headings
- Active Transport, Cell Nucleus
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Apoptosis/drug effects
- Bortezomib/pharmacology
- Cell Line, Tumor
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cell Nucleus/pathology
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm/drug effects
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Hydrazines/pharmacology
- Karyopherins/antagonists & inhibitors
- Karyopherins/metabolism
- Mice, Inbred NOD
- Mice, SCID
- Multiple Myeloma/drug therapy
- Multiple Myeloma/enzymology
- Multiple Myeloma/genetics
- Multiple Myeloma/pathology
- NF-KappaB Inhibitor alpha/genetics
- NF-KappaB Inhibitor alpha/metabolism
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Oligopeptides/pharmacology
- Proteasome Endopeptidase Complex/metabolism
- Protein Stability
- Proteolysis
- RNA Interference
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Receptors, Cytoplasmic and Nuclear/metabolism
- Time Factors
- Transcription, Genetic
- Transfection
- Triazoles/pharmacology
- Xenograft Model Antitumor Assays
- Exportin 1 Protein
Collapse
Affiliation(s)
- Joel G. Turner
- Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | - Jana L. Dawson
- Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Juan Gomez
- Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Alexis A. Bauer
- Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Steven Grant
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Yun Dai
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Kenneth H. Shain
- Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Mark Meads
- Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | - Daniel M. Sullivan
- Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Blood & Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
72
|
Aggarwal N, Korenbaum E, Mahadeva R, Immenschuh S, Grau V, Dinarello CA, Welte T, Janciauskiene S. α-Linoleic acid enhances the capacity of α-1 antitrypsin to inhibit lipopolysaccharide induced IL-1β in human blood neutrophils. Mol Med 2016; 22:680-693. [PMID: 27452044 DOI: 10.2119/molmed.2016.00119] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/05/2016] [Indexed: 11/06/2022] Open
Abstract
Alpha1-antitrypsin (A1AT, SERPINA1), a major circulating inhibitor of neutrophil elastase (NE) and proteinase-3 (PR3), has been proposed to reduce the processing and release of IL-1β. Since the anti-inflammatory properties of A1AT are influenced by the presence of polyunsaturated fatty acids, we compared effects of fatty acid-free (A1AT-0) and α-linoleic acid bound (A1AT-LA) forms of A1AT on lipopolysaccharide (LPS)-induced synthesis of IL-1β precursor and the release of IL-1β from human blood neutrophils. The presence of A1AT-LA or A1AT-0 significantly reduced LPS induced release of mature IL-1β. However, only A1AT-LA reduced both steady state mRNA levels of IL-1β and the secretion of mature IL-1β. In LPS-stimulated neutrophils, mRNA levels of TLR2/4, NFKBIA, P2RX7, NLRP3, and CASP1 decreased significantly in the presence of A1AT-LA but not A1AT-0. A1AT-0 and A1AT-LA did not inhibit the direct enzymatic activity of caspase-1, but we observed complexes of either form of A1AT with NE and PR3. Consistent with the effect on TLR and IL-1β gene expression, only A1AT-LA inhibited LPS-induced gene expression of NE and PR3. Increased gene expression of PPAR-γ was observed in A1AT-LA treated neutrophils without of LPS stimulation, and the selective PPAR-γ antagonist (GW9662) prevented the reduction in IL-1β by A1AT-LA. We conclude from our data, that the ability of A1AT to reduce TLR and IL-1β gene expression depends on its association with LA. Moreover, the anti-inflammatory properties of A1AT-LA are likely to be mediated by the activation of PPAR-γ.
Collapse
Affiliation(s)
- Nupur Aggarwal
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Elena Korenbaum
- Institute of Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Ravi Mahadeva
- Cambridge NIHR Biomedical Research Centre, Department of Respiratory Medicine, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Stephan Immenschuh
- Institute for Transfusion Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Veronika Grau
- Department of General and Thoracic Surgery, Laboratory of Experimental Surgery, Justus-Liebig-University Giessen, Giessen, Germany
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045.,Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tobias Welte
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Sabina Janciauskiene
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| |
Collapse
|
73
|
Sun Q, Chen X, Zhou Q, Burstein E, Yang S, Jia D. Inhibiting cancer cell hallmark features through nuclear export inhibition. Signal Transduct Target Ther 2016; 1:16010. [PMID: 29263896 PMCID: PMC5661660 DOI: 10.1038/sigtrans.2016.10] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/28/2016] [Accepted: 05/31/2016] [Indexed: 02/05/2023] Open
Abstract
Treating cancer through inhibition of nuclear export is one of the best examples of basic research translation into clinical application. Nuclear export factor chromosomal region maintenance 1 (CRM1; Xpo1 and exportin-1) controls cellular localization and function of numerous proteins that are critical for the development of many cancer hallmarks. The diverse actions of CRM1 are likely to explain the broad ranging anti-cancer potency of CRM1 inhibitors observed in pre-clinical studies and/or clinical trials (phase I–III) on both advanced-stage solid and hematological tumors. In this review, we compare and contrast the mechanisms of action of different CRM1 inhibitors, and discuss the potential benefit of unexplored non-covalent CRM1 inhibitors. This emerging field has uncovered that nuclear export inhibition is well poised as an attractive target towards low-toxicity broad-spectrum potent anti-cancer therapy.
Collapse
Affiliation(s)
- Qingxiang Sun
- State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China.,Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Xueqin Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiao Zhou
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Ezra Burstein
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA.,Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Shengyong Yang
- State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Da Jia
- State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China.,West China 2nd University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
74
|
Di Ventura B, Kuhlman B. Go in! Go out! Inducible control of nuclear localization. Curr Opin Chem Biol 2016; 34:62-71. [PMID: 27372352 DOI: 10.1016/j.cbpa.2016.06.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/06/2016] [Accepted: 06/07/2016] [Indexed: 12/19/2022]
Abstract
Cells have evolved a variety of mechanisms to regulate the enormous complexity of processes taking place inside them. One mechanism consists in tightly controlling the localization of macromolecules, keeping them away from their place of action until needed. Since a large fraction of the cellular response to external stimuli is mediated by gene expression, it is not surprising that transcriptional regulators are often subject to stimulus-induced nuclear import or export. Here we review recent methods in chemical biology and optogenetics for controlling the nuclear localization of proteins of interest inside living cells. These methods allow researchers to regulate protein activity with exquisite spatiotemporal control, and open up new possibilities for studying the roles of proteins in a broad array of cellular processes and biological functions.
Collapse
Affiliation(s)
- Barbara Di Ventura
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), University of Heidelberg, Heidelberg, Germany.
| | - Brian Kuhlman
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
75
|
Leukemia-Associated Nup214 Fusion Proteins Disturb the XPO1-Mediated Nuclear-Cytoplasmic Transport Pathway and Thereby the NF-κB Signaling Pathway. Mol Cell Biol 2016; 36:1820-35. [PMID: 27114368 DOI: 10.1128/mcb.00158-16] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 04/14/2016] [Indexed: 02/07/2023] Open
Abstract
Nuclear-cytoplasmic transport through nuclear pore complexes is mediated by nuclear transport receptors. Previous reports have suggested that aberrant nuclear-cytoplasmic transport due to mutations or overexpression of nuclear pore complexes and nuclear transport receptors is closely linked to diseases. Nup214, a component of nuclear pore complexes, has been found as chimeric fusion proteins in leukemia. Among various Nup214 fusion proteins, SET-Nup214 and DEK-Nup214 have been shown to be engaged in tumorigenesis, but their oncogenic mechanisms remain unclear. In this study, we examined the functions of the Nup214 fusion proteins by focusing on their effects on nuclear-cytoplasmic transport. We found that SET-Nup214 and DEK-Nup214 interact with exportin-1 (XPO1)/CRM1 and nuclear RNA export factor 1 (NXF1)/TAP, which mediate leucine-rich nuclear export signal (NES)-dependent protein export and mRNA export, respectively. SET-Nup214 and DEK-Nup214 decreased the XPO1-mediated nuclear export of NES proteins such as cyclin B and proteins involved in the NF-κB signaling pathway by tethering XPO1 onto nuclear dots where Nup214 fusion proteins are localized. We also demonstrated that SET-Nup214 and DEK-Nup214 expression inhibited NF-κB-mediated transcription by abnormal tethering of the complex containing p65 and its inhibitor, IκB, in the nucleus. These results suggest that SET-Nup214 and DEK-Nup214 perturb the regulation of gene expression through alteration of the nuclear-cytoplasmic transport system.
Collapse
|
76
|
The Ubiquitination of NF-κB Subunits in the Control of Transcription. Cells 2016; 5:cells5020023. [PMID: 27187478 PMCID: PMC4931672 DOI: 10.3390/cells5020023] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/03/2016] [Accepted: 05/06/2016] [Indexed: 02/06/2023] Open
Abstract
Nuclear factor (NF)-κB has evolved as a latent, inducible family of transcription factors fundamental in the control of the inflammatory response. The transcription of hundreds of genes involved in inflammation and immune homeostasis require NF-κB, necessitating the need for its strict control. The inducible ubiquitination and proteasomal degradation of the cytoplasmic inhibitor of κB (IκB) proteins promotes the nuclear translocation and transcriptional activity of NF-κB. More recently, an additional role for ubiquitination in the regulation of NF-κB activity has been identified. In this case, the ubiquitination and degradation of the NF-κB subunits themselves plays a critical role in the termination of NF-κB activity and the associated transcriptional response. While there is still much to discover, a number of NF-κB ubiquitin ligases and deubiquitinases have now been identified which coordinate to regulate the NF-κB transcriptional response. This review will focus the regulation of NF-κB subunits by ubiquitination, the key regulatory components and their impact on NF-κB directed transcription.
Collapse
|
77
|
Fagerlund R, Behar M, Fortmann KT, Lin YE, Vargas JD, Hoffmann A. Anatomy of a negative feedback loop: the case of IκBα. J R Soc Interface 2016; 12:0262. [PMID: 26311312 DOI: 10.1098/rsif.2015.0262] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The magnitude, duration and oscillation of cellular signalling pathway responses are often limited by negative feedback loops, defined as an 'activator-induced inhibitor' regulatory motif. Within the NFκB signalling pathway, a key negative feedback regulator is IκBα. We show here that, contrary to current understanding, NFκB-inducible expression is not sufficient for providing effective negative feedback. We then employ computational simulations of NFκB signalling to identify IκBα molecular properties that are critical for proper negative feedback control and test the resulting predictions in biochemical and single-cell live-imaging studies. We identified nuclear import and nuclear export of IκBα and the IκBα-NFκB complex, as well as the free IκBα half-life, as key determinants of post-induction repression of NFκB and the potential for subsequent reactivation. Our work emphasizes that negative feedback is an emergent systems property determined by multiple molecular and biophysical properties in addition to the required 'activator-induced inhibitor' relationship.
Collapse
Affiliation(s)
- Riku Fagerlund
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Marcelo Behar
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Karen T Fortmann
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Y Eason Lin
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095, USA Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, CA 90095, USA
| | - Jesse D Vargas
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095, USA Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, CA 90095, USA
| | - Alexander Hoffmann
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095, USA Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
78
|
Rinkenbaugh AL, Baldwin AS. The NF-κB Pathway and Cancer Stem Cells. Cells 2016; 5:cells5020016. [PMID: 27058560 PMCID: PMC4931665 DOI: 10.3390/cells5020016] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 02/07/2023] Open
Abstract
The NF-κB transcription factor pathway is a crucial regulator of inflammation and immune responses. Additionally, aberrant NF-κB signaling has been identified in many types of cancer. Downstream of key oncogenic pathways, such as RAS, BCR-ABL, and Her2, NF-κB regulates transcription of target genes that promote cell survival and proliferation, inhibit apoptosis, and mediate invasion and metastasis. The cancer stem cell model posits that a subset of tumor cells (cancer stem cells) drive tumor initiation, exhibit resistance to treatment, and promote recurrence and metastasis. This review examines the evidence for a role for NF-κB signaling in cancer stem cell biology.
Collapse
Affiliation(s)
- Amanda L Rinkenbaugh
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Albert S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
79
|
Asymmetric arginine dimethylation of RelA provides a repressive mark to modulate TNFα/NF-κB response. Proc Natl Acad Sci U S A 2016; 113:4326-31. [PMID: 27051065 DOI: 10.1073/pnas.1522372113] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Nuclear factor kappa B (NF-κB) is an inducible transcription factor that plays critical roles in immune and stress responses and is often implicated in pathologies, including chronic inflammation and cancer. Although much has been learned about NF-κB-activating pathways, the specific repression of NF-κB is far less well understood. Here we identified the type I protein arginine methyltransferase 1 (PRMT1) as a restrictive factor controlling TNFα-induced activation of NF-κB. PRMT1 forms a cellular complex with NF-κB through direct interaction with the Rel homology domain of RelA. We demonstrate that PRMT1 methylates RelA at evolutionary conserved R30, located in the DNA-binding L1 loop, which is a critical residue required for DNA binding. Asymmetric R30 dimethylation inhibits the binding of RelA to DNA and represses NF-κB target genes in response to TNFα. Molecular dynamics simulations of the DNA-bound RelA:p50 predicted structural changes in RelA caused by R30 methylation or a mutation that interferes with the stability of the DNA-NF-κB complex. Our findings provide evidence for the asymmetric arginine dimethylation of RelA and unveil a unique mechanism controlling TNFα/NF-κB signaling.
Collapse
|
80
|
Senol SP, Temiz M, Guden DS, Cecen P, Sari AN, Sahan-Firat S, Falck JR, Dakarapu R, Malik KU, Tunctan B. Contribution of PPARα/β/γ, AP-1, importin-α3, and RXRα to the protective effect of 5,14-HEDGE, a 20-HETE mimetic, against hypotension, tachycardia, and inflammation in a rat model of septic shock. Inflamm Res 2016; 65:367-87. [DOI: 10.1007/s00011-016-0922-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 10/10/2015] [Accepted: 01/29/2016] [Indexed: 12/18/2022] Open
|
81
|
Niopek D, Wehler P, Roensch J, Eils R, Di Ventura B. Optogenetic control of nuclear protein export. Nat Commun 2016; 7:10624. [PMID: 26853913 PMCID: PMC4748110 DOI: 10.1038/ncomms10624] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/05/2016] [Indexed: 12/21/2022] Open
Abstract
Active nucleocytoplasmic transport is a key mechanism underlying protein regulation in eukaryotes. While nuclear protein import can be controlled in space and time with a portfolio of optogenetic tools, protein export has not been tackled so far. Here we present a light-inducible nuclear export system (LEXY) based on a single, genetically encoded tag, which enables precise spatiotemporal control over the export of tagged proteins. A constitutively nuclear, chromatin-anchored LEXY variant expands the method towards light inhibition of endogenous protein export by sequestering cellular CRM1 receptors. We showcase the utility of LEXY for cell biology applications by regulating a synthetic repressor as well as human p53 transcriptional activity with light. LEXY is a powerful addition to the optogenetic toolbox, allowing various novel applications in synthetic and cell biology.
Collapse
Affiliation(s)
- Dominik Niopek
- Department of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Department of Bioinformatics and Functional Genomics, Synthetic Biology Group, Institute for Pharmacy and Biotechnology (IPMB), University of Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Pierre Wehler
- Department of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Julia Roensch
- Department of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Roland Eils
- Department of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Department of Bioinformatics and Functional Genomics, Synthetic Biology Group, Institute for Pharmacy and Biotechnology (IPMB), University of Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Barbara Di Ventura
- Department of Bioinformatics and Functional Genomics, Synthetic Biology Group, Institute for Pharmacy and Biotechnology (IPMB), University of Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| |
Collapse
|
82
|
Crochiere M, Kashyap T, Kalid O, Shechter S, Klebanov B, Senapedis W, Saint-Martin JR, Landesman Y. Deciphering mechanisms of drug sensitivity and resistance to Selective Inhibitor of Nuclear Export (SINE) compounds. BMC Cancer 2015; 15:910. [PMID: 26573568 PMCID: PMC4647283 DOI: 10.1186/s12885-015-1790-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 10/15/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Exportin 1 (XPO1) is a well-characterized nuclear export protein whose expression is up-regulated in many types of cancers and functions to transport key tumor suppressor proteins (TSPs) from the nucleus. Karyopharm Therapeutics has developed a series of small-molecule Selective Inhibitor of Nuclear Export (SINE) compounds, which have been shown to block XPO1 function both in vitro and in vivo. The drug candidate, selinexor (KPT-330), is currently in Phase-II/IIb clinical trials for treatment of both hematologic and solid tumors. The present study sought to decipher the mechanisms that render cells either sensitive or resistant to treatment with SINE compounds, represented by KPT-185, an early analogue of KPT-330. METHODS Using the human fibrosarcoma HT1080 cell line, resistance to SINE was acquired over a period of 10 months of constant incubation with increasing concentration of KPT-185. Cell viability was assayed by MTT. Immunofluorescence was used to compare nuclear export of TSPs. Fluorescence activated cell sorting (FACS), quantitative polymerase chain reaction (qPCR), and immunoblots were used to measure effects on cell cycle, gene expression, and cell death. RNA from naïve and drug treated parental and resistant cells was analyzed by Affymetrix microarrays. RESULTS Treatment of HT1080 cells with gradually increasing concentrations of SINE resulted in >100 fold decrease in sensitivity to SINE cytotoxicity. Resistant cells displayed prolonged cell cycle, reduced nuclear accumulation of TSPs, and similar changes in protein expression compared to parental cells, however the magnitude of the protein expression changes were more significant in parental cells. Microarray analyses comparing parental to resistant cells indicate that a number of key signaling pathways were altered in resistant cells including expression changes in genes involved in adhesion, apoptosis, and inflammation. While the patterns of changes in transcription following drug treatment are similar in parental and resistant cells, the extent of response was more robust in the parental cells. CONCLUSIONS These results suggest that SINE resistance is conferred by alterations in signaling pathways downstream of XPO1 inhibition. Modulation of these pathways could potentially overcome the resistance to nuclear export inhibitors.
Collapse
Affiliation(s)
- Marsha Crochiere
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | - Trinayan Kashyap
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | - Ori Kalid
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | - Sharon Shechter
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | - Boris Klebanov
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | - William Senapedis
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | | | - Yosef Landesman
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| |
Collapse
|
83
|
Zhu J, Cynader MS, Jia W. TDP-43 Inhibits NF-κB Activity by Blocking p65 Nuclear Translocation. PLoS One 2015; 10:e0142296. [PMID: 26571498 PMCID: PMC4646651 DOI: 10.1371/journal.pone.0142296] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 10/20/2015] [Indexed: 12/22/2022] Open
Abstract
TDP-43 (TAR DNA binding protein 43) is a heterogeneous nuclear ribonucleoprotein (hnRNP) that has been found to play an important role in neurodegenerative diseases. TDP-43’s involvement in nuclear factor-kappaB pathways has been reported in both neurons and microglial cells. The NF-κB pathway targets hundreds of genes, many of which are involved in inflammation, immunity and cancer. p50/p65 (p50/RelA) heterodimers, as the major Rel complex in the NF-κB family, are induced by diverse external physiological stimuli and modulate transcriptional activity in almost all cell types. Both p65 and TDP-43 translocation occur through the classic nuclear transportation system. In this study, we report that TDP-43 overexpression prevents TNF-α induced p65 nuclear translocation in a dose dependent manner, and that this further inhibits p65 transactivation activity. The inhibition by TDP-43 does not occur through preventing IκB degradation but probably by competing for the nuclear transporter-importin α3 (KPNA4). This competition is dependent on the presence of the nuclear localization signal (NLS) in TDP-43. Silencing TDP-43 using a specific siRNA also increased p65 nuclear localization upon TNF-α stimulation, suggesting that endogenous TDP-43 may be a default suppressor of the NF-κB pathway. Our results indicate that TDP-43 may play an important role in regulating the levels of NF-κB activity by controlling the nuclear translocation of p65.
Collapse
Affiliation(s)
- Jingyan Zhu
- Brain Research Center, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T2B5, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Max S. Cynader
- Brain Research Center, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T2B5, Canada
| | - William Jia
- Brain Research Center, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T2B5, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- * E-mail:
| |
Collapse
|
84
|
Walther W, Kobelt D, Bauer L, Aumann J, Stein U. Chemosensitization by diverging modulation by short-term and long-term TNF-α action on ABCB1 expression and NF-κB signaling in colon cancer. Int J Oncol 2015; 47:2276-85. [PMID: 26463665 DOI: 10.3892/ijo.2015.3189] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 08/19/2015] [Indexed: 11/05/2022] Open
Abstract
Multidrug resistance (MDR) is a major cause for cancer chemotherapy failure. Among the numerous strategies to overcome persistent action of proinflammatory cytokines, such as tumor necrosis factor α (TNF-α) permits downregulation of MDR-associated genes, including ATP-binding cassette, subfamily B 1 gene (ABCB1). A key regulator of ABCB1 expression is the transcription factor nuclear factor κ light chain enhancer (NF-κB)/p65. We analyzed diverging short- and long-term effects of TNF-α regarding modulation of NF-κB/p65 signaling and ABCB1 expression in colon cancer cells. Highly resistant ABCB1 overexpressing human HCT15 colorectal carcinoma cells were subjected to short- (30-120 min) or long-term (24-96 h) TNF-α treatment. TNF-α mediated modulation of ABCB1 expression was analyzed by real-time RT-PCR and western blot analysis. The TNF-mediated chemosensitization was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) cytotoxicity assay. The involvement of TNF receptors and of NF-κB/p65 signaling was analyzed by western blot analysis, ABCB1 promoter analysis and electrophoretic mobility shift assay (EMSA). The study revealed, that long-term, but not short-term TNF-α treatment leads to TNF-receptor 1 (TNFR1) mediated downregulation of ABCB1 resulting in sensitization towards drug treatment. It dampens NF-κB/p65 activation and nuclear factor of κ light polypeptide gene enhancer in B-cells inhibitor α (IκBα) resynthesis, associated with reduced nuclear accumulation of NF-κB/p65 and reduced binding to its consensus sequence in the ABCB1 promoter. The study reveals the diverging effects of short- or long-term TNF-α action and provides novel insights on downregulation of ABCB1 expression by TNF-mediated repression of NF-κB signaling.
Collapse
Affiliation(s)
- Wolfgang Walther
- Experimental and Clinical Research Center, Charité, University Medicine Berlin and the Max-Delbrück-Center for Molecular Medicine, D-13125 Berlin, Germany
| | - Dennis Kobelt
- Max-Delbrück-Center for Molecular Medicine, D-13125 Berlin, Germany
| | - Lisa Bauer
- Max-Delbrück-Center for Molecular Medicine, D-13125 Berlin, Germany
| | - Jutta Aumann
- Experimental and Clinical Research Center, Charité, University Medicine Berlin and the Max-Delbrück-Center for Molecular Medicine, D-13125 Berlin, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité, University Medicine Berlin and the Max-Delbrück-Center for Molecular Medicine, D-13125 Berlin, Germany
| |
Collapse
|
85
|
McKenna S, Gossling M, Bugarini A, Hill E, Anderson AL, Rancourt RC, Balasubramaniyan N, El Kasmi KC, Wright CJ. Endotoxemia Induces IκBβ/NF-κB-Dependent Endothelin-1 Expression in Hepatic Macrophages. THE JOURNAL OF IMMUNOLOGY 2015; 195:3866-79. [PMID: 26342031 DOI: 10.4049/jimmunol.1501017] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 08/03/2015] [Indexed: 12/20/2022]
Abstract
Elevated serum concentrations of the vasoactive protein endothelin-1 (ET-1) occur in the setting of systemic inflammatory response syndrome and contribute to distal organ hypoperfusion and pulmonary hypertension. Thus, understanding the cellular source and transcriptional regulation of systemic inflammatory stress-induced ET-1 expression may reveal therapeutic targets. Using a murine model of LPS-induced septic shock, we demonstrate that the hepatic macrophage is the primary source of elevated circulating ET-1, rather than the endothelium as previously proposed. Using pharmacologic inhibitors, ET-1 promoter luciferase assays, and by silencing and overexpressing NF-κB inhibitory protein IκB expression, we demonstrate that LPS-induced ET-1 expression occurs via an NF-κB-dependent pathway. Finally, the specific role of the cRel/p65 inhibitory protein IκBβ was evaluated. Although cytoplasmic IκBβ inhibits activity of cRel-containing NF-κB dimers, nuclear IκBβ stabilizes NF-κB/DNA binding and enhances gene expression. Using targeted pharmacologic therapies to specifically prevent IκBβ/NF-κB signaling, as well as mice genetically modified to overexpress IκBβ, we show that nuclear IκBβ is both necessary and sufficient to drive LPS-induced ET-1 expression. Together, these results mechanistically link the innate immune response mediated by IκBβ/NF-κB to ET-1 expression and potentially reveal therapeutic targets for patients with Gram-negative septic shock.
Collapse
Affiliation(s)
- Sarah McKenna
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045
| | - Megan Gossling
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045
| | - Alejandro Bugarini
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045
| | - Elizabeth Hill
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045
| | - Aimee L Anderson
- Hepatology and Nutrition, Digestive Health Institute, Section of Gastroenterology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045; and
| | - Raymond C Rancourt
- Pediatric Airway Research Center, Section of Pulmonology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045
| | - Natarajan Balasubramaniyan
- Hepatology and Nutrition, Digestive Health Institute, Section of Gastroenterology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045; and
| | - Karim C El Kasmi
- Hepatology and Nutrition, Digestive Health Institute, Section of Gastroenterology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045; and
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045;
| |
Collapse
|
86
|
Xing S, Zhang B, Hua R, Tai WCS, Zeng Z, Xie B, Huang C, Xue J, Xiong S, Yang J, Liu S, Li H. URG4/URGCP enhances the angiogenic capacity of human hepatocellular carcinoma cells in vitro via activation of the NF-κB signaling pathway. BMC Cancer 2015; 15:368. [PMID: 25947641 PMCID: PMC4437676 DOI: 10.1186/s12885-015-1378-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 04/27/2015] [Indexed: 01/12/2023] Open
Abstract
Background Angiogenesis is essential for tumor growth. Hepatocellular carcinoma (HCC) is characterized by hypervascularity; high levels of angiogenesis are associated with poor prognosis and a highly invasive phenotype in HCC. Up-regulated gene-4 (URG4), also known as upregulator of cell proliferation (URGCP), is overexpressed in multiple tumor types and has been suggested to act as an oncogene. This study aimed to elucidate the effect of URG4/URGCP on the angiogenic capacity of HCC cells in vitro. Methods Expression of URG4/URGCP in HCC cell lines and normal liver epithelial cell lines was examined by Western blotting and quantitative real-time PCR. URG4/URGCP was stably overexpressed or transiently knocked down using a shRNA in two HCC cell lines. The human umbilical vein endothelial cell (HUVEC) tubule formation and Transwell migration assays and chicken chorioallantoic membrane (CAM) assay were used to examine the angiogenic capacity of conditioned media from URG4/URGCP-overexpressing and knockdown cells. A luciferase reporter assay was used to examine the transcriptional activity of nuclear factor kappa – light – chain - enhancer of activated B cells (NF-κB). NF-κB was inhibited by overexpressing degradation-resistant mutant inhibitor of κB (IκB)-α. Expression of vascular endothelial growth factor C (VEGFC), tumor necrosis factor-α (TNFα), interleukin (IL)-6, IL-8 and v-myc avian myelocytomatosis viral oncogene homolog (MYC) were examined by quantitative real-time PCR; VEGFC protein expression was analyzed using an ELISA. Results URG4/URGCP protein and mRNA expression were significantly upregulated in HCC cell lines. Overexpressing URG4/URGCP enhanced - while silencing URG4/URGCP decreased - the capacity of HCC cell conditioned media to induce HUVEC tubule formation and migration and neovascularization in the CAM assay. Furthermore, overexpressing URG4/URGCP increased - whereas knockdown of URG4/URGCP decreased - VEGFC expression, NF-κB transcriptional activity, the levels of phosphorylated (but not total) IκB kinase (IKK) and IκB-α, and expression of TNFα, IL-6, IL-8 and MYC in HCC cells. Additionally, inhibition of NF-κB activity in HCC cells abrogated URG4/URGCP-induced NF-κB activation and angiogenic capacity. Conclusions This study suggests that URG4/URGCP plays an important pro-angiogenic role in HCC via a mechanism linked to activation of the NF-κB pathway; URG4/URGCP may represent a potential target for anti-angiogenic therapy in HCC. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1378-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sizhong Xing
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, P.R. China. .,Department of Gastroenterology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, P.R. China. .,Department of Internal Medicine, Baoan People's Hospital, Shenzhen, 518101, P.R. China.
| | - Bing Zhang
- Department of Medical Imaging, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, P.R. China.
| | - Ruixi Hua
- Department of Oncology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, P.R. China.
| | - William Chi-shing Tai
- Center for Cancer and Inflammation Research, Institute of Integrated Bioinformedicine and Translational Science, Hong Kong Baptist University, Hong Kong, S.A.R., China.
| | - Zhirong Zeng
- Department of Gastroenterology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, P.R. China.
| | - Binhui Xie
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, P.R. China.
| | - Chenghui Huang
- Department of Internal Medicine, Baoan People's Hospital, Shenzhen, 518101, P.R. China.
| | - Jisu Xue
- Department of Internal Medicine, Baoan People's Hospital, Shenzhen, 518101, P.R. China.
| | - Shiqiu Xiong
- Department of Biochemistry, University of Leicester, Leicester, UK.
| | - Jianyong Yang
- Department of Medical Imaging, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, P.R. China.
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, P.R. China.
| | - Heping Li
- Department of Medical Imaging, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, P.R. China. .,Department of Oncology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, P.R. China.
| |
Collapse
|
87
|
Ch'ng WC, Abd-Aziz N, Ong MH, Stanbridge EJ, Shafee N. Human renal carcinoma cells respond to Newcastle disease virus infection through activation of the p38 MAPK/NF-κB/IκBα pathway. Cell Oncol (Dordr) 2015; 38:279-88. [PMID: 25930675 DOI: 10.1007/s13402-015-0229-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2015] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Newcastle disease virus (NDV) is an oncolytic virus that is known to have a higher preference to cancer cells than to normal cells. It has been proposed that this higher preference may be due to defects in the interferon (IFN) responses of cancer cells. The exact mechanism underlying this process, however, remains to be resolved. In the present study, we examined the antiviral response towards NDV infection of clear cell renal cell carcinoma (ccRCC) cells. ccRCC is associated with mutations of the von Hippel-Lindau tumor suppressor gene VHL, whose protein product is important for eliciting cellular responses to changes in oxygen levels. The most common first line treatment strategy of ccRCC includes IFN. Unfortunately, most ccRCC cases are diagnosed at a late stage and often are resistant to IFN-based therapies. Alternative treatment approaches, including virotherapy using oncolytic viruses, are currently being investigated. The present study was designed to investigate the mechanistic pathways underlying the response of ccRCC cells to oncolytic NDV infection. METHODS AND RESULTS We found that NDV induces activation of NF-κB in ccRCC cells by inducing phosphorylation and subsequent degradation of IκBα. IκBα was found to be phosphorylated as early as 1 hour post-infection and to result in rapid NF-κB nuclear translocation and activation. Importantly, p38 MAPK phosphorylation was found to occur upstream of the NDV-induced NF-κB activation. Restoration of VHL in ccRCC cells did not result in a reduction of this phosphorylation. A similar phenomenon was also observed in several other cancer-derived cell lines. CONCLUSION Our data provide evidence for involvement of the p38 MAPK/NF-κB/IκBα pathway in NDV infection and subsequent induction of apoptosis in ccRCC cells.
Collapse
Affiliation(s)
- Wei-Choong Ch'ng
- Department of Microbiology, Faculty of Biotechnology & Biomolecular Sciences, and Institute of Biosciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Malaysia
| | | | | | | | | |
Collapse
|
88
|
Chen Z, He Y, Song C, Dong Z, Su Z, Xue J. Sericin can reduce hippocampal neuronal apoptosis by activating the Akt signal transduction pathway in a rat model of diabetes mellitus. Neural Regen Res 2015; 7:197-201. [PMID: 25767499 PMCID: PMC4353114 DOI: 10.3969/j.issn.1673-5374.2012.03.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 11/15/2011] [Indexed: 11/18/2022] Open
Abstract
In the present study, a rat model of type 2 diabetes mellitus was established by continuous peritoneal injection of streptozotocin. Following intragastric perfusion of sericin for 35 days, blood glucose levels significantly reduced, neuronal apoptosis in the hippocampal CA1 region decreased, hippocampal phosphorylated Akt and nuclear factor kappa B expression were enhanced, but Bcl-xL/Bcl-2 associated death promoter expression decreased. Results demonstrated that sericin can reduce hippocampal neuronal apoptosis in a rat model of diabetes mellitus by regulating abnormal changes in the Akt signal transduction pathway.
Collapse
Affiliation(s)
- Zhihong Chen
- Department of Human Anatomy, Chengde Medical College, Chengde 067000, Hebei Province, China
| | - Yaqiang He
- Department of Human Anatomy, Chengde Medical College, Chengde 067000, Hebei Province, China
| | - Chengjun Song
- Department of Human Anatomy, Chengde Medical College, Chengde 067000, Hebei Province, China
| | - Zhijun Dong
- Affiliated Hospital of Chengde Medical College, Chengde 067000, Hebei Province, China
| | - Zhejun Su
- Affiliated Hospital of Chengde Medical College, Chengde 067000, Hebei Province, China
| | - Jingfeng Xue
- Department of Human Anatomy, Chengde Medical College, Chengde 067000, Hebei Province, China
| |
Collapse
|
89
|
Li X, Zhang C, Qiao W, Zhou X, Sun M. NFKB1 -94ins/del ATTG polymorphism increases osteosarcoma risk in a Chinese Han population. Int J Clin Exp Med 2015; 8:1420-1423. [PMID: 25785149 PMCID: PMC4358604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 12/15/2014] [Indexed: 06/04/2023]
Abstract
Osteosarcoma is one of the most common bone malignancies. The Nuclear factor-κB1 (NFKB1) gene plays an important role in the pathogenesis of osteosarcoma. The objective of this study aimed to detect the potential association between NFKB1 -94 ins/del ATTG polymorphism and osteosarcoma susceptibility in Chinese Han population. We recruited 220 osteosarcoma patients and 222 cancer-free controls in this case-control study. The NFKB1 -94 ins/del ATTG polymorphism by the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. Patients with ID genotype and II genotype showed higher risk of osteosarcoma than individuals with DD genotype (OR=1.54, 95% CI 1.00-2.44, P=0.05; OR=1.72, 95% CI 1.01-2.93, P=0.04), respectively. Subjects with ID or II genotype also showed increased risk of osteosarcoma (OR=1.60, 95% CI 1.04-2.47, P=0.03). In addition, I allele was significantly associated with osteosarcoma risk (OR=1.31, 95% CI 1.01-1.71, P=0.04). We also found that this polymorphism was significantly associated with advanced osteosarcoma risk (OR=3.43, 95% CI 1.61-7.36, P=0.001) and metastatic osteosarcoma risk (OR=2.33, 95% CI 1.22-5.03, P=0.01). In conclusion, our findings indicate that osteosarcoma is associated with the NFKB1 promoter -94ins/del ATTG polymorphism.
Collapse
Affiliation(s)
- Xiyi Li
- Department of Pathology, Cancer Hospital, Fudan UniversityShanghai 200032, China
- Department of Oncology, Shanghai Medical School, Fudan UniversityShanghai 200032, China
- Department of Orthopedics, Changzheng Hospital, Second Military Medical UniversityShanghai 200003, China
| | - Chenglin Zhang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical UniversityShanghai 200003, China
| | - Wei Qiao
- Department of Pathology, Cancer Hospital, Fudan UniversityShanghai 200032, China
- Department of Oncology, Shanghai Medical School, Fudan UniversityShanghai 200032, China
| | - Xuhui Zhou
- Department of Orthopedics, Changzheng Hospital, Second Military Medical UniversityShanghai 200003, China
| | - Menghong Sun
- Department of Pathology, Cancer Hospital, Fudan UniversityShanghai 200032, China
- Department of Oncology, Shanghai Medical School, Fudan UniversityShanghai 200032, China
| |
Collapse
|
90
|
Han X, Wang J, Shen Y, Zhang N, Wang S, Yao J, Shi Y. CRM1 as a new therapeutic target for non-Hodgkin lymphoma. Leuk Res 2015; 39:38-46. [PMID: 25466285 DOI: 10.1016/j.leukres.2014.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 08/13/2014] [Accepted: 10/09/2014] [Indexed: 11/30/2022]
Abstract
The chromosomal region maintenance 1 (CRM1) may serve as a novel target for cancer treatment. Here, we investigated the anti non-Hodgkin lymphoma (NHL) activity of two novel CRM1 inhibitors (KPT-185 and KPT-276) in vitro and in vivo. KPT-185 displayed potent antiproliferative properties and induced cell-cycle arrest and apoptosis in several NHL cell lines and patients' tumor cells. The antitumor activity mainly consisted of inducing caspase cleavage and downregulating the expression of antiapoptotic proteins such as CRM1, nuclear factor-κB, and survivin. Furthermore, oral administration of KPT-276 significantly suppressed tumor growth in mice with Jeko-1 xenograft without any major toxic effects.
Collapse
Affiliation(s)
- Xiaohong Han
- Department of Medical Oncology, Cancer Institute/Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing 100021, China
| | - Jianfei Wang
- Department of Medical Oncology, Cancer Institute/Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing 100021, China
| | - Yinchen Shen
- Department of Medical Oncology, Cancer Institute/Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing 100021, China
| | - Ningning Zhang
- Department of Medical Oncology, Cancer Institute/Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing 100021, China
| | - Shuai Wang
- Department of Medical Oncology, Cancer Institute/Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing 100021, China
| | - Jiarui Yao
- Department of Medical Oncology, Cancer Institute/Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing 100021, China
| | - Yuankai Shi
- Department of Medical Oncology, Cancer Institute/Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing 100021, China.
| |
Collapse
|
91
|
Gravina GL, Senapedis W, McCauley D, Baloglu E, Shacham S, Festuccia C. Nucleo-cytoplasmic transport as a therapeutic target of cancer. J Hematol Oncol 2014; 7:85. [PMID: 25476752 PMCID: PMC4272779 DOI: 10.1186/s13045-014-0085-1] [Citation(s) in RCA: 202] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 11/09/2014] [Indexed: 12/19/2022] Open
Abstract
Shuttling of specific proteins out of the nucleus is essential for the regulation of the cell cycle and proliferation of both normal and malignant tissues. Dysregulation of this fundamental process may affect many other important cellular processes such as tumor growth, inflammatory response, cell cycle, and apoptosis. It is known that XPO1 (Exportin-1/Chromosome Region Maintenance 1/CRM1) is the main mediator of nuclear export in many cell types. Nuclear proteins exported to the cytoplasm by XPO1 include the drug targets topoisomerase IIα (topo IIα) and BCR-ABL and tumor suppressor proteins such as Rb, APC, p53, p21, and p27. XPO1 can mediate cell proliferation through several pathways: (i) the sub-cellular localization of NES-containing oncogenes and tumor suppressor proteins, (ii) the control of the mitotic apparatus and chromosome segregation, and (iii) the maintenance of nuclear and chromosomal structures. The XPO1 protein is elevated in ovarian carcinoma, glioma, osteosarcoma, pancreatic and cervical cancer. There is a growing body of research indicating that XPO1 may have an important role as a prognostic marker in solid tumors. Because of this, nuclear export inhibition through XPO1 is a potential target for therapeutic intervention in many cancers. The best understood XPO1 inhibitors are the small molecule nuclear export inhibitors (NEIs; Leptomycin B and derivatives, ratjadones, PKF050-638, valtrate, ACA, CBS9106, selinexor/KPT-330, and verdinexor/KPT-335). Selinexor and verdinexor are orally bioavailable, highly potent, small molecules that are classified as Selective Inhibitors of Nuclear Export (SINE). KPT-330 is the only NEI currently in Phase I/II human clinical trials in hematological and solid cancers. Of all the potential targets in nuclear cytoplasmic transport, the nuclear export receptor XPO1 remains the best understood and most advanced therapeutic target for the treatment of cancer.
Collapse
Affiliation(s)
- Giovanni Luca Gravina
- />Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | | | - Dilara McCauley
- />Karyopharm Therapeutics, Inc., 85 Wells Avenue, Newton, MA USA
| | - Erkan Baloglu
- />Karyopharm Therapeutics, Inc., 85 Wells Avenue, Newton, MA USA
| | - Sharon Shacham
- />Karyopharm Therapeutics, Inc., 85 Wells Avenue, Newton, MA USA
| | - Claudio Festuccia
- />Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
92
|
Intronic variants in the NFKB1 gene may influence hearing forecast in patients with unilateral sensorineural hearing loss in Meniere's disease. PLoS One 2014; 9:e112171. [PMID: 25397881 PMCID: PMC4232390 DOI: 10.1371/journal.pone.0112171] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 10/13/2014] [Indexed: 12/24/2022] Open
Abstract
Meniere's disease is an episodic vestibular syndrome associated with sensorineural hearing loss (SNHL) and tinnitus. Patients with MD have an elevated prevalence of several autoimmune diseases (rheumatoid arthritis, systemic lupus erythematosus, ankylosing spondylitis and psoriasis), which suggests a shared autoimmune background. Functional variants of several genes involved in the NF-κB pathway, such as REL, TNFAIP3, NFKB1 and TNIP1, have been associated with two or more immune-mediated diseases and allelic variations in the TLR10 gene may influence bilateral affectation and clinical course in MD. We have genotyped 716 cases of MD and 1628 controls by using the ImmunoChip, a high-density genotyping array containing 186 autoimmune loci, to explore the association of immune system related-loci with sporadic MD. Although no single nucleotide polymorphism (SNP) reached a genome-wide significant association (p<10−8), we selected allelic variants in the NF-kB pathway for further analyses to evaluate the impact of these SNPs in the clinical outcome of MD in our cohort. None of the selected SNPs increased susceptibility for MD in patients with uni or bilateral SNHL. However, two potential regulatory variants in the NFKB1 gene (rs3774937 and rs4648011) were associated with a faster hearing loss progression in patients with unilateral SNHL. So, individuals with unilateral MD carrying the C allele in rs3774937 or G allele in rs4648011 had a shorter mean time to reach hearing stage 3 (>40 dB HL) (log-rank test, corrected p values were p = 0.009 for rs3774937 and p = 0.003 for rs4648011, respectively). No variants influenced hearing in bilateral MD. Our data support that the allelic variants rs3774937 and rs4648011 can modify hearing outcome in patients with MD and unilateral SNHL.
Collapse
|
93
|
Withaferin A disrupts ubiquitin-based NEMO reorganization induced by canonical NF-κB signaling. Exp Cell Res 2014; 331:58-72. [PMID: 25304104 DOI: 10.1016/j.yexcr.2014.09.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 09/03/2014] [Accepted: 09/27/2014] [Indexed: 01/18/2023]
Abstract
The NF-κB family of transcription factors regulates numerous cellular processes, including cell proliferation and survival responses. The constitutive activation of NF-κB has also emerged as an important oncogenic driver in many malignancies, such as activated B-cell like diffuse large B cell lymphoma, among others. In this study, we investigated the impact and mechanisms of action of Withaferin A, a naturally produced steroidal lactone, against both signal-inducible as well as constitutive NF-κB activities. We found that Withaferin A is a robust inhibitor of canonical and constitutive NF-κB activities, leading to apoptosis of certain lymphoma lines. In the canonical pathway induced by TNF, Withaferin A did not disrupt RIP1 polyubiquitination or NEMO-IKKβ interaction and was a poor direct IKKβ inhibitor, but prevented the formation of TNF-induced NEMO foci which colocalized with TNF ligand. While GFP-NEMO efficiently formed TNF-induced foci, a GFP-NEMO(Y308S) mutant that is defective in binding to polyubiquitin chains did not form foci. Our study reveals that Withaferin A is a novel type of IKK inhibitor which acts by disrupting NEMO reorganization into ubiquitin-based signaling structures in vivo.
Collapse
|
94
|
Sari AN, Korkmaz B, Serin MS, Kacan M, Unsal D, Buharalioglu CK, Firat SS, Manhati VL, Falck JR, Malik KU, Tunctan B. Effects of 5,14-HEDGE, a 20-HETE mimetic, on lipopolysaccharide-induced changes in MyD88/TAK1/IKKβ/IκB-α/NF-κB pathway and circulating miR-150, miR-223, and miR-297 levels in a rat model of septic shock. Inflamm Res 2014; 63:741-56. [PMID: 24915805 PMCID: PMC4158117 DOI: 10.1007/s00011-014-0747-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Revised: 05/15/2014] [Accepted: 05/26/2014] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVES We have previously demonstrated that a stable synthetic analog of 20-hydroxyeicosatetraenoic acid (20-HETE), N-(20-hydroxyeicosa-5[Z],14[Z]-dienoyl)glycine (5,14-HEDGE), which mimics the effects of endogenously produced 20-HETE, prevents vascular hyporeactivity, hypotension, tachycardia, inflammation, and mortality in a rodent model of septic shock. The present study was performed to determine whether decreased renal and cardiovascular expression and activity of myeloid differentiation factor 88 (MyD88)/transforming growth factor-activated kinase 1 (TAK1)/inhibitor of κB (IκB) kinase β (IKKβ)/IκB-α/nuclear factor-κB (NF-κB) pathway and reduced circulating microRNA (miR)-150, miR-223, and miR-297 expression levels participate in the protective effect of 5,14-HEDGE against hypotension, tachycardia, and inflammation in response to systemic administration of lipopolysaccharide (LPS). METHODS Conscious male Wistar rats received saline (4 ml/kg) or LPS (10 mg/kg) at time 0. Blood pressure and heart rate were measured using a tail-cuff device. Separate groups of LPS-treated rats were given 5,14-HEDGE (30 mg/kg) 1 h after injection of saline or LPS. The rats were killed 4 h after LPS challenge and blood, kidney, heart, thoracic aorta, and superior mesenteric artery were collected for measurement of the protein expression. RESULTS LPS-induced fall in blood pressure and rise in heart rate were associated with increased MyD88 expression and phosphorylation of TAK1 and IκB-α in cytosolic fractions of the tissues. LPS also caused an increase in both unphosphorylated and phosphorylated NF-κB p65 proteins in the cytosolic and nuclear fractions as well as nuclear translocation of NF-κB p65. In addition, serum miR-150, miR-223, and miR-297 expression levels were increased in LPS-treated rats. These effects of LPS were prevented by 5,14-HEDGE. CONCLUSIONS These results suggest that downregulation of MyD88/TAK1/IKKβ/IκB-α/NF-κB pathway as well as decreased circulating miR-150, miR-223, and miR-297 expression levels participate in the protective effect of 5,14-HEDGE against hypotension, tachycardia, and inflammation in the rat model of septic shock.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Arterial Pressure/drug effects
- Disease Models, Animal
- Heart Rate/drug effects
- Hydroxyeicosatetraenoic Acids
- I-kappa B Kinase/metabolism
- I-kappa B Proteins/metabolism
- Kidney/drug effects
- Kidney/metabolism
- Lipopeptides/pharmacology
- Lipopeptides/therapeutic use
- Lipopolysaccharides
- MAP Kinase Kinase Kinases/metabolism
- Male
- Mesenteric Artery, Superior/drug effects
- Mesenteric Artery, Superior/metabolism
- MicroRNAs/blood
- Myeloid Differentiation Factor 88/metabolism
- Myocardium/metabolism
- Protective Agents/pharmacology
- Protective Agents/therapeutic use
- Rats, Wistar
- Shock, Septic/blood
- Shock, Septic/drug therapy
- Shock, Septic/metabolism
- Shock, Septic/physiopathology
- Transcription Factor RelA/metabolism
Collapse
Affiliation(s)
- A. Nihal Sari
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Belma Korkmaz
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Mehmet Sami Serin
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Meltem Kacan
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Demet Unsal
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | | | - Seyhan Sahan Firat
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Vijay L. Manhati
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John R. Falck
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kafait U. Malik
- Department of Pharmacology, College of Medicine, University of Tennessee, Center for Health Sciences, Memphis, TN, USA
| | - Bahar Tunctan
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| |
Collapse
|
95
|
Perwitasari O, Johnson S, Yan X, Howerth E, Shacham S, Landesman Y, Baloglu E, McCauley D, Tamir S, Tompkins SM, Tripp RA. Verdinexor, a novel selective inhibitor of nuclear export, reduces influenza a virus replication in vitro and in vivo. J Virol 2014; 88:10228-43. [PMID: 24965445 PMCID: PMC4136318 DOI: 10.1128/jvi.01774-14] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 06/18/2014] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Influenza is a global health concern, causing death, morbidity, and economic losses. Chemotherapeutics that target influenza virus are available; however, rapid emergence of drug-resistant strains is common. Therapeutic targeting of host proteins hijacked by influenza virus to facilitate replication is an antiviral strategy to reduce the development of drug resistance. Nuclear export of influenza virus ribonucleoprotein (vRNP) from infected cells has been shown to be mediated by exportin 1 (XPO1) interaction with viral nuclear export protein tethered to vRNP. RNA interference screening has identified XPO1 as a host proinfluenza factor where XPO1 silencing results in reduced influenza virus replication. The Streptomyces metabolite XPO1 inhibitor leptomycin B (LMB) has been shown to limit influenza virus replication in vitro; however, LMB is toxic in vivo, which makes it unsuitable for therapeutic use. In this study, we tested the anti-influenza virus activity of a new class of orally available small-molecule selective inhibitors of nuclear export, specifically, the XPO1 antagonist KPT-335 (verdinexor). Verdinexor was shown to potently and selectively inhibit vRNP export and effectively inhibited the replication of various influenza virus A and B strains in vitro, including pandemic H1N1 virus, highly pathogenic H5N1 avian influenza virus, and the recently emerged H7N9 strain. In vivo, prophylactic and therapeutic administration of verdinexor protected mice against disease pathology following a challenge with influenza virus A/California/04/09 or A/Philippines/2/82-X79, as well as reduced lung viral loads and proinflammatory cytokine expression, while having minimal toxicity. These studies show that verdinexor acts as a novel anti-influenza virus therapeutic agent. IMPORTANCE Antiviral drugs represent important means of influenza virus control. However, substantial resistance to currently approved influenza therapeutic drugs has developed. New antiviral approaches are required to address drug resistance and reduce the burden of influenza virus-related disease. This study addressed critical preclinical studies for the development of verdinexor (KPT-335) as a novel antiviral drug. Verdinexor blocks progeny influenza virus genome nuclear export, thus effectively inhibiting virus replication. Verdinexor was found to limit the replication of various strains of influenza A and B viruses, including a pandemic H1N1 influenza virus strain, a highly pathogenic H5N1 avian influenza virus strain, and a recently emerging H7N9 influenza virus strain. Importantly, oral verdinexor treatments, given prophylactically or therapeutically, were efficacious in limiting lung virus burdens in influenza virus-infected mice, in addition to limiting lung proinflammatory cytokine expression, pathology, and death. Thus, this study demonstrated that verdinexor is efficacious against influenza virus infection in vitro and in vivo.
Collapse
Affiliation(s)
- Olivia Perwitasari
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, Georgia, USA
| | - Scott Johnson
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, Georgia, USA
| | - Xiuzhen Yan
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, Georgia, USA
| | - Elizabeth Howerth
- Department of Pathology, University of Georgia College of Veterinary Medicine, Athens, Georgia, USA
| | - Sharon Shacham
- Karyopharm Therapeutics, Inc., Natick, Massachusetts, USA
| | | | - Erkan Baloglu
- Karyopharm Therapeutics, Inc., Natick, Massachusetts, USA
| | | | - Sharon Tamir
- Karyopharm Therapeutics, Inc., Natick, Massachusetts, USA
| | - S Mark Tompkins
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, Georgia, USA
| | - Ralph A Tripp
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, Georgia, USA
| |
Collapse
|
96
|
Yang X, Cheng L, Yao L, Ren H, Zhang S, Min X, Chen X, Zhang J, Li M. Involvement of chromosome region maintenance 1 (CRM1) in the formation and progression of esophageal squamous cell carcinoma. Med Oncol 2014; 31:155. [PMID: 25148895 DOI: 10.1007/s12032-014-0155-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 07/29/2014] [Indexed: 11/29/2022]
Abstract
Chromosome region maintenance 1 (CRM1) has been related to several malignancies. The predictive value of CRM1 in the malignance and prognosis of esophageal squamous cell carcinoma (ESCC), however, is not clear yet. In this study, we displayed that CRM1 expression was up-regulated in ESCC using immunohistochemistry and Western blot. Statistical analysis demonstrated that patients with high CRM1 levels indicated shorter survival period. We further found that silencing CRM1 caused apoptosis in ESCC cell lines. Moreover, knockdown of CRM1 disturbed the expression of tumor suppressor proteins and inhibited NF-κB activity in ESCC cell lines, especially if the cell line was treated with 5-fluorouracil. In consequence, our results for the first time indicated that CRM1 was dysregulated in ESCC, and suppression of CRM1 expression which resulted in inhibiting of NF-κB signaling might be developed into a new strategy in ESCC therapy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Squamous Cell/chemistry
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/mortality
- Cell Line, Tumor
- Disease Progression
- Esophageal Neoplasms/chemistry
- Esophageal Neoplasms/metabolism
- Esophageal Neoplasms/mortality
- Esophageal Squamous Cell Carcinoma
- Esophagus/chemistry
- Esophagus/metabolism
- Female
- Humans
- Immunohistochemistry
- Karyopherins/analysis
- Karyopherins/genetics
- Karyopherins/metabolism
- Male
- Mice
- Mice, Nude
- Middle Aged
- NF-kappa B/metabolism
- Prognosis
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Receptors, Cytoplasmic and Nuclear/analysis
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Survival Analysis
- Exportin 1 Protein
Collapse
Affiliation(s)
- Xiaojing Yang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Jiangsu, 226001, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Zhao B, Barrera LA, Ersing I, Willox B, Schmidt SCS, Greenfeld H, Zhou H, Mollo SB, Shi TT, Takasaki K, Jiang S, Cahir-McFarland E, Kellis M, Bulyk ML, Kieff E, Gewurz BE. The NF-κB genomic landscape in lymphoblastoid B cells. Cell Rep 2014; 8:1595-606. [PMID: 25159142 DOI: 10.1016/j.celrep.2014.07.037] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 06/09/2014] [Accepted: 07/21/2014] [Indexed: 01/17/2023] Open
Abstract
The nuclear factor κB (NF-κΒ) subunits RelA, RelB, cRel, p50, and p52 are each critical for B cell development and function. To systematically characterize their responses to canonical and noncanonical NF-κB pathway activity, we performed chromatin immunoprecipitation followed by high-throughput DNA sequencing (ChIP-seq) analysis in lymphoblastoid B cell lines (LCLs). We found a complex NF-κB-binding landscape, which did not readily reflect the two NF-κB pathway paradigms. Instead, 10 subunit-binding patterns were observed at promoters and 11 at enhancers. Nearly one-third of NF-κB-binding sites lacked κB motifs and were instead enriched for alternative motifs. The oncogenic forkhead box protein FOXM1 co-occupied nearly half of NF-κB-binding sites and was identified in protein complexes with NF-κB on DNA. FOXM1 knockdown decreased NF-κB target gene expression and ultimately induced apoptosis, highlighting FOXM1 as a synthetic lethal target in B cell malignancy. These studies provide a resource for understanding mechanisms that underlie NF-κB nuclear activity and highlight opportunities for selective NF-κB blockade.
Collapse
Affiliation(s)
- Bo Zhao
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Luis A Barrera
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA; Committee on Higher Degrees in Biophysics, Harvard University, Cambridge, MA 02138, USA; Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA 02115, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ina Ersing
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Bradford Willox
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Stefanie C S Schmidt
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Hannah Greenfeld
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Hufeng Zhou
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah B Mollo
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Tommy T Shi
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kaoru Takasaki
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Sizun Jiang
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Ellen Cahir-McFarland
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Manolis Kellis
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Martha L Bulyk
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA; Committee on Higher Degrees in Biophysics, Harvard University, Cambridge, MA 02138, USA; Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Elliott Kieff
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Benjamin E Gewurz
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
98
|
Erlejman AG, De Leo SA, Mazaira GI, Molinari AM, Camisay MF, Fontana V, Cox MB, Piwien-Pilipuk G, Galigniana MD. NF-κB transcriptional activity is modulated by FK506-binding proteins FKBP51 and FKBP52: a role for peptidyl-prolyl isomerase activity. J Biol Chem 2014; 289:26263-26276. [PMID: 25104352 DOI: 10.1074/jbc.m114.582882] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Hsp90 binding immunophilins FKBP51 and FKBP52 modulate steroid receptor trafficking and hormone-dependent biological responses. With the purpose to expand this model to other nuclear factors that are also subject to nuclear-cytoplasmic shuttling, we analyzed whether these immunophilins modulate NF-κB signaling. It is demonstrated that FKBP51 impairs both the nuclear translocation rate of NF-κB and its transcriptional activity. The inhibitory action of FKBP51 requires neither the peptidylprolyl-isomerase activity of the immunophilin nor its association with Hsp90. The TPR domain of FKBP51 is essential. On the other hand, FKBP52 favors the nuclear retention time of RelA, its association to a DNA consensus binding sequence, and NF-κB transcriptional activity, the latter effect being strongly dependent on the peptidylprolyl-isomerase activity and also on the TPR domain of FKBP52, but its interaction with Hsp90 is not required. In unstimulated cells, FKBP51 forms endogenous complexes with cytoplasmic RelA. Upon cell stimulation with phorbol ester, the NF-κB soluble complex exchanges FKBP51 for FKBP52, and the NF-κB biological effect is triggered. Importantly, FKBP52 is functionally recruited to the promoter region of NF-κB target genes, whereas FKBP51 is released. Competition assays demonstrated that both immunophilins antagonize one another, and binding assays with purified proteins suggest that the association of RelA and immunophilins could be direct. These observations suggest that the biological action of NF-κB in different cell types could be positively regulated by a high FKBP52/FKBP51 expression ratio by favoring NF-κB nuclear retention, recruitment to the promoter regions of target genes, and transcriptional activity.
Collapse
Affiliation(s)
- Alejandra G Erlejman
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Química Biológica de Ciencias Exactas y Naturales (IQUIBICEN)/Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, C1428ADN Argentina
| | - Sonia A De Leo
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Química Biológica de Ciencias Exactas y Naturales (IQUIBICEN)/Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, C1428ADN Argentina
| | - Gisela I Mazaira
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Química Biológica de Ciencias Exactas y Naturales (IQUIBICEN)/Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, C1428ADN Argentina
| | - Alejandro M Molinari
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Química Biológica de Ciencias Exactas y Naturales (IQUIBICEN)/Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, C1428ADN Argentina
| | - María Fernanda Camisay
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Química Biológica de Ciencias Exactas y Naturales (IQUIBICEN)/Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, C1428ADN Argentina
| | - Vanina Fontana
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Química Biológica de Ciencias Exactas y Naturales (IQUIBICEN)/Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, C1428ADN Argentina
| | - Marc B Cox
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas, El Paso, Texas 79968
| | - Graciela Piwien-Pilipuk
- Laboratorio de Arquitectura Nuclear, Instituto de Biología y Medicina Experimental/CONICET, Buenos Aires C1428ADN, Argentina, and
| | - Mario D Galigniana
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Química Biológica de Ciencias Exactas y Naturales (IQUIBICEN)/Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, C1428ADN Argentina,; Laboratorio de Receptores Nucleares, Instituto de Biología y Medicina Experimental/CONICET, Buenos Aires C1428ADN, Argentina.
| |
Collapse
|
99
|
Senapedis WT, Baloglu E, Landesman Y. Clinical translation of nuclear export inhibitors in cancer. Semin Cancer Biol 2014; 27:74-86. [DOI: 10.1016/j.semcancer.2014.04.005] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 04/10/2014] [Indexed: 01/18/2023]
|
100
|
Sun H, Hattori N, Chien W, Sun Q, Sudo M, E-Ling GL, Ding L, Lim SL, Shacham S, Kauffman M, Nakamaki T, Koeffler HP. KPT-330 has antitumour activity against non-small cell lung cancer. Br J Cancer 2014; 111:281-91. [PMID: 24946002 PMCID: PMC4102938 DOI: 10.1038/bjc.2014.260] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/14/2014] [Accepted: 04/23/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND We investigated the biologic and pharmacologic activities of a chromosome region maintenance 1 (CRM1) inhibitor against human non-small cell lung cancer (NSCLC) cells both in vitro and in vivo. METHODS The in vitro and in vivo effects of a novel CRM1 inhibitor (KPT-330) for a large number of anticancer parameters were evaluated using a large panel of 11 NSCLC cell lines containing different key driver mutations. Mice bearing human NSCLC xenografts were treated with KPT-330, and tumour growth was assessed. RESULTS KPT-330 inhibited proliferation and induced cell cycle arrest and apoptosis-related proteins in 11 NSCLC cells lines. Moreover, the combination of KPT-330 with cisplatin synergistically enhanced the cell kill of the NSCLC cells in vitro. Human NSCLC tumours growing in immunodeficient mice were markedly inhibited by KPT-330. Also, KPT-330 was effective even against NSCLC cells with a transforming mutation of either exon 20 of EGFR, TP53, phosphatase and tensin homologue, RAS or PIK3CA, suggesting the drug might be effective against a variety of lung cancers irrespective of their driver mutation. CONCLUSIONS Our results support clinical testing of KPT-330 as a novel therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- H Sun
- Department of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - N Hattori
- Cancer Science Institute of Singapore, National Cancer Institute, NUS, Singapore, Singapore
| | - W Chien
- Cancer Science Institute of Singapore, National Cancer Institute, NUS, Singapore, Singapore
| | - Q Sun
- Cancer Science Institute of Singapore, National Cancer Institute, NUS, Singapore, Singapore
| | - M Sudo
- Cancer Science Institute of Singapore, National Cancer Institute, NUS, Singapore, Singapore
| | - G L E-Ling
- Cancer Science Institute of Singapore, National Cancer Institute, NUS, Singapore, Singapore
| | - L Ding
- Cancer Science Institute of Singapore, National Cancer Institute, NUS, Singapore, Singapore
| | - S L Lim
- Cancer Science Institute of Singapore, National Cancer Institute, NUS, Singapore, Singapore
| | - S Shacham
- Karyopharm Therapeutics, Boston, MA 01760, USA
| | - M Kauffman
- Karyopharm Therapeutics, Boston, MA 01760, USA
| | - T Nakamaki
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - H P Koeffler
- Department of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Science Institute of Singapore, National Cancer Institute, NUS, Singapore, Singapore
| |
Collapse
|