1
|
Msweli S, Pakala SB, Syed K. NF-κB Transcription Factors: Their Distribution, Family Expansion, Structural Conservation, and Evolution in Animals. Int J Mol Sci 2024; 25:9793. [PMID: 39337282 PMCID: PMC11432056 DOI: 10.3390/ijms25189793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
The Nuclear Factor Kappa B (NF-κB) transcription factor family consists of five members: RelA (p65), RelB, c-Rel, p50 (p105/NF-κB1), and p52 (p100/NF-κB2). This family is considered a master regulator of classical biochemical pathways such as inflammation, immunity, cell proliferation, and cell death. The proteins in this family have a conserved Rel homology domain (RHD) with the following subdomains: DNA binding domain (RHD-DBD) and dimerization domain (RHD-DD). Despite the importance of the NF-κB family in biology, there is a lack of information with respect to their distribution patterns, evolution, and structural conservation concerning domains and subdomains in animals. This study aims to address this critical gap regarding NF-κB proteins. A comprehensive analysis of NF-κB family proteins revealed their distinct distribution in animals, with differences in protein sizes, conserved domains, and subdomains (RHD-DBD and RHD-DD). For the first time, NF-κB proteins with multiple RHD-DBDs and RHD-DDs have been identified, and in some cases, this is due to subdomain duplication. The presence of RelA/p65 exclusively in vertebrates shows that innate immunity originated in fishes, followed by amphibians, reptiles, aves, and mammals. Phylogenetic analysis showed that NF-κB family proteins grouped according to animal groups, signifying structural conservation after speciation. The evolutionary analysis of RHDs suggests that NF-κB family members p50/p105 and c-Rel may have been the first to emerge in arthropod ancestors, followed by RelB, RelA, and p52/p100.
Collapse
Affiliation(s)
- Siphesihle Msweli
- Department of Biochemistry and Microbiology, Faculty of Science, Agriculture and Engineering, University of Zululand, KwaDlangezwa 3886, South Africa; (S.M.); (S.B.P.)
| | - Suresh B. Pakala
- Department of Biochemistry and Microbiology, Faculty of Science, Agriculture and Engineering, University of Zululand, KwaDlangezwa 3886, South Africa; (S.M.); (S.B.P.)
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500-046, India
| | - Khajamohiddin Syed
- Department of Biochemistry and Microbiology, Faculty of Science, Agriculture and Engineering, University of Zululand, KwaDlangezwa 3886, South Africa; (S.M.); (S.B.P.)
| |
Collapse
|
2
|
Han W, Xiong W, Sun W, Liu W, Zhang Y, Li C, Gu N, Shen Y, Qiu Z, Li C, Zhao Y, Zhao R. YTHDC1 Mitigates Apoptosis in Bone Marrow Mesenchymal Stem Cells by Inhibiting NfƙBiα and Augmenting Cardiac Function Following Myocardial Infarction. Cell Transplant 2024; 33:9636897241290910. [PMID: 39466658 PMCID: PMC11528794 DOI: 10.1177/09636897241290910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/21/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024] Open
Abstract
The therapeutic efficacy of bone marrow mesenchymal stem cells (BMSCs) in myocardial infarction (MI) is hindered by poor cell survival. This study explored the role of N6-methyladenosine (m6A) regulation, specifically YTHDC1, in improving BMSC transplantation for MI. By screening m6A-related regulators in hypoxia and serum deprivation (HSD)-induced BMSC apoptosis, YTHDC1 was found to be downregulated. Overexpression of Ythdc1 in BMSCs reduced apoptosis markers, reactive oxygen species (ROS) release, and improved cell survival under HSD conditions. Conversely, Ythdc1 knockdown enhanced apoptosis. In rat MI models, transplantation of Ythdc1-overexpressing BMSCs improved cardiac function and reduced myocardial fibrosis. Mechanistically, YTHDC1 interacts with nuclear factor kappa B (NF-κB) inhibitor-alpha mRNA, suggesting its involvement in BMSC survival pathways. This study identifies YTHDC1 as a potential target to enhance BMSC efficacy in MI therapy.
Collapse
Affiliation(s)
- Weiyu Han
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Weidong Xiong
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Weixing Sun
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Cardiology, People’s Hospital of Honghuagang District, Zunyi, China
| | - Weiwei Liu
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yu Zhang
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chaofu Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ning Gu
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Youcheng Shen
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhimei Qiu
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chaozhong Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yongchao Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ranzun Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
3
|
Chen K, Tang L, Nong X. Artesunate targets cellular metabolism to regulate the Th17/Treg cell balance. Inflamm Res 2023; 72:1037-1050. [PMID: 37024544 DOI: 10.1007/s00011-023-01729-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023] Open
Abstract
INTRODUCTION Metabolic reprogramming is one of the important mechanisms of cell differentiation, and different cells have different preferences for energy sources. During the differentiation of naive CD4 + T cells into Th17 and Treg cells, these cells show specific energy metabolism characteristics. Th17 cells depend on enhanced glycolysis, fatty acid synthesis, and glutaminolysis. In contrast, Treg cells are dependent on oxidative phosphorylation, fatty acid oxidation, and amino acid depletion. As a potent antimalarial drug, artesunate has been shown to modulate the Th17/Treg imbalance and regulate cell metabolism. METHODOLOGY Relevant literatures on ART, cellular metabolism, glycolysis, lipid metabolism, amino acid metabolism, CD4 + T cells, Th17 cells, and Treg cells published from January 1, 2010 to now were searched in PubMed database. CONCLUSION In this review, we will highlight recent advances in which artesunate can restore the Th17/Treg imbalance in disease states by altering T-cell metabolism to influence differentiation and lineage selection. Data from the current study show that few studies have focused on the effect of ART on cellular metabolism. ART can affect the metabolic characteristics of T cells (glycolysis, lipid metabolism, and amino acid metabolism) and interfere with their differentiation lineage, thereby regulating the balance of Th17/Treg and alleviating the symptoms of the disease.
Collapse
Affiliation(s)
- Kun Chen
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Liying Tang
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaolin Nong
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China.
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
4
|
De Re V, Repetto O, Mussolin L, Brisotto G, Elia C, Lopci E, d’Amore ESG, Burnelli R, Mascarin M. Promising drugs and treatment options for pediatric and adolescent patients with Hodgkin lymphoma. Front Cell Dev Biol 2022; 10:965803. [PMID: 36506094 PMCID: PMC9729954 DOI: 10.3389/fcell.2022.965803] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Currently-available therapies for newly-diagnosed pediatric and adolescent patients with Hodgkin lymphoma result in >95% survival at 5 years. Long-term survivors may suffer from long-term treatment-related side effects, however, so the past 20 years have seen clinical trials for children and adolescents with HL gradually abandon the regimens used in adults in an effort to improve this situation. Narrower-field radiotherapy can reduce long-term toxicity while maintaining good tumor control. Various risk-adapted chemo-radiotherapy strategies have been used. Early assessment of tumor response with interim positron emission tomography and/or measuring metabolic tumor volume has been used both to limit RT in patients with favorable characteristics and to adopt more aggressive therapies in patients with a poor response. Most classical Hodgkin's lymphoma relapses occur within 3 years of initial treatment, while relapses occurring 5 years or more after diagnosis are rare. As the outcome for patients with relapsed/refractory classical Hodgkin lymphoma remains unsatisfactory, new drugs have been proposed for its prevention or treatment. This review summarizes the important advances made in recent years in the management of pediatric and adolescent with classical Hodgkin lymphoma, and the novel targeted treatments for relapsed and refractory classical Hodgkin lymphoma.
Collapse
Affiliation(s)
- Valli De Re
- Immunopatologia e Biomarcatori Oncologici, Dipartimento di Ricerca e Diagnostica Avanzata dei Tumori, CRO Aviano, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Aviano, Italy,*Correspondence: Valli De Re, ; Maurizio Mascarin,
| | - Ombretta Repetto
- Immunopatologia e Biomarcatori Oncologici, Dipartimento di Ricerca e Diagnostica Avanzata dei Tumori, CRO Aviano, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Aviano, Italy
| | - Lara Mussolin
- Pediatric Hemato-Oncology Unit, Department of Women’s and Children’s Health, University of Padua, Padua, Italy
| | - Giulia Brisotto
- Immunopatologia e Biomarcatori Oncologici, Dipartimento di Ricerca e Diagnostica Avanzata dei Tumori, CRO Aviano, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Aviano, Italy
| | - Caterina Elia
- AYA Oncology and Pediatric Radiotherapy Unit, Centro di Riferimento Oncologico IRCCS, Aviano, Italy
| | - Egesta Lopci
- Nuclear Medicine, IRCCS—Humanitas Research Hospital, Rozzano, MI, Italy
| | | | - Roberta Burnelli
- Pediatric Hematology-Oncology Unit, Azienda Ospedaliera Universitaria, Ospedale Sant’Anna, Ferrara, Italy
| | - Maurizio Mascarin
- AYA Oncology and Pediatric Radiotherapy Unit, Centro di Riferimento Oncologico IRCCS, Aviano, Italy,*Correspondence: Valli De Re, ; Maurizio Mascarin,
| |
Collapse
|
5
|
Vitali L, Merlini A, Galvagno F, Proment A, Sangiolo D. Biological and Exploitable Crossroads for the Immune Response in Cancer and COVID-19. Biomedicines 2022; 10:2628. [PMID: 36289890 PMCID: PMC9599827 DOI: 10.3390/biomedicines10102628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/05/2022] [Accepted: 10/14/2022] [Indexed: 12/15/2022] Open
Abstract
The outbreak of novel coronavirus disease 2019 (COVID-19) has exacted a disproportionate toll on cancer patients. The effects of anticancer treatments and cancer patients' characteristics shared significant responsibilities for this dismal outcome; however, the underlying immunopathological mechanisms are far from being completely understood. Indeed, despite their different etiologies, SARS-CoV-2 infection and cancer unexpectedly share relevant immunobiological connections. In the pathogenesis and natural history of both conditions, there emerges the centrality of the immune response, orchestrating the timed appearance, functional and dysfunctional roles of multiple effectors in acute and chronic phases. A significant number (more than 600) of observational and interventional studies have explored the interconnections between COVID-19 and cancer, focusing on aspects as diverse as psychological implications and prognostic factors, with more than 4000 manuscripts published so far. In this review, we reported and discussed the dynamic behavior of the main cytokines and immune system signaling pathways involved in acute vs. early, and chronic vs. advanced stages of SARS-CoV-2 infection and cancer. We highlighted the biological similarities and active connections within these dynamic disease scenarios, exploring and speculating on possible therapeutic crossroads from one setting to the other.
Collapse
Affiliation(s)
- Letizia Vitali
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, 10060 Candiolo, Italy
- Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Alessandra Merlini
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, 10060 Candiolo, Italy
- Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Federica Galvagno
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, 10060 Candiolo, Italy
- Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Alessia Proment
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, 10060 Candiolo, Italy
- Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Dario Sangiolo
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 Km 3.95, 10060 Candiolo, Italy
- Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy
| |
Collapse
|
6
|
Shan DD, Zheng QX, Chen Z. Go-Ichi-Ni-San 2: A potential biomarker and therapeutic target in human cancers. World J Gastrointest Oncol 2022; 14:1892-1902. [PMID: 36310704 PMCID: PMC9611433 DOI: 10.4251/wjgo.v14.i10.1892] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/15/2022] [Accepted: 09/06/2022] [Indexed: 02/05/2023] Open
Abstract
Cancer incidence and mortality are increasing globally, leading to its rising status as a leading cause of death. The Go-Ichi-Ni-San (GINS) complex plays a crucial role in DNA replication and the cell cycle. The GINS complex consists of four subunits encoded by the GINS1, GINS2, GINS3, and GINS4 genes. Recent findings have shown that GINS2 expression is upregulated in many diseases, particularly tumors. For example, increased GINS2 expression has been found in cervical cancer, gastric adenocarcinoma, glioma, non-small cell lung cancer, and pancreatic cancer. It correlates with the clinicopathological characteristics of the tumors. In addition, high GINS2 expression plays a pro-carcinogenic role in tumor development by promoting tumor cell proliferation and migration, inhibiting tumor cell apoptosis, and blocking the cell cycle. This review describes the upregulation of GINS2 expression in most human tumors and the pathway of GINS2 in tumor development. GINS2 may serve as a new marker for tumor diagnosis and a new biological target for therapy.
Collapse
Affiliation(s)
- Dan-Dan Shan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Qiu-Xian Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
7
|
Smulski CR, Zhang L, Burek M, Teixidó Rubio A, Briem JS, Sica MP, Sevdali E, Vigolo M, Willen L, Odermatt P, Istanbullu D, Herr S, Cavallari M, Hess H, Rizzi M, Eibel H, Schneider P. Ligand-independent oligomerization of TACI is controlled by the transmembrane domain and regulates proliferation of activated B cells. Cell Rep 2022; 38:110583. [PMID: 35354034 DOI: 10.1016/j.celrep.2022.110583] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 11/03/2021] [Accepted: 03/07/2022] [Indexed: 12/23/2022] Open
Abstract
In mature B cells, TACI controls class-switch recombination and differentiation into plasma cells during T cell-independent antibody responses. TACI binds the ligands BAFF and APRIL. Approximately 10% of patients with common variable immunodeficiency (CVID) carry TACI mutations, of which A181E and C172Y are in the transmembrane domain. Residues A181 and C172 are located on distinct sides of the transmembrane helix, which is predicted by molecular modeling to spontaneously assemble into trimers and dimers. In human B cells, these mutations impair ligand-dependent (C172Y) and -independent (A181E) TACI multimerization and signaling, as well as TACI-enhanced proliferation and/or IgA production. Genetic inactivation of TACI in primary human B cells impaired survival of CpG-activated cells in the absence of ligand. These results identify the transmembrane region of TACI as an active interface for TACI multimerization in signal transduction, in particular for ligand-independent signals. These functions are perturbed by CVID-associated mutations.
Collapse
Affiliation(s)
- Cristian R Smulski
- Department of Biochemistry, University of Lausanne, Ch. des Boveresses 155, 1066 Epalinges, Switzerland; Faculty of Medicine and Medical Center, University of Freiburg, Department of Rheumatology and Center for Chronic Immunodeficiency, Breisacherstr. 115, 79106 Freiburg, Germany; Medical Physics Department, Centro Atómico Bariloche, Comisión Nacional de Energía Atómica (CNEA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Avenida E- Bustillo 9500, R8402AGP Río Negro, San Carlos de Bariloche, Argentina.
| | - Luyao Zhang
- Faculty of Medicine and Medical Center, University of Freiburg, Department of Rheumatology and Center for Chronic Immunodeficiency, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Malte Burek
- Faculty of Medicine and Medical Center, University of Freiburg, Department of Rheumatology and Center for Chronic Immunodeficiency, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Ariadna Teixidó Rubio
- Faculty of Medicine and Medical Center, University of Freiburg, Department of Rheumatology and Center for Chronic Immunodeficiency, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Jana-Susann Briem
- Faculty of Medicine and Medical Center, University of Freiburg, Department of Rheumatology and Center for Chronic Immunodeficiency, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Mauricio P Sica
- Medical Physics Department, Centro Atómico Bariloche, Comisión Nacional de Energía Atómica (CNEA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Avenida E- Bustillo 9500, R8402AGP Río Negro, San Carlos de Bariloche, Argentina; Instituto de Energía y Desarrollo Sustentable, Centro Atómico Bariloche, Comisión Nacional de Energía Atómica (CNEA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Avenida E- Bustillo 9500, R8402AGP Río Negro, San Carlos de Bariloche, Argentina
| | - Eirini Sevdali
- Faculty of Medicine and Medical Center, University of Freiburg, Department of Rheumatology and Center for Chronic Immunodeficiency, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Michele Vigolo
- Department of Biochemistry, University of Lausanne, Ch. des Boveresses 155, 1066 Epalinges, Switzerland
| | - Laure Willen
- Department of Biochemistry, University of Lausanne, Ch. des Boveresses 155, 1066 Epalinges, Switzerland
| | - Patricia Odermatt
- Faculty of Medicine and Medical Center, University of Freiburg, Department of Rheumatology and Center for Chronic Immunodeficiency, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Duygu Istanbullu
- Faculty of Medicine and Medical Center, University of Freiburg, Department of Rheumatology and Center for Chronic Immunodeficiency, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Stephanie Herr
- Faculty of Medicine and Medical Center, University of Freiburg, Department of Rheumatology and Center for Chronic Immunodeficiency, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Marco Cavallari
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestr. 18, 79104 Freiburg, Germany
| | | | - Marta Rizzi
- Faculty of Medicine and Medical Center, University of Freiburg, Department of Rheumatology and Center for Chronic Immunodeficiency, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Hermann Eibel
- Faculty of Medicine and Medical Center, University of Freiburg, Department of Rheumatology and Center for Chronic Immunodeficiency, Breisacherstr. 115, 79106 Freiburg, Germany
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Ch. des Boveresses 155, 1066 Epalinges, Switzerland.
| |
Collapse
|
8
|
Sheth M, Esfandiari L. Bioelectric Dysregulation in Cancer Initiation, Promotion, and Progression. Front Oncol 2022; 12:846917. [PMID: 35359398 PMCID: PMC8964134 DOI: 10.3389/fonc.2022.846917] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer is primarily a disease of dysregulation – both at the genetic level and at the tissue organization level. One way that tissue organization is dysregulated is by changes in the bioelectric regulation of cell signaling pathways. At the basis of bioelectricity lies the cellular membrane potential or Vmem, an intrinsic property associated with any cell. The bioelectric state of cancer cells is different from that of healthy cells, causing a disruption in the cellular signaling pathways. This disruption or dysregulation affects all three processes of carcinogenesis – initiation, promotion, and progression. Another mechanism that facilitates the homeostasis of cell signaling pathways is the production of extracellular vesicles (EVs) by cells. EVs also play a role in carcinogenesis by mediating cellular communication within the tumor microenvironment (TME). Furthermore, the production and release of EVs is altered in cancer. To this end, the change in cell electrical state and in EV production are responsible for the bioelectric dysregulation which occurs during cancer. This paper reviews the bioelectric dysregulation associated with carcinogenesis, including the TME and metastasis. We also look at the major ion channels associated with cancer and current technologies and tools used to detect and manipulate bioelectric properties of cells.
Collapse
Affiliation(s)
- Maulee Sheth
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, United States
| | - Leyla Esfandiari
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, United States
- Department of Electrical Engineering and Computer Science, University of Cincinnati, Cincinnati, OH, United States
- Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH, United States
- *Correspondence: Leyla Esfandiari,
| |
Collapse
|
9
|
Mindt BC, Krisna SS, Duerr CU, Mancini M, Richer L, Vidal SM, Gerondakis S, Langlais D, Fritz JH. The NF-κB Transcription Factor c-Rel Modulates Group 2 Innate Lymphoid Cell Effector Functions and Drives Allergic Airway Inflammation. Front Immunol 2021; 12:664218. [PMID: 34867937 PMCID: PMC8635195 DOI: 10.3389/fimmu.2021.664218] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 08/27/2021] [Indexed: 01/03/2023] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) play a key role in the initiation and orchestration of early type 2 immune responses. Upon tissue damage, ILC2s are activated by alarmins such as IL-33 and rapidly secrete large amounts of type 2 signature cytokines. ILC2 activation is governed by a network of transcriptional regulators including nuclear factor (NF)-κB family transcription factors. While it is known that activating IL-33 receptor signaling results in downstream NF-κB activation, the underlying molecular mechanisms remain elusive. Here, we found that the NF-κB subunit c-Rel is required to mount effective innate pulmonary type 2 immune responses. IL-33-mediated activation of ILC2s in vitro as well as in vivo was found to induce c-Rel mRNA and protein expression. In addition, we demonstrate that IL-33-mediated activation of ILC2s leads to nuclear translocation of c-Rel in pulmonary ILC2s. Although c-Rel was found to be a critical mediator of innate pulmonary type 2 immune responses, ILC2-intrinsic deficiency of c-Rel did not have an impact on the developmental capacity of ILC2s nor affected homeostatic numbers of lung-resident ILC2s at steady state. Moreover, we demonstrate that ILC2-intrinsic deficiency of c-Rel alters the capacity of ILC2s to upregulate the expression of ICOSL and OX40L, key stimulatory receptors, and the expression of type 2 signature cytokines IL-5, IL-9, IL-13, and granulocyte-macrophage colony-stimulating factor (GM-CSF). Collectively, our data using Rel−/− mice suggest that c-Rel promotes acute ILC2-driven allergic airway inflammation and suggest that c-Rel may contribute to the pathophysiology of ILC2-mediated allergic airway disease. It thereby represents a promising target for the treatment of allergic asthma, and evaluating the effect of established c-Rel inhibitors in this context would be of great clinical interest.
Collapse
Affiliation(s)
- Barbara C. Mindt
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - Sai Sakktee Krisna
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Physiology, McGill University, Montréal, QC, Canada
| | - Claudia U. Duerr
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Berlin, Germany
| | - Mathieu Mancini
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Lara Richer
- Department of Pathology, McGill University, Montréal, QC, Canada
| | - Silvia M. Vidal
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Steven Gerondakis
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - David Langlais
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- McGill University Genome Centre, Montreal, QC, Canada
| | - Jörg H. Fritz
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Department of Physiology, McGill University, Montréal, QC, Canada
- FOCiS Centre of Excellence in Translational Immunology (CETI), Montréal, QC, Canada
- *Correspondence: Jörg H. Fritz,
| |
Collapse
|
10
|
Lee SR, Qian M, Yang W, Diedrich JD, Raetz E, Yang W, Dong Q, Devidas M, Pei D, Yeoh A, Cheng C, Pui CH, Evans WE, Mullighan CG, Hunger SP, Savic D, Relling MV, Loh ML, Yang JJ. Genome-Wide Association Study of Susceptibility Loci for TCF3-PBX1 Acute Lymphoblastic Leukemia in Children. J Natl Cancer Inst 2021; 113:933-937. [PMID: 32882024 PMCID: PMC8487647 DOI: 10.1093/jnci/djaa133] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/14/2020] [Accepted: 08/20/2020] [Indexed: 01/03/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common cancer in children. TCF3-PBX1 fusion defines a common molecular subtype of ALL with unique clinical features, but the molecular basis of its inherited susceptibility is unknown. In a genome-wide association study of 1494 ALL cases and 2057 non-ALL controls, we identified a germline risk locus located in an intergenic region between BCL11A and PAPOLG: rs2665658, P = 1.88 × 10-8 for TCF3-PBX1 ALL vs non-ALL, and P = 1.70 × 10-8 for TCF3-PBX1 ALL vs other-ALL. The lead variant was validated in a replication cohort, and conditional analyses pointed to a single causal variant with subtype-specific effect. The risk variant is located in a regulatory DNA element uniquely activated in ALL cells with the TCF3-PBX1 fusion and may distally modulate the transcription of the adjacent gene REL. Our results expand the understanding of subtype-specific ALL susceptibility and highlight plausible interplay between germline variants and somatic genomic abnormalities in ALL pathogenesis.
Collapse
Affiliation(s)
- Shawn H. R Lee
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
- Khoo Teck Puat–National University Children’s Medical Institute, National University Health System, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Maoxiang Qian
- Institute of Pediatrics and Department of Hematology and Oncology, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wentao Yang
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Jonathan D Diedrich
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Elizabeth Raetz
- Division of Pediatric Hematology and Oncology, New York University Langone Health, New York, NY, USA
| | - Wenjian Yang
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Qian Dong
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Deqing Pei
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Allen Yeoh
- Khoo Teck Puat–National University Children’s Medical Institute, National University Health System, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Cheng Cheng
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - William E Evans
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Stephen P Hunger
- Department of Pediatrics and Center for Childhood Cancer Research, Children’s Hospital of Philadelphia and the Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Savic
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Mary V Relling
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Mignon L Loh
- Department of Pediatrics, Benioff Children’s Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Jun J Yang
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
11
|
Fulford TS, Grumont R, Wirasinha RC, Ellis D, Barugahare A, Turner SJ, Naeem H, Powell D, Lyons PA, Smith KGC, Scheer S, Zaph C, Klein U, Daley SR, Gerondakis S. c-Rel employs multiple mechanisms to promote the thymic development and peripheral function of regulatory T cells in mice. Eur J Immunol 2021; 51:2006-2026. [PMID: 33960413 DOI: 10.1002/eji.202048900] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 03/12/2021] [Accepted: 05/05/2021] [Indexed: 01/03/2023]
Abstract
The NF-κB transcription factor c-Rel is a critical regulator of Treg ontogeny, controlling multiple points of the stepwise developmental pathway. Here, we found that the thymic Treg defect in c-Rel-deficient (cRel-/- ) mice is quantitative, not qualitative, based on analyses of TCR repertoire and TCR signaling strength. However, these parameters are altered in the thymic Treg-precursor population, which is also markedly diminished in cRel-/- mice. Moreover, c-Rel governs the transcriptional programme of both thymic and peripheral Tregs, controlling a core of genes involved with immune signaling, and separately in the periphery, cell cycle progression. Last, the immune suppressive function of peripheral cRel-/- tTregs is diminished in a lymphopenic model of T cell proliferation and is associated with decreased stability of Foxp3 expression. Collectively, we show that c-Rel is a transcriptional regulator that controls multiple aspects of Treg development, differentiation, and function via distinct mechanisms.
Collapse
Affiliation(s)
- Thomas S Fulford
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Raelene Grumont
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Rushika C Wirasinha
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Darcy Ellis
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Adele Barugahare
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia.,Monash Bioinformatics Platform, School of Biomedical Sciences, Monash University, Melbourne, Australia
| | - Stephen J Turner
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia.,Department of Microbiology, Monash University, Melbourne, Australia
| | - Haroon Naeem
- Monash Bioinformatics Platform, School of Biomedical Sciences, Monash University, Melbourne, Australia
| | - David Powell
- Monash Bioinformatics Platform, School of Biomedical Sciences, Monash University, Melbourne, Australia
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, England, UK.,Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge, England, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, England, UK.,Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge, England, UK
| | - Sebastian Scheer
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Colby Zaph
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Ulf Klein
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James's, University of Leeds, Leeds, LS2 7TF
| | - Stephen R Daley
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Steve Gerondakis
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| |
Collapse
|
12
|
Kober-Hasslacher M, Oh-Strauß H, Kumar D, Soberon V, Diehl C, Lech M, Engleitner T, Katab E, Fernández-Sáiz V, Piontek G, Li H, Menze B, Ziegenhain C, Enard W, Rad R, Böttcher JP, Anders HJ, Rudelius M, Schmidt-Supprian M. c-Rel gain in B cells drives germinal center reactions and autoantibody production. J Clin Invest 2021; 130:3270-3286. [PMID: 32191641 DOI: 10.1172/jci124382] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 03/11/2020] [Indexed: 12/11/2022] Open
Abstract
Single-nucleotide polymorphisms and locus amplification link the NF-κB transcription factor c-Rel to human autoimmune diseases and B cell lymphomas, respectively. However, the functional consequences of enhanced c-Rel levels remain enigmatic. Here, we overexpressed c-Rel specifically in mouse B cells from BAC-transgenic gene loci and demonstrate that c-Rel protein levels linearly dictated expansion of germinal center B (GCB) cells and isotype-switched plasma cells. c-Rel expression in B cells of otherwise c-Rel-deficient mice fully rescued terminal B cell differentiation, underscoring its critical B cell-intrinsic roles. Unexpectedly, in GCB cells transcription-independent regulation produced the highest c-Rel protein levels among B cell subsets. In c-Rel-overexpressing GCB cells this caused enhanced nuclear translocation, a profoundly altered transcriptional program, and increased proliferation. Finally, we provide a link between c-Rel gain and autoimmunity by showing that c-Rel overexpression in B cells caused autoantibody production and renal immune complex deposition.
Collapse
Affiliation(s)
- Maike Kober-Hasslacher
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, Munich, Germany.,Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Hyunju Oh-Strauß
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, Munich, Germany.,Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Dilip Kumar
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Valeria Soberon
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, Munich, Germany.,Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Carina Diehl
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, Munich, Germany.,Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Maciej Lech
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany
| | - Thomas Engleitner
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Molecular Oncology and Functional Genomics and
| | - Eslam Katab
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany.,Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany
| | - Vanesa Fernández-Sáiz
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany.,Department of Medicine III, School of Medicine, Technical University of Munich, Munich, Germany
| | - Guido Piontek
- Institute of Pathology, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany
| | - Hongwei Li
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany.,Department of Informatics, Technical University of Munich, Munich, Germany
| | - Björn Menze
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany.,Department of Informatics, Technical University of Munich, Munich, Germany
| | - Christoph Ziegenhain
- Anthropology and Human Genomics, Department of Biology II, Ludwig-Maximilians-Universität, Martinsried, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Department of Biology II, Ludwig-Maximilians-Universität, Martinsried, Germany
| | - Roland Rad
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Molecular Oncology and Functional Genomics and
| | - Jan P Böttcher
- Institute of Molecular Immunology and Experimental Oncology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Hans-Joachim Anders
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany
| | - Martina Rudelius
- Institute of Pathology, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany
| | - Marc Schmidt-Supprian
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, Munich, Germany.,Max Planck Institute of Biochemistry, Martinsried, Germany.,Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
13
|
Labbozzetta M, Notarbartolo M, Poma P. Can NF-κB Be Considered a Valid Drug Target in Neoplastic Diseases? Our Point of View. Int J Mol Sci 2020; 21:ijms21093070. [PMID: 32349210 PMCID: PMC7246796 DOI: 10.3390/ijms21093070] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/24/2020] [Accepted: 04/25/2020] [Indexed: 02/07/2023] Open
Abstract
Multidrug resistance (MDR), of the innate and acquired types, is one of major problems in treating tumor diseases with a good chance of success. In this review, we examine the key role of nuclear factor-kappa B (NF-κB) to induce MDR in three tumor models characterized precisely by innate or acquired MDR, in particular triple negative breast cancer (TNBC), hepatocellular carcinoma (HCC), and acute myeloid leukemia (AML). We also present different pharmacological approaches that our group have employed to reduce the expression/activation of this transcriptional factor and thus to restore chemo-sensitivity. Finally, we examine the latest scientific evidence found by other groups, the most significant clinical trials regarding NF-κB, and new perspectives on the possibility to consider this transcriptional factor a valid drug target in neoplastic diseases.
Collapse
|
14
|
Abstract
Aim: PTPIP51 interacts with NFκB signaling at the RelA and IκB level. NFκB signaling is linked to the initiation, progression and metastasis of breast cancer. Her2-amplified breast cancer cells frequently display activation of the NFκB signaling. We aimed to clarify the effects of NFκB inhibition on the NFκB- and MAPK-related interactome of PTPIP51 and cell viability in HaCat cells and SKBR3 cells. Results: IKK-16 selectively reduced cell viability in SKBR3 cells. PDTC induced a formation of the Raf1/14-3-3/PTPIP51 complex in SKBR3 cells, indicating a shift of PTPIP51 into MAPK signaling. Conclusion: IKK-16 selectively inhibits cell viability of SKBR3 cells. In addition, PTPIP51 might serve as the mediator between NFκB signaling and the MAPK pathway in SKBR3. Breast cancer is the most common cancerous disease among women. Prognosis and therapy of breast cancer depends on the expression of hormone and surface receptors such as Her2, which promote tumor growth and invasion via activation of downstream signaling pathways. NFκB signaling represents a downstream signaling pathway that can be activated by Her2. In this study, we demonstrated that inhibition of NFκB signaling with IKK-16 reduces cell viability in breast cancer cells with amplified Her2. Furthermore, we identified PTPIP51 as a potential mediator of crosstalks between the MAPK pathway and NFκB signaling. This signaling pathway could therefore be a target for future drug development.
Collapse
|
15
|
Miao Y, Medeiros LJ, Li Y, Li J, Young KH. Genetic alterations and their clinical implications in DLBCL. Nat Rev Clin Oncol 2019; 16:634-652. [PMID: 31127191 DOI: 10.1038/s41571-019-0225-1] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Diffuse large B cell lymphoma (DLBCL) is a highly heterogeneous lymphoid neoplasm with variations in gene expression profiles and genetic alterations, which lead to substantial variations in clinical course and response to therapy. The advent of high-throughput genome sequencing platforms, and especially whole-exome sequencing, has helped to define the genetic landscape of DLBCL. In the past 10 years, these studies have identified many genetic alterations in DLBCL, some of which are specific to B cell lymphomas, whereas others can also be observed in other types of cancer. These aberrations result in altered activation of a wide range of signalling pathways and other cellular processes, including those involved in B cell differentiation, B cell receptor signalling, activation of the NF-κB pathway, apoptosis and epigenetic regulation. Further elaboration of the genetics of DLBCL will not only improve our understanding of disease pathogenesis but also provide further insight into disease classification, prognostication and therapeutic targets. In this Review, we describe the current understanding of the prevalence and causes of specific genetic alterations in DLBCL and their role in disease development and progression. We also summarize the available clinical data on therapies designed to target the aberrant pathways driven by these alterations.
Collapse
Affiliation(s)
- Yi Miao
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yong Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jianyong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Ken H Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Graduate School of Biomedical Sciences, University of Texas Health Science Center, Houston, TX, USA.
| |
Collapse
|
16
|
Konczalla L, Perez DR, Wenzel N, Wolters-Eisfeld G, Klemp C, Lüddeke J, Wolski A, Landschulze D, Meier C, Buchholz A, Yao D, Hofmann BT, Graß JK, Spriestersbach SL, Grupp K, Schumacher U, Betzel C, Kapis S, Nuguid T, Steinberg P, Püschel K, Sauter G, Bockhorn M, Uzunoglu FG, Izbicki JR, Güngör C, El Gammal AT. Biperiden and mepazine effectively inhibit MALT1 activity and tumor growth in pancreatic cancer. Int J Cancer 2019; 146:1618-1630. [PMID: 31291468 DOI: 10.1002/ijc.32567] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/31/2019] [Accepted: 06/21/2019] [Indexed: 12/14/2022]
Abstract
MALT1 is a key mediator of NF-κB signaling and a main driver of B-cell lymphomas. Remarkably, MALT1 is expressed in the majority of pancreatic ductal adenocarcinomas (PDACs) as well, but absent from normal exocrine pancreatic tissue. Following, MALT1 shows off to be a specific target in cancer cells of PDAC without affecting regular pancreatic cells. Therefore, we studied the impact of pharmacological MALT1 inhibition in pancreatic cancer and showed promising effects on tumor progression. Mepazine (Mep), a phenothiazine derivative, is a known potent MALT1 inhibitor. Newly, we described that biperiden (Bip) is a potent MALT1 inhibitor with even less pharmacological side effects. Thus, Bip is a promising drug leading to reduced proliferation and increased apoptosis in PDAC cells in vitro and in vivo. By compromising MALT1 activity, nuclear translocation of c-Rel is prevented. c-Rel is critical for NF-κB-dependent inhibition of apoptosis. Hence, off-label use of Bip or Mep represents a promising new therapeutic approach to PDAC treatment. Regularly, the Anticholinergicum Bip is used to treat neurological side effects of Phenothiazines, like extrapyramidal symptoms.
Collapse
Affiliation(s)
- Leonie Konczalla
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel R Perez
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nadine Wenzel
- Institute for Food Toxicology and Analytical Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Gerrit Wolters-Eisfeld
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Clarissa Klemp
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johanna Lüddeke
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Annika Wolski
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dirk Landschulze
- Institute of Organic Chemistry, University of Hamburg, Hamburg, Germany
| | - Chris Meier
- Institute of Organic Chemistry, University of Hamburg, Hamburg, Germany
| | - Anika Buchholz
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dichao Yao
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bianca T Hofmann
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia K Graß
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah L Spriestersbach
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Grupp
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Betzel
- Laboratory for Structural Biology of Infection and Inflammation, Department of Chemistry, c/o DESY, University of Hamburg, Hamburg, Germany
| | - Svetlana Kapis
- Laboratory for Structural Biology of Infection and Inflammation, Department of Chemistry, c/o DESY, University of Hamburg, Hamburg, Germany
| | - Theresa Nuguid
- Laboratory for Structural Biology of Infection and Inflammation, Department of Chemistry, c/o DESY, University of Hamburg, Hamburg, Germany
| | - Pablo Steinberg
- Institute for Food Toxicology and Analytical Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Klaus Püschel
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximillian Bockhorn
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Faik G Uzunoglu
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob R Izbicki
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cenap Güngör
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander T El Gammal
- Department of General, Visceral, and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
17
|
Gamboa-Cedeño AM, Castillo M, Xiao W, Waldmann TA, Ranuncolo SM. Alternative and canonical NF-kB pathways DNA-binding hierarchies networks define Hodgkin lymphoma and Non-Hodgkin diffuse large B Cell lymphoma respectively. J Cancer Res Clin Oncol 2019; 145:1437-1448. [PMID: 30941572 PMCID: PMC8317045 DOI: 10.1007/s00432-019-02909-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/25/2019] [Indexed: 12/22/2022]
Abstract
PURPOSE Despite considerable evidence that supports the NF-kB role in the immune system and lymphomagenesis, it is unclear whether specific NF-kB dimers control a particular set of genes that account for their biological functions. Our previous work showed that Hodgkin Lymphoma (HL) is unique, among germinal center (GC)-derived lymphomas, with respect to its dependency on Rel-B to survive. In contrast, diffuse large B-Cell lymphoma (DLBCL) including both Activated B-Cell-Like and Germinal Center B-Cell-Like, requires cREL and Rel-A to survive and it is not affected by Rel-B depletion. These findings highlighted the activity of specific NF-kB subunits in different GC-derived lymphomas. METHODS Sequenced chromatin immunoprecipitated DNA fragments (ChIP-Seq) analysis revealed an extensive NF-kB DNA-binding network in DLBCL and HL. The ChIP-Seq data was merged with microarray analysis following the Rel-A, Rel-B or cRel knockdown to determine effectively regulated genes. RESULTS Downstream target analysis showed enrichment for cell cycle control, among other signatures. Rel-B and cRel controlled different genes within the same signature in HL and DLBCL, respectively. BCL2 was exclusively controlled by Rel-B in HL. Both mRNA and protein levels decreased following Rel-B depletion meanwhile there was no change upon cRel knock-down. BCL2 exogenous expression partially rescued the death induced by decreased Rel-B in HL cells. CONCLUSION The Rel-B hierarchical network defined HL and the cRel hierarchical network characterized DLBCL. Each Rel member performs specific functions in distinct GC-derived lymphomas. This result should be considered for the development of targeted therapies that are aimed to selectively inhibit individual NF-kB dimers.
Collapse
Affiliation(s)
- Angélica María Gamboa-Cedeño
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET-Instituto Universitario del Hospital Italiano-Hospital Italiano de Buenos Aires, Potosí 4240 C.P., C1183AEG, Buenos Aires, Argentina
| | - Mariángeles Castillo
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET-Instituto Universitario del Hospital Italiano-Hospital Italiano de Buenos Aires, Potosí 4240 C.P., C1183AEG, Buenos Aires, Argentina
| | - Wenming Xiao
- Center for Information Technology, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research (CCR), NCI-NIH, Bethesda, MD, USA
| | - Stella Maris Ranuncolo
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET-Instituto Universitario del Hospital Italiano-Hospital Italiano de Buenos Aires, Potosí 4240 C.P., C1183AEG, Buenos Aires, Argentina.
- Departamento de Histología y Biología Celular, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.
- National Council of Scientific and Technological Research (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
18
|
Ramsey KM, Chen W, Marion JD, Bergqvist S, Komives EA. Exclusivity and Compensation in NFκB Dimer Distributions and IκB Inhibition. Biochemistry 2019; 58:2555-2563. [PMID: 31033276 DOI: 10.1021/acs.biochem.9b00008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The NFκB transcription factor family members RelA, p50, and cRel form homo- and heterodimers that are inhibited by IκBα, IκBβ, and IκBε. These NFκB family members have diverse biological functions, and their expression profiles differ, leading to different concentrations in different tissue types. Here we present definitive biophysical measurements of the NFκB dimer affinities and inhibitor affinities to better understand dimer exchange and how the presence of inhibitors may alter the equilibrium concentrations of NFκB dimers in the cellular context. Fluorescence anisotropy binding experiments were performed at low concentrations to mimic intracellular concentrations. We report binding affinities much stronger than those that had been previously reported by non-equilibrium gel shift and analytical ultracentrifugation assays. The results reveal a wide range of NFκB dimer affinities and a strong preference of each IκB for a small subset of NFκB dimers. Once the preferred IκB is bound, dimer exchange no longer occurs over a period of days. A mathematical model of the cellular distribution of these canonical NFκB transcription factors based on the revised binding affinities recapitulates intracellular observations and provides simple, precise explanations for observed cellular phenomena.
Collapse
Affiliation(s)
- Kristen M Ramsey
- Department of Chemistry and Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92092-0378 , United States
| | - Wei Chen
- Department of Chemistry and Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92092-0378 , United States
| | - James D Marion
- Department of Chemistry and Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92092-0378 , United States
| | - Simon Bergqvist
- Department of Chemistry and Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92092-0378 , United States
| | - Elizabeth A Komives
- Department of Chemistry and Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92092-0378 , United States
| |
Collapse
|
19
|
The impact of NF-κB signaling on pathogenesis and current treatment strategies in multiple myeloma. Blood Rev 2019; 34:56-66. [DOI: 10.1016/j.blre.2018.11.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 11/14/2018] [Accepted: 11/22/2018] [Indexed: 12/13/2022]
|
20
|
Grewal RK, Chetty M, Abayomi EA, Tomuleasa C, Fromm JR. Use of flow cytometry in the phenotypic diagnosis of hodgkin's lymphoma. CYTOMETRY PART B-CLINICAL CYTOMETRY 2018; 96:116-127. [PMID: 30350336 DOI: 10.1002/cyto.b.21724] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/18/2018] [Accepted: 06/26/2018] [Indexed: 12/22/2022]
Abstract
Hodgkin's lymphoma (HL) has a unique immunophenotype derived from immunohistochemistry (positive for CD15, CD30, and Pax-5; negative for CD3, CD20 in most cases, and CD45). The knowledge gained over recent years enables better diagnosis, prognosis, and treatment of HL. Flow cytometry as a tool for the diagnosis of classic HL has not been useful in the past due to the difficulty in isolating Reed-Sternberg cells as they are admixed in a rich inflammatory background which consists mainly of T cells, B cells, eosinophils, histiocytes, and plasma cells. However, in the recent past, several studies have tried to identify Reed-Sternberg cells using flow cytometry on fine needle aspiration or tissue biopsy of lymph nodes to confirm or supplement immunohistochemistry staining in diagnosis. Newer and more sensitive tools such as flow cytometry can be used for diagnosis, technology that may have been difficult in the past for diagnosis of this lymphoma subtype. Using flow cytometry, diagnosis is faster and could lead to point-of-care technology especially where we have typical immunophenotype signatures. © 2018 International Clinical Cytometry Society.
Collapse
Affiliation(s)
- Ravnit-Kaur Grewal
- MBCHB South African National Bioinformatics Institute, University of the Western Cape, Bellville, South Africa
| | - Manogari Chetty
- Department of Oral and Molecular Biology, Faculty of Dentistry, University of the Western Cape, Cape Town, South Africa
| | | | - Ciprian Tomuleasa
- Department of Hematology/Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy-Ion Chiricuta Oncology Institute, Cluj Napoca, Romania
| | - Jonathan R Fromm
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
21
|
Gao HY, Wang W, Luo XG, Jiang YF, He X, Xu P, Chen X, Li XY. Screening of prognostic risk microRNAs for acute myeloid leukemia. ACTA ACUST UNITED AC 2018; 23:747-755. [PMID: 29781401 DOI: 10.1080/10245332.2018.1475860] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Objectives This study aimed to investigate the risk miRNAs (microRNAs) for AML (acute myeloid leukemia) prognosis and related regulatory mechanisms. Methods MiRNA and gene expression data, as well as clinical data of 176 patients were first downloaded from TCGA. Then miRNAs and genes significantly affecting the survival time based on KM survival curve were identified using Log Rank test. Next, COX proportional-hazard regression analysis was performed to screen the risk miRNAs (P-value < 0.05). Common genes from survival analysis and predicted by miRWalk were used to construct the miRNA regulatory network with the risk miRNAs. Finally, a protein-protein interaction (PPI) network was constructed, as well as functional annotation and pathway enrichment analysis. Results The survival analysis revealed 33 miRNAs and 1,377 genes significantly affecting the survival time. HR values of nine miRNAs (up-regulated hsa-mir-606, 520a, 137, 362, 599, 600, 202, 639and down-regulated 502) were either >1 or <1. The miRNA regulatory network contained 477 nodes and 944 edges. The top ten genes of the constructed PPI network were EGFR, EIF4G1, REL, TOP1, COL14A1, HDAC3, MRPL49, PSMA2, TOP2A and VCAM1 successively. According to pathway enrichment analysis, 6 KEGG pathways and 6 REACTOME pathways were obtained respectively. Conclusion Up-regulated hsa-mir-520a, 599, 606, 137 and 362 may increase the prognostic risk for AML patients via regulating the expression of corresponding target genes, especially COL14A1, HDAC3, REL, EGFR, PSMA2, EIF4G1, MRPL49 and TOP1.
Collapse
Affiliation(s)
- Hai-Yan Gao
- a Department of Hematology , The Second Affiliated Hospital of Harbin Medical University , Harbin , People's Republic of China
| | - Wei Wang
- a Department of Hematology , The Second Affiliated Hospital of Harbin Medical University , Harbin , People's Republic of China
| | - Xin-Guo Luo
- b Department of Hematology , Jinhua People's Hospital , Jinhua , People's Republic of China
| | - Yong-Fang Jiang
- a Department of Hematology , The Second Affiliated Hospital of Harbin Medical University , Harbin , People's Republic of China
| | - Xin He
- a Department of Hematology , The Second Affiliated Hospital of Harbin Medical University , Harbin , People's Republic of China
| | - Ping Xu
- a Department of Hematology , The Second Affiliated Hospital of Harbin Medical University , Harbin , People's Republic of China
| | - Xi Chen
- a Department of Hematology , The Second Affiliated Hospital of Harbin Medical University , Harbin , People's Republic of China
| | - Xiao-Yun Li
- a Department of Hematology , The Second Affiliated Hospital of Harbin Medical University , Harbin , People's Republic of China
| |
Collapse
|
22
|
Moore D, Walker SI, Levin M. Cancer as a disorder of patterning information: computational and biophysical perspectives on the cancer problem. CONVERGENT SCIENCE PHYSICAL ONCOLOGY 2017. [DOI: 10.1088/2057-1739/aa8548] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
23
|
Mohammadi SM, Mohammadnejad D, Hosseinpour Feizi AA, Movassaghpour AA, Montazersaheb S, Nozad Charoudeh H. Inhibition of c-REL using siRNA increased apoptosis and decreased proliferation in pre-B ALL blasts: Therapeutic implications. Leuk Res 2017; 61:53-61. [PMID: 28892661 DOI: 10.1016/j.leukres.2017.08.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 08/18/2017] [Accepted: 08/21/2017] [Indexed: 01/03/2023]
Abstract
The c-Rel transcription factor is a unique member of the NF-kB family that has a role in apoptosis, proliferation and cell survival. Overexpression of c-Rel is detected in many human B cell tumors, including B-cell leukemia and several cancers. The study aimed to investigate the effects of c-Rel siRNA on the proliferation and apoptosis of relapsed pre-B acute leukemia cells. The c-Rel siRNA was transfected into Leukemia cells using an Amaxa cell line Nucleofector kit L (Lonza). Quantitative real-time RT-PCR (qRT-PCR) and western blot were done to measure the expression levels of mRNA and protein, respectively. The flow cytometry was used to analyze the effect of c-Rel siRNA on the apoptosis and proliferation of Leukemia cells. Observed c-Rel expression in the 5 pre-B Acute lymphoblastic leukemia (ALL) patients were higher than the normal cells. The c-Rel siRNA transfection significantly blocked the expression of c-Rel mRNA in a time-dependent manner, leading to a strong growth inhibition and enhanced apoptosis (P<0.05). Our results demonstrated that c-Rel plays a fundamental role in the survival. Therefore, c-Rel can be considered as an attractive target for gene therapy in ALL patients. Also siRNA-mediated silencing of this gene may be a novel strategy in ALL treatment.
Collapse
Affiliation(s)
| | - Daryosh Mohammadnejad
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Anatomical Sciences Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | | | |
Collapse
|
24
|
Slotta C, Schlüter T, Ruiz-Perera LM, Kadhim HM, Tertel T, Henkel E, Hübner W, Greiner JFW, Huser T, Kaltschmidt B, Kaltschmidt C. CRISPR/Cas9-mediated knockout of c-REL in HeLa cells results in profound defects of the cell cycle. PLoS One 2017; 12:e0182373. [PMID: 28767691 PMCID: PMC5540532 DOI: 10.1371/journal.pone.0182373] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/17/2017] [Indexed: 12/17/2022] Open
Abstract
Cervical cancer is the fourth common cancer in women resulting worldwide in 266,000 deaths per year. Belonging to the carcinomas, new insights into cervical cancer biology may also have great implications for finding new treatment strategies for other kinds of epithelial cancers. Although the transcription factor NF-κB is known as a key player in tumor formation, the relevance of its particular subunits is still underestimated. Here, we applied CRISPR/Cas9n-mediated genome editing to successfully knockout the NF-κB subunit c-REL in HeLa Kyoto cells as a model system for cervical cancers. We successfully generated a homozygous deletion in the c-REL gene, which we validated using sequencing, qPCR, immunocytochemistry, western blot analysis, EMSA and analysis of off-target effects. On the functional level, we observed the deletion of c-REL to result in a significantly decreased cell proliferation in comparison to wildtype (wt) without affecting apoptosis. The impaired proliferative behavior of c-REL-/- cells was accompanied by a strongly decreased amount of the H2B protein as well as a significant delay in the prometaphase of mitosis compared to c-REL+/+ HeLa Kyoto cells. c-REL-/- cells further showed significantly decreased expression levels of c-REL target genes in comparison to wt. In accordance to our proliferation data, we observed the c-REL knockout to result in a significantly increased resistance against the chemotherapeutic agents 5-Fluoro-2'-deoxyuridine (5-FUDR) and cisplatin. In summary, our findings emphasize the importance of c-REL signaling in a cellular model of cervical cancer with direct clinical implications for the development of new treatment strategies.
Collapse
Affiliation(s)
- Carsten Slotta
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
| | - Thomas Schlüter
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
| | | | | | - Tobias Tertel
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
| | - Elena Henkel
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
| | - Wolfgang Hübner
- Biomolecular Photonics, University of Bielefeld, Bielefeld, Germany
| | | | - Thomas Huser
- Biomolecular Photonics, University of Bielefeld, Bielefeld, Germany
| | - Barbara Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- AG Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany
| | | |
Collapse
|
25
|
Yao Y, Vasoya D, Kgosana L, Smith LP, Gao Y, Wang X, Watson M, Nair V. Activation of gga-miR-155 by reticuloendotheliosis virus T strain and its contribution to transformation. J Gen Virol 2017; 98:810-820. [PMID: 28113043 PMCID: PMC5657028 DOI: 10.1099/jgv.0.000718] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The v-rel oncoprotein encoded by reticuloendotheliosis virus T strain (Rev-T) is a member of the rel/NF-κB family of transcription factors capable of transformation of primary chicken spleen and bone marrow cells. Rapid transformation of avian haematopoietic cells by v-rel occurs through a process of deregulation of multiple protein-encoding genes through its direct effect on their promoters. More recently, upregulation of oncogenic miR-155 and its precursor pre-miR-155 was demonstrated in both Rev-T-infected chicken embryo fibroblast cultures and Rev-T-induced B-cell lymphomas. Through electrophoresis mobility shift assay and reporter analysis on the gga-miR-155 promoter, we showed that the v-rel-induced miR-155 overexpression occurred by the direct binding to one of the putative NF-κB binding sites. Using the v-rel-induced transformation model on chicken embryonic splenocyte cultures, we could demonstrate a dynamic increase in miR-155 levels during the transformation. Transcriptome profiles of lymphoid cells transformed by v-rel showed upregulation of miR-155 accompanied by downregulation of a number of putative miR-155 targets such as Pu.1 and CEBPβ. We also showed that v-rel could rescue the suppression of miR-155 expression observed in Marek's disease virus (MDV)-transformed cell lines, where its functional viral homologue MDV-miR-M4 is overexpressed. Demonstration of gene expression changes affecting major molecular pathways, including organismal injury and cancer in avian macrophages transfected with synthetic mature miR-155, underlines its potential direct role in transformation. Our study suggests that v-rel-induced transformation involves a complex set of events mediated by the direct activation of NF-κB targets, together with inhibitory effects on microRNA targets.
Collapse
Affiliation(s)
- Yongxiu Yao
- Avian Viral Disease Programme & UK-China Centre of Excellence on Avian Disease Research, The Pirbright Institute, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, UK
| | - Deepali Vasoya
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush EH25 9RG, UK
| | - Lydia Kgosana
- Avian Viral Disease Programme & UK-China Centre of Excellence on Avian Disease Research, The Pirbright Institute, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, UK
| | - Lorraine P Smith
- Avian Viral Disease Programme & UK-China Centre of Excellence on Avian Disease Research, The Pirbright Institute, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, UK
| | - Yulong Gao
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Xiaomei Wang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Mick Watson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush EH25 9RG, UK
| | - Venugopal Nair
- Avian Viral Disease Programme & UK-China Centre of Excellence on Avian Disease Research, The Pirbright Institute, Pirbright, Ash Road, Guildford, Surrey GU24 0NF, UK
| |
Collapse
|
26
|
Fallahi S, Mohammadi SM, Tayefi Nasrabadi H, Alihemmati A, Samadi N, Gholami S, Shanehbandi D, Nozad Charoudeh H. Impact of C-rel inhibition of cord blood-derived B-, T-, and NK cells. J Immunotoxicol 2017; 14:15-22. [DOI: 10.1080/1547691x.2016.1250849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Shirin Fallahi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyede Momeneh Mohammadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Tayefi Nasrabadi
- Tissue Engineering Research Group, Advanced Research School, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Alihemmati
- Tissue Engineering Research Group, Advanced Research School, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naser Samadi
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanaz Gholami
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
27
|
Osorio FG, Soria-Valles C, Santiago-Fernández O, Freije JMP, López-Otín C. NF-κB signaling as a driver of ageing. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 326:133-74. [PMID: 27572128 DOI: 10.1016/bs.ircmb.2016.04.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
NF-κB signaling exerts essential roles in immunity and cellular stress responses, regulating many functions related with organism innate defense. Besides, NF-κB altered signaling has been causally linked to ageing and diverse pathological conditions. We discuss herein the functional involvement of this signaling pathway in ageing, visiting recent experimental evidence about NF-κB activation in this complex process, its functional consequences and the novel biological functions raised from these works. Moreover, we discuss ageing intervention strategies based on NF-κB inhibition, which have demonstrated to be effective at delaying and even reverting different ageing manifestations in human and mouse models of both normal and accelerated ageing. Altogether, the current evidence supports that NF-κB activation constitutes a driving force of the ageing process and a preferential target for rejuvenation-aimed approaches.
Collapse
Affiliation(s)
- F G Osorio
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Oviedo, Oviedo, Spain
| | - C Soria-Valles
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Oviedo, Oviedo, Spain
| | - O Santiago-Fernández
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Oviedo, Oviedo, Spain
| | - J M P Freije
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Oviedo, Oviedo, Spain
| | - C López-Otín
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Oviedo, Oviedo, Spain.
| |
Collapse
|
28
|
Raymond A, Liu B, Liang H, Wei C, Guindani M, Lu Y, Liang S, St John LS, Molldrem J, Nagarajan L. A role for BMP-induced homeobox gene MIXL1 in acute myelogenous leukemia and identification of type I BMP receptor as a potential target for therapy. Oncotarget 2015; 5:12675-93. [PMID: 25544748 PMCID: PMC4350356 DOI: 10.18632/oncotarget.2564] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 10/02/2014] [Indexed: 01/03/2023] Open
Abstract
Mesoderm Inducer in Xenopus Like1 (MIXL1), a paired-type homeobox transcription factor induced by TGF-β family of ligands is required for early embryonic specification of mesoderm and endoderm. Retrovirally transduced Mixl1 is reported to induce acute myelogenous leukemia (AML) with a high penetrance. But the mechanistic underpinnings of MIXL1 mediated leukemogenesis are unknown. Here, we establish the protooncogene c-REL to be a transcriptional target of MIXL1 by genome wide chromatin immune precipitation. Accordingly, expression of c-REL and its downstream targets BCL2L1 and BCL2A2 are elevated in MIXL1 expressing cells. Notably, MIXL1 regulates c-REL through a zinc finger binding motif, potentially by a MIXL1–Zinc finger protein transcriptional complex. Furthermore, MIXL1 expression is detected in the cancer genome atlas (TCGA) AML samples in a pattern mutually exclusive from that of HOXA9, CDX2 and HLX suggesting the existence of a core, yet distinct HOX transcriptional program. Finally, we demonstrate MIXL1 to be induced by BMP4 and not TGF-β in primary human hematopoietic stem and progenitor cells. Consequently, MIXL1 expressing AML cells are preferentially sensitive to the BMPR1 kinase inhibitor LDN-193189. These findings support the existence of a novel MIXL1-c REL mediated survival axis in AML that can be targeted by BMPR1 inhibitors. (MIXL1- human gene, Mixl1- mouse ortholog, MIXL1- protein)
Collapse
Affiliation(s)
- Aaron Raymond
- Department of Genetics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. Graduate Program in Genes and Development, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bin Liu
- Department of Genetics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. Center for Cancer Genetics and Genomics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hong Liang
- Department of Genetics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Caimiao Wei
- Department of Biostatistics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michele Guindani
- Department of Biostatistics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yue Lu
- Dept. of Leukemia, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. Dept. of Molecular Carcinogenesis, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shoudan Liang
- Dept. of Bioinformatics and Computational Biology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lisa S St John
- Section of Transplantation Immunology, Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeff Molldrem
- Section of Transplantation Immunology, Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lalitha Nagarajan
- Department of Genetics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. Graduate Program in Genes and Development, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. Center for Cancer Genetics and Genomics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. Dept. of Leukemia, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. Graduate Program in Human Molecular Genetics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. Center for Stem cell and Developmental biology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
29
|
ATM deficiency promotes development of murine B-cell lymphomas that resemble diffuse large B-cell lymphoma in humans. Blood 2015; 126:2291-301. [PMID: 26400962 DOI: 10.1182/blood-2015-06-654749] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 09/19/2015] [Indexed: 12/17/2022] Open
Abstract
The serine-threonine kinase ataxia-telangiectasia mutated (ATM) plays a central role in maintaining genomic integrity. In mice, ATM deficiency is exclusively associated with T-cell lymphoma development, whereas B-cell tumors predominate in human ataxia-telangiectasia patients. We demonstrate in this study that when T cells are removed as targets for lymphomagenesis and as mediators of immune surveillance, ATM-deficient mice exclusively develop early-onset immunoglobulin M(+) B-cell lymphomas that do not transplant to immunocompetent mice and that histologically and genetically resemble the activated B cell-like (ABC) subset of human diffuse large B-cell lymphoma (DLBCL). These B-cell lymphomas show considerable chromosomal instability and a recurrent genomic amplification of a 4.48-Mb region on chromosome 18 that contains Malt1 and is orthologous to a region similarly amplified in human ABC DLBCL. Of importance, amplification of Malt1 in these lymphomas correlates with their dependence on nuclear factor (NF)-κB, MALT1, and B-cell receptor (BCR) signaling for survival, paralleling human ABC DLBCL. Further, like some human ABC DLBCLs, these mouse B-cell lymphomas also exhibit constitutive BCR-dependent NF-κB activation. This study reveals that ATM protects against development of B-cell lymphomas that model human ABC DLBCL and identifies a potential role for T cells in preventing the emergence of these tumors.
Collapse
|
30
|
Abstract
NF-κB comprises a family of five transcription factors that form distinct protein complexes, which bind to consensus DNA sequences at promoter regions of responsive genes regulating cellular processes. The past three decades have witnessed remarkable progress in understanding the NF-κB signaling pathway in physiologic and pathologic conditions. The role of NF-κB in human cancer initiation, development, metastasis, and resistance to treatment has drawn particular attention. A significant number of human cancers have constitutive NF-κB activity due to the inflammatory microenvironment and various oncogenic mutations. NF-κB activity not only promotes tumor cells' proliferation, suppresses apoptosis, and attracts angiogenesis, but it also induces epithelial-mesenchymal transition, which facilitates distant metastasis. In certain circumstances, NF-κB activation may also remodel local metabolism and anergize the immune system to favor tumor growth. Suppression of NF-κB in myeloid cells or tumor cells usually leads to tumor regression, which makes the NF-κB pathway a promising therapeutic target. However, because of its vital role in various biologic activities, components of the NF-κB pathway need to be carefully selected and evaluated to design targeted therapies.
Collapse
Affiliation(s)
- Yifeng Xia
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
| | - Shen Shen
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
| | - Inder M Verma
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California.
| |
Collapse
|
31
|
Himmelstein DS, Baranzini SE. Heterogeneous Network Edge Prediction: A Data Integration Approach to Prioritize Disease-Associated Genes. PLoS Comput Biol 2015; 11:e1004259. [PMID: 26158728 PMCID: PMC4497619 DOI: 10.1371/journal.pcbi.1004259] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/26/2015] [Indexed: 12/13/2022] Open
Abstract
The first decade of Genome Wide Association Studies (GWAS) has uncovered a wealth of disease-associated variants. Two important derivations will be the translation of this information into a multiscale understanding of pathogenic variants and leveraging existing data to increase the power of existing and future studies through prioritization. We explore edge prediction on heterogeneous networks—graphs with multiple node and edge types—for accomplishing both tasks. First we constructed a network with 18 node types—genes, diseases, tissues, pathophysiologies, and 14 MSigDB (molecular signatures database) collections—and 19 edge types from high-throughput publicly-available resources. From this network composed of 40,343 nodes and 1,608,168 edges, we extracted features that describe the topology between specific genes and diseases. Next, we trained a model from GWAS associations and predicted the probability of association between each protein-coding gene and each of 29 well-studied complex diseases. The model, which achieved 132-fold enrichment in precision at 10% recall, outperformed any individual domain, highlighting the benefit of integrative approaches. We identified pleiotropy, transcriptional signatures of perturbations, pathways, and protein interactions as influential mechanisms explaining pathogenesis. Our method successfully predicted the results (with AUROC = 0.79) from a withheld multiple sclerosis (MS) GWAS despite starting with only 13 previously associated genes. Finally, we combined our network predictions with statistical evidence of association to propose four novel MS genes, three of which (JAK2, REL, RUNX3) validated on the masked GWAS. Furthermore, our predictions provide biological support highlighting REL as the causal gene within its gene-rich locus. Users can browse all predictions online (http://het.io). Heterogeneous network edge prediction effectively prioritized genetic associations and provides a powerful new approach for data integration across multiple domains. For complex human diseases, identifying the genes harboring susceptibility variants has taken on medical importance. Disease-associated genes provide clues for elucidating disease etiology, predicting disease risk, and highlighting therapeutic targets. Here, we develop a method to predict whether a given gene and disease are associated. To capture the multitude of biological entities underlying pathogenesis, we constructed a heterogeneous network, containing multiple node and edge types. We built on a technique developed for social network analysis, which embraces disparate sources of data to make predictions from heterogeneous networks. Using the compendium of associations from genome-wide studies, we learned the influential mechanisms underlying pathogenesis. Our findings provide a novel perspective about the existence of pervasive pleiotropy across complex diseases. Furthermore, we suggest transcriptional signatures of perturbations are an underutilized resource amongst prioritization approaches. For multiple sclerosis, we demonstrated our ability to prioritize future studies and discover novel susceptibility genes. Researchers can use these predictions to increase the statistical power of their studies, to suggest the causal genes from a set of candidates, or to generate evidence-based experimental hypothesis.
Collapse
Affiliation(s)
- Daniel S. Himmelstein
- Biological & Medical Informatics, University of California, San Francisco, San Francisco, California, United States of America
| | - Sergio E. Baranzini
- Biological & Medical Informatics, University of California, San Francisco, San Francisco, California, United States of America
- Department of Neurology, University of California, San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
32
|
Laguna T, Notario L, Pippa R, Fontela MG, Vázquez BN, Maicas M, Aguilera-Montilla N, Corbí ÁL, Odero MD, Lauzurica P. New insights on the transcriptional regulation of CD69 gene through a potent enhancer located in the conserved non-coding sequence 2. Mol Immunol 2015; 66:171-9. [PMID: 25801305 DOI: 10.1016/j.molimm.2015.02.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/25/2015] [Accepted: 02/27/2015] [Indexed: 01/03/2023]
Abstract
The CD69 type II C-type lectin is one of the earliest indicators of leukocyte activation acting in lymphocyte migration and cytokine secretion. CD69 expression in hematopoietic lineage undergoes rapid changes depending on the cell-lineage, the activation state or the localization of the cell where it is expressed, suggesting a complex and tightly controlled regulation. Here we provide new insights on the transcriptional regulation of CD69 gene in mammal species. Through in silico studies, we analyzed several regulatory features of the 4 upstream conserved non-coding sequences (CNS 1-4) previously described, confirming a major function of CNS2 in the transcriptional regulation of CD69. In addition, multiple transcription binding sites are identified in the CNS2 region by DNA cross-species conservation analysis. By functional approaches we defined a core region of 226bp located within CNS2 as the main enhancer element of CD69 transcription in the hematopoietic cells analyzed. By chromatin immunoprecipitation, binding of RUNX1 to the core-CNS2 was shown in a T cell line. In addition, we found an activating but not essential role of RUNX1 in CD69 gene transcription by site-directed mutagenesis and RNA silencing, probably through the interaction with this potent enhancer specifically in the hematopoietic lineage. In summary, in this study we contribute with new evidences to the landscape of the transcriptional regulation of the CD69 gene.
Collapse
Affiliation(s)
- Teresa Laguna
- Activación Immunológica Group, Carlos III National Health Institute (ISCIII), Majadahonda, Spain
| | - Laura Notario
- Activación Immunológica Group, Carlos III National Health Institute (ISCIII), Majadahonda, Spain
| | - Raffaella Pippa
- Laboratory of Genetics, Division of Oncology, CIMA, University of Navarra, Pamplona, Spain
| | - Miguel G Fontela
- Activación Immunológica Group, Carlos III National Health Institute (ISCIII), Majadahonda, Spain
| | - Berta N Vázquez
- Activación Immunológica Group, Carlos III National Health Institute (ISCIII), Majadahonda, Spain
| | - Miren Maicas
- Laboratory of Genetics, Division of Oncology, CIMA, University of Navarra, Pamplona, Spain
| | - Noemí Aguilera-Montilla
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Ángel L Corbí
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - María D Odero
- Laboratory of Genetics, Division of Oncology, CIMA, University of Navarra, Pamplona, Spain
| | - Pilar Lauzurica
- Activación Immunológica Group, Carlos III National Health Institute (ISCIII), Majadahonda, Spain.
| |
Collapse
|
33
|
Li F, Zhang J, Arfuso F, Chinnathambi A, Zayed ME, Alharbi SA, Kumar AP, Ahn KS, Sethi G. NF-κB in cancer therapy. Arch Toxicol 2015; 89:711-31. [PMID: 25690730 DOI: 10.1007/s00204-015-1470-4] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/05/2015] [Indexed: 02/06/2023]
Abstract
The transcription factor nuclear factor kappa B (NF-κB) has attracted increasing attention in the field of cancer research from last few decades. Aberrant activation of this transcription factor is frequently encountered in a variety of solid tumors and hematological malignancies. NF-κB family members and their regulated genes have been linked to malignant transformation, tumor cell proliferation, survival, angiogenesis, invasion/metastasis, and therapeutic resistance. In this review, we highlight the diverse molecular mechanism(s) by which the NF-κB pathway is constitutively activated in different types of human cancers, and the potential role of various oncogenic genes regulated by this transcription factor in cancer development and progression. Additionally, various pharmacological approaches employed to target the deregulated NF-κB signaling pathway, and their possible therapeutic potential in cancer therapy is also discussed briefly.
Collapse
Affiliation(s)
- Feng Li
- Department of Pharmacology, Yong Loo Lin School of Medicine, Cancer Science Institute, National University of Singapore, Singapore, 117597, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Robles-Espinoza CD, Velasco-Herrera MDC, Hayward NK, Adams DJ. Telomere-regulating genes and the telomere interactome in familial cancers. Mol Cancer Res 2015; 13:211-22. [PMID: 25244922 PMCID: PMC4278843 DOI: 10.1158/1541-7786.mcr-14-0305] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Telomeres are repetitive sequence structures at the ends of linear chromosomes that consist of double-stranded DNA repeats followed by a short single-stranded DNA protrusion. Telomeres need to be replicated in each cell cycle and protected from DNA-processing enzymes, tasks that cells execute using specialized protein complexes such as telomerase (that includes TERT), which aids in telomere maintenance and replication, and the shelterin complex, which protects chromosome ends. These complexes are also able to interact with a variety of other proteins, referred to as the telomere interactome, to fulfill their biological functions and control signaling cascades originating from telomeres. Given their essential role in genomic maintenance and cell-cycle control, germline mutations in telomere-regulating proteins and their interacting partners have been found to underlie a variety of diseases and cancer-predisposition syndromes. These syndromes can be characterized by progressively shortening telomeres, in which carriers can present with organ failure due to stem cell senescence among other characteristics, or can also present with long or unprotected telomeres, providing an alternative route for cancer formation. This review summarizes the critical roles that telomere-regulating proteins play in cell-cycle control and cell fate and explores the current knowledge on different cancer-predisposing conditions that have been linked to germline defects in these proteins and their interacting partners.
Collapse
Affiliation(s)
| | | | - Nicholas K Hayward
- Oncogenomics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - David J Adams
- Experimental Cancer Genetics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
35
|
Odqvist L, Montes-Moreno S, Sánchez-Pacheco RE, Young KH, Martín-Sánchez E, Cereceda L, Sánchez-Verde L, Pajares R, Mollejo M, Fresno MF, Mazorra F, Ruíz-Marcellán C, Sánchez-Beato M, Piris MA. NFκB expression is a feature of both activated B-cell-like and germinal center B-cell-like subtypes of diffuse large B-cell lymphoma. Mod Pathol 2014; 27:1331-1337. [PMID: 24603590 DOI: 10.1038/modpathol.2014.34] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 12/19/2013] [Indexed: 02/07/2023]
Abstract
The activation of nuclear factor kappa B (NFκB) transcription factor family is considered to have a key role in diffuse large B-cell lymphoma (DLBCL) pathogenesis and is associated with a specific molecular subtype, the activated B-cell-like (ABC) subtype. We evaluated the expression of NFκB by immunohistochemistry in a large series of DLBCL cases. The five different NFκB family members (NFκB1, NFκB2, RELA, RELB, and REL) showed a heterogeneous expression pattern with the vast majority of cases being positive for at least one factor. Two independent series of tumor samples were classified into germinal center B-cell-like (GCB) or ABC subtypes using different approaches, immunohistochemistry, or gene expression profiling, and the expression of NFκB family members was assessed. Notably, no significant differences regarding the expression of the different NFκB members were detected between the two subtypes, suggesting that NFκB signaling is a prominent feature not only in the ABC subtype, but also in the GCB tumors. Of the five transcription factors, only REL expression had a significant clinical impact on R-CHOP-treated diffuse large B-cell lymphoma, identifying a subgroup of patients with superior clinical outcome.
Collapse
Affiliation(s)
- Lina Odqvist
- 1] Molecular Pathology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain [2] Cancer Genomics Group, Valdecilla Biomedical Research Institute (IFIMAV), Santander, Spain
| | - Santiago Montes-Moreno
- Pathology Department, Hospital Universitario Marques de Valdecilla, Universidad de Cantabria, IFIMAV, Santander, Spain
| | - Roxana E Sánchez-Pacheco
- Pathology Department, Hospital Universitario Marques de Valdecilla, Universidad de Cantabria, IFIMAV, Santander, Spain
| | - Ken H Young
- UT MD Anderson Cancer Center, Houston, TX, USA
| | - Esperanza Martín-Sánchez
- 1] Molecular Pathology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain [2] Cancer Genomics Group, Valdecilla Biomedical Research Institute (IFIMAV), Santander, Spain
| | - Laura Cereceda
- Cancer Genomics Group, Valdecilla Biomedical Research Institute (IFIMAV), Santander, Spain
| | | | | | | | | | - Francisco Mazorra
- Pathology Department, Hospital Universitario Marques de Valdecilla, Universidad de Cantabria, IFIMAV, Santander, Spain
| | | | - Margarita Sánchez-Beato
- Oncology-Hematology Area, Instituto Investigación Sanitaria Hospital Universitario Puerta de Hierro-Majadahonda, Calle Joaquín Rodrígo 2, Madrid, Spain
| | - Miguel A Piris
- 1] Cancer Genomics Group, Valdecilla Biomedical Research Institute (IFIMAV), Santander, Spain [2] Pathology Department, Hospital Universitario Marques de Valdecilla, Universidad de Cantabria, IFIMAV, Santander, Spain
| |
Collapse
|
36
|
Neo WH, Lim JF, Grumont R, Gerondakis S, Su IH. c-Rel regulates Ezh2 expression in activated lymphocytes and malignant lymphoid cells. J Biol Chem 2014; 289:31693-31707. [PMID: 25266721 DOI: 10.1074/jbc.m114.574517] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The polycomb group protein Ezh2 is a histone methyltransferase that modifies chromatin structure to alter gene expression during embryonic development, lymphocyte activation, and tumorigenesis. The mechanism by which Ezh2 expression is regulated is not well defined. In the current study, we report that c-Rel is a critical activator of Ezh2 transcription in lymphoid cells. In activated primary murine B and T cells, plus human leukemia and multiple myeloma cell lines, recruitment of c-Rel to the first intron of the Ezh2 locus promoted Ezh2 mRNA expression. This up-regulation was abolished in activated c-Rel-deficient lymphocytes and by c-Rel knockdown in Jurkat T cells. Treatment of malignant cells with the c-Rel inhibitor pentoxifylline not only reduced c-Rel nuclear translocation and Ezh2 expression, but also enhanced their sensitivity to the Ezh2-specific drug, GSK126 through increased growth inhibition and cell death. In summary, our demonstration that c-Rel regulates Ezh2 expression in lymphocytes and malignant lymphoid cells reveals a novel transcriptional network in transformed lymphoid cells expressing high levels of Ezh2 that provides a molecular justification for combinatorial drug therapy.
Collapse
Affiliation(s)
- Wen Hao Neo
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Republic of Singapore and
| | - Jun Feng Lim
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Republic of Singapore and
| | - Raelene Grumont
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia
| | - Steve Gerondakis
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia
| | - I-Hsin Su
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Republic of Singapore and.
| |
Collapse
|
37
|
Alves BN, Tsui R, Almaden J, Shokhirev MN, Davis-Turak J, Fujimoto J, Birnbaum H, Ponomarenko J, Hoffmann A. IκBε is a key regulator of B cell expansion by providing negative feedback on cRel and RelA in a stimulus-specific manner. THE JOURNAL OF IMMUNOLOGY 2014; 192:3121-32. [PMID: 24591377 DOI: 10.4049/jimmunol.1302351] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The transcription factor NF-κB is a regulator of inflammatory and adaptive immune responses, yet only IκBα was shown to limit NF-κB activation and inflammatory responses. We investigated another negative feedback regulator, IκBε, in the regulation of B cell proliferation and survival. Loss of IκBε resulted in increased B cell proliferation and survival in response to both antigenic and innate stimulation. NF-κB activity was elevated during late-phase activation, but the dimer composition was stimulus specific. In response to IgM, cRel dimers were elevated in IκBε-deficient cells, yet in response to LPS, RelA dimers also were elevated. The corresponding dimer-specific sequences were found in the promoters of hyperactivated genes. Using a mathematical model of the NF-κB-signaling system in B cells, we demonstrated that kinetic considerations of IκB kinase-signaling input and IκBε's interactions with RelA- and cRel-specific dimers could account for this stimulus specificity. cRel is known to be the key regulator of B cell expansion. We found that the RelA-specific phenotype in LPS-stimulated cells was physiologically relevant: unbiased transcriptome profiling revealed that the inflammatory cytokine IL-6 was hyperactivated in IκBε(-/-) B cells. When IL-6R was blocked, LPS-responsive IκBε(-/-) B cell proliferation was reduced to near wild-type levels. Our results provide novel evidence for a critical role for immune-response functions of IκBε in B cells; it regulates proliferative capacity via at least two mechanisms involving cRel- and RelA-containing NF-κB dimers. This study illustrates the importance of kinetic considerations in understanding the functional specificity of negative-feedback regulators.
Collapse
Affiliation(s)
- Bryce N Alves
- Signaling Systems Laboratory, Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Jones MR, Liu C, Wilson AK. Molecular dynamics studies of the protein-protein interactions in inhibitor of κB kinase-β. J Chem Inf Model 2014; 54:562-72. [PMID: 24437505 DOI: 10.1021/ci400720n] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Activation of the inhibitor of κB kinase subunit β (IKKβ) oligomer initiates a cascade that results in the translocation of transcription factors involved in mediating immune responses. Dimerization of IKKβ is required for its activation. Coarse-grained and atomistic molecular dynamics simulations were used to investigate the conformation-activity and structure-activity relationships within the oligomer assembly of IKKβ that are impacted upon activation, mutation, and binding of ATP. Intermolecular interactions, free energies, and conformational changes were compared among several conformations, including a monomer, two different dimers, and the tetramer. Modifications to the activation segment induce conformational changes that disrupt dimerization and suggest that the multimeric assembly mediates a global stability for the enzyme that influences the activity of IKKβ.
Collapse
Affiliation(s)
- Michael R Jones
- Department of Chemistry and Center for Advanced Scientific Computing and Modeling, University of North Texas , Denton, Texas 76203-5017, United States
| | | | | |
Collapse
|
39
|
Jia XF, Shen L, Fan H, Xie JW, Zeng YY, Chen X, Zhu R. Significance of NF-κB p65 expression in ulcerative colitis and colorectal adenocarcinoma. Shijie Huaren Xiaohua Zazhi 2014; 22:279-285. [DOI: 10.11569/wcjd.v22.i2.279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of nuclear factor κB (NF-κB) p65 protein in the colon mucosa of patients with ulcerative colitis (UC) or colorectal adenocarcinoma (CA) and healthy volunteers.
METHODS: Specimens were obtained from 12 healthy volunteers, 16 patients with UC and 18 patients with CA. Fresh colonic tissues were obtained by endoscopic punch biopsies. The diagnosis was confirmed pathologically. Reverse transcription-polymerase chain reaction and immunohistochemistry were used to examine the expression of NF-κB p65 at both mRNA and protein levels in the colon mucosa of different groups.
RESULTS: The expression rate of NF-κB p65 mRNA and protein was 0.10% ± 0.03% and 2.06%±0.70% in the normal colon mucosa, 0.96% ± 0.11% and 36.16% ± 6.99% in UC, and 0.42% ± 0.77% and 9.54% ± 2.77% in CA, respectively. The expression of NF-κB p65 was significantly higher in UC than in CA and normal mucosa (all P < 0.01). Although the expression of NF-κB p65 in CA was higher than that in normal colon mucosa at both mRNA and protein levels, the expression intensity was not as strong as that reported in the literature.
CONCLUSION: Increased expression of NF-κB p65 may play an important role in the pathogenesis of UC, and anti-NF-κB therapy should be examined further for use as a potential therapy. NF-κB p65 may also be involved in the pathogenesis of CA.
Collapse
|
40
|
López Y, Patil A, Nakai K. Identification of novel motif patterns to decipher the promoter architecture of co-expressed genes in Arabidopsis thaliana. BMC SYSTEMS BIOLOGY 2013; 7 Suppl 3:S10. [PMID: 24555803 PMCID: PMC3852273 DOI: 10.1186/1752-0509-7-s3-s10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background The understanding of the mechanisms of transcriptional regulation remains a challenge for molecular biologists in the post-genome era. It is hypothesized that the regulatory regions of genes expressed in the same tissue or cell type share a similar structure. Though several studies have analyzed the promoters of genes expressed in specific metazoan tissues or cells, little research has been done in plants. Hence finding specific patterns of motifs to explain the promoter architecture of co-expressed genes in plants could shed light on their transcription mechanism. Results We identified novel patterns of sets of motifs in promoters of genes co-expressed in four different plant structures (PSs) and in the entire plant in Arabidopsis thaliana. Sets of genes expressed in four PSs (flower, seed, root, shoot) and housekeeping genes expressed in the entire plant were taken from a database of co-expressed genes in A. thaliana. PS-specific motifs were predicted using three motif-discovery algorithms, 8 of which are novel, to the best of our knowledge. A support vector machine was trained using the average upstream distance of the identified motifs from the translation start site on both strands of binding sites. The correctly classified promoters per PS were used to construct specific patterns of sets of motifs to describe the promoter architecture of those co-expressed genes. The discovered PS-specific patterns were tested in the entire A. thaliana genome, correctly identifying 77.8%, 81.2%, 70.8% and 53.7% genes expressed in petal differentiation, synergid cells, root hair and trichome, as well as 88.4% housekeeping genes. Conclusions We present five patterns of sets of motifs which describe the promoter architecture of co-expressed genes in five PSs with the ability to predict them from the entire A. thaliana genome. Based on these findings, we conclude that the positioning and orientation of transcription factor binding sites at specific distances from the translation start site is a reliable measure to differentiate promoters of genes expressed in different A. thaliana structures from background genomic promoters. Our method can be used to predict novel motifs and decipher a similar promoter architecture for genes co-expressed in A. thaliana under different conditions.
Collapse
|
41
|
Burkitt MD, Williams JM, Duckworth CA, O'Hara A, Hanedi A, Varro A, Caamaño JH, Pritchard DM. Signaling mediated by the NF-κB sub-units NF-κB1, NF-κB2 and c-Rel differentially regulate Helicobacter felis-induced gastric carcinogenesis in C57BL/6 mice. Oncogene 2013; 32:5563-73. [PMID: 23975431 PMCID: PMC3898319 DOI: 10.1038/onc.2013.334] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 06/03/2013] [Accepted: 06/17/2013] [Indexed: 12/15/2022]
Abstract
The classical nuclear factor-kappaB (NF-κB) signaling pathway has been shown to be important in a number of models of inflammation-associated cancer. In a mouse model of Helicobacter-induced gastric cancer, impairment of classical NF-κB signaling in the gastric epithelium led to the development of increased preneoplastic pathology, however the role of specific NF-κB proteins in Helicobacter-associated gastric cancer development remains poorly understood. To investigate this C57BL/6, Nfkb1−/−, Nfkb2−/− and c-Rel−/− mice were infected with Helicobacter felis for 6 weeks or 12 months. Bacterial colonization, gastric atrophy and preneoplastic changes were assessed histologically and cytokine expression was assessed by qPCR. Nfkb1−/− mice developed spontaneous gastric atrophy when maintained for 12 months in conventional animal house conditions. They also developed more pronounced gastric atrophy after short-term H. felis colonization with a similar extent of preneoplasia to wild-type (WT) mice after 12 months. c-Rel−/− mice developed a similar degree of gastric atrophy to WT mice; 3 of 6 of these animals also developed lymphoproliferative lesions after 12 months of infection. Nfkb2−/− mice developed minimal gastric epithelial pathology even 12 months after H. felis infection. These findings demonstrate that NF-κB1- and NF-κB2-mediated signaling pathways differentially regulate the epithelial consequences of H. felis infection in the stomach, while c-Rel-mediated signaling also appears to modulate the risk of lymphomagenesis in gastric mucosa-associated lymphoid tissue.
Collapse
Affiliation(s)
- M D Burkitt
- Department of Gastroenterology, The Henry Wellcome Laboratories, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - J M Williams
- Department of Gastroenterology, The Henry Wellcome Laboratories, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - C A Duckworth
- Department of Gastroenterology, The Henry Wellcome Laboratories, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - A O'Hara
- Department of Gastroenterology, The Henry Wellcome Laboratories, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - A Hanedi
- Department of Gastroenterology, The Henry Wellcome Laboratories, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - A Varro
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - J H Caamaño
- IBR-School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - D M Pritchard
- Department of Gastroenterology, The Henry Wellcome Laboratories, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
42
|
CluGene: A Bioinformatics Framework for the Identification of Co-Localized, Co-Expressed and Co-Regulated Genes Aimed at the Investigation of Transcriptional Regulatory Networks from High-Throughput Expression Data. PLoS One 2013; 8:e66196. [PMID: 23823315 PMCID: PMC3688840 DOI: 10.1371/journal.pone.0066196] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 05/05/2013] [Indexed: 01/03/2023] Open
Abstract
The full understanding of the mechanisms underlying transcriptional regulatory networks requires unravelling of complex causal relationships. Genome high-throughput technologies produce a huge amount of information pertaining gene expression and regulation; however, the complexity of the available data is often overwhelming and tools are needed to extract and organize the relevant information. This work starts from the assumption that the observation of co-occurrent events (in particular co-localization, co-expression and co-regulation) may provide a powerful starting point to begin unravelling transcriptional regulatory networks. Co-expressed genes often imply shared functional pathways; co-expressed and functionally related genes are often co-localized, too; moreover, co-expressed and co-localized genes are also potential targets for co-regulation; finally, co-regulation seems more frequent for genes mapped to proximal chromosome regions. Despite the recognized importance of analysing co-occurrent events, no bioinformatics solution allowing the simultaneous analysis of co-expression, co-localization and co-regulation is currently available. Our work resulted in developing and valuating CluGene, a software providing tools to analyze multiple types of co-occurrences within a single interactive environment allowing the interactive investigation of combined co-expression, co-localization and co-regulation of genes. The use of CluGene will enhance the power of testing hypothesis and experimental approaches aimed at unravelling transcriptional regulatory networks. The software is freely available at http://bioinfolab.unipg.it/.
Collapse
|
43
|
Yu Q, Zhou C, Wang J, Chen L, Zheng S, Zhang J. A functional insertion/deletion polymorphism in the promoter of PDCD6IP is associated with the susceptibility of hepatocellular carcinoma in a Chinese population. DNA Cell Biol 2013; 32:451-7. [PMID: 23777424 DOI: 10.1089/dna.2013.2061] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide. Apart from environmental factors such as hepatitis B virus (HBV) or hepatitis C virus, alcohol abuse, and exposure to dietary aflatoxin, genetic factors are also involved in the pathogenesis of HCC. By analyzing 390 HCC cases and 431 healthy controls in a Chinese population, we used a candidate gene approach to evaluate the association between a 15-bp insertion/deletion (indel) polymorphism (rs28381975) in the promoter region of the programmed cell death 6 interacting protein (PDCD6IP) gene and HCC susceptibility. Logistic regression analysis demonstrated that subjects carrying ins/del or ins/ins genotypes had significantly increased risk for HCC than individuals carrying del/del genotypes (adjusted odds ratio=1.39, 95% confidence interval=1.01-1.91, p=0.033]. Carrying the 15-bp insertion allele was associated with a 1.26-fold risk for HCC (95% CI=1.04-1.54, p=0.018). Moreover, significant differences were observed within HCC patients concerning genotypic frequencies of rs28381975 after stratifying by tumor stages and HBV infection. Computational modeling suggests that rs28381975 could disrupt the binding patterns of c-rel, a key subunit of nuclear factor-kappaB transcription factor. Further luciferase-based transient transfection assays revealed that rs28381975 can affect the promoter activity of PDCD6IP, indicating its possible functional significance. Taken together, our data suggest that common genetic variations in PDCD6IP may influence HCC risk, possibly through promoter activity-mediated regulation. Replication of our studies in other populations and further functional analysis will strengthen our understanding of this association.
Collapse
Affiliation(s)
- Qiang Yu
- Department of Gastroenterology, Suzhou Municipal Hospital, Affiliated to Nanjing Medical University, Suzhou, Jiangsu, China
| | | | | | | | | | | |
Collapse
|
44
|
Fan Y, Mao R, Yang J. NF-κB and STAT3 signaling pathways collaboratively link inflammation to cancer. Protein Cell 2013; 4:176-85. [PMID: 23483479 DOI: 10.1007/s13238-013-2084-3] [Citation(s) in RCA: 535] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Accepted: 09/03/2012] [Indexed: 12/18/2022] Open
Abstract
Although links between cancer and inflammation were firstly proposed in the nineteenth century, the molecular mechanism has not yet been clearly understood. Epidemiological studies have identified chronic infections and inflammation as major risk factors for various types of cancer. NF-κB transcription factors and the signaling pathways are central coordinators in innate and adaptive immune responses. STAT3 regulates the expression of a variety of genes in response to cellular stimuli, and thus plays a key role in cell growth and apoptosis. Recently, roles of NF-κB and STAT3 in colon, gastric and liver cancers have been extensively investigated. The activation and interaction between STAT3 and NF-κB play vital roles in control of the communication between cancer cells and inflammatory cells. NF-κB and STAT3 are two major factors controlling the ability of pre-neoplastic and malignant cells to resist apoptosis-based tumor-surveillance and regulating tumor angiogenesis and invasiveness. Understanding the molecular mechanisms of NF-κB and STAT3 cooperation in cancer will offer opportunities for the design of new chemo-preventive and chemotherapeutic approaches.
Collapse
Affiliation(s)
- Yihui Fan
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | |
Collapse
|
45
|
Chernet BT, Levin M. Transmembrane voltage potential is an essential cellular parameter for the detection and control of tumor development in a Xenopus model. Dis Model Mech 2013; 6:595-607. [PMID: 23471912 PMCID: PMC3634644 DOI: 10.1242/dmm.010835] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Understanding mechanisms that orchestrate cell behavior into appropriately patterned tissues and organs within the organism is an essential element of preventing, detecting and treating cancer. Bioelectric signals (resting transmembrane voltage potential gradients in all cells) underlie an important and broadly conserved set of control mechanisms that regulate pattern formation. We tested the role of transmembrane potential in tumorigenesis mediated by canonical oncogenes in Xenopus laevis. Depolarized membrane potential (Vmem) was a characteristic of induced tumor-like structures (ITLSs) generated by overexpression of Gli1, KrasG12D, Xrel3 or p53Trp248. This bioelectric signature was also present in precursor ITLS sites. Vmem is a bioelectric marker that reveals ITLSs before they become histologically and morphologically apparent. Moreover, voltage was functionally important: overexpression of hyperpolarizing ion transporters caused a return to normal Vmem and significantly reduced ITLS formation in vivo. To characterize the molecular mechanism by which Vmem change regulates ITLS phenotypes, we performed a suppression screen. Vmem hyperpolarization was transduced into downstream events via Vmem-regulated activity of SLC5A8, a sodium-butyrate exchanger previously implicated in human cancer. These data indicate that butyrate, a histone deacetylase (HDAC) inhibitor, might be responsible for transcriptional events that mediate suppression of ITLSs by hyperpolarization. Vmem is a convenient cellular parameter by which tumors induced by human oncogenes can be detected in vivo and represents a new diagnostic modality. Moreover, control of resting membrane potential is functionally involved in the process by which oncogene-bearing cells depart from normal morphogenesis programs to form tumors. Modulation of Vmem levels is a novel and promising strategy for tumor normalization.
Collapse
Affiliation(s)
- Brook T Chernet
- Center for Regenerative and Developmental Biology and Department of Biology, Tufts University, 200 Boston Avenue, Medford, MA 02155, USA
| | | |
Collapse
|
46
|
Gilmore TD, Gerondakis S. The c-Rel Transcription Factor in Development and Disease. Genes Cancer 2012; 2:695-711. [PMID: 22207895 DOI: 10.1177/1947601911421925] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 08/08/2011] [Indexed: 12/21/2022] Open
Abstract
c-Rel is a member of the nuclear factor κB (NF-κB) transcription factor family. Unlike other NF-κB proteins that are expressed in a variety of cell types, high levels of c-Rel expression are found primarily in B and T cells, with many c-Rel target genes involved in lymphoid cell growth and survival. In addition to c-Rel playing a major role in mammalian B and T cell function, the human c-rel gene (REL) is a susceptibility locus for certain autoimmune diseases such as arthritis, psoriasis, and celiac disease. The REL locus is also frequently altered (amplified, mutated, rearranged), and expression of REL is increased in a variety of B and T cell malignancies and, to a lesser extent, in other cancer types. Thus, agents that modulate REL activity may have therapeutic benefits for certain human cancers and chronic inflammatory diseases.
Collapse
|
47
|
Abstract
Cigarette smoke (CS), a major risk factor for developing lung cancer, is known to activate transcriptional activator nuclear factor kappa B (NF-κB). However, the underlying mechanism of this activation remains unclear because of conflicting reports. As NF-κB has a pivotal role in the generation and maintenance of malignancies, efforts were targeted towards understanding its activation mechanism using both ex vivo and in vivo studies. The results show that CS-induced NF-κB activation mechanism is different from that of other pro-inflammatory signals such as lipopolysaccharide (LPS). The NF-κB dimer that translocates to the nucleus upon stimulation with CS is predominantly composed of c-Rel/p50 and this translocation involves degradation of I-κBɛ and not I-κBα. This degradation of I-κBɛ depends on IKKβ activity, which preferentially targets I-κBɛ. Consistently, CS-activated form of IKKβ was found to be different from that involved in LPS activation as neither Ser177 nor Ser181 of IKKβ is crucial for CS-induced NF-κB activation. Thus, unlike other pro-inflammatory stimulations where p65 and I-κBα have a central role, the predominantly active signaling cascade in CS-induced NF-κB activation in the lung epithelial cells comprises of IKKβ–I-κBɛ–c-Rel/p50. Thus, this study uncovers a new axis of NF-κB activation wherein I-κBɛ and c-Rel have the central role.
Collapse
|
48
|
Yeo AT, Porco JA, Gilmore TD. Bcl-XL, but not Bcl-2, can protect human B-lymphoma cell lines from parthenolide-induced apoptosis. Cancer Lett 2011; 318:53-60. [PMID: 22155272 DOI: 10.1016/j.canlet.2011.11.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2011] [Revised: 11/29/2011] [Accepted: 11/30/2011] [Indexed: 12/19/2022]
Abstract
In this report, we investigated the effects of the natural product parthenolide on human B-lymphoma cell lines. We show that parthenolide inhibited NF-κB transcription factor c-Rel (REL). In addition, the sensitivity of several human B-lymphoma cell lines to parthenolide-induced apoptosis inversely correlated with their levels of anti-apoptosis protein Bcl-X(L). Furthermore, ectopic expression of Bcl-X(L) (but not Bcl-2) in two B-lymphoma cell lines decreased their sensitivity to parthenolide-induced apoptosis. Finally, over-expression of a transforming mutant of REL, which increased expression of endogenous Bcl-X(L), decreased the sensitivity of BJAB B-lymphoma cells to parthenolide-induced apoptosis. These results demonstrate that the NF-κB target gene products Bcl-X(L) and Bcl-2 can play different roles in protecting B-lymphoma cells from chemical-induced apoptosis.
Collapse
Affiliation(s)
- Alan T Yeo
- Department of Biology, Boston University, Boston, MA 02215, USA
| | | | | |
Collapse
|
49
|
Lu H, Yang X, Duggal P, Allen CT, Yan B, Cohen J, Nottingham L, Romano RA, Sinha S, King KE, Weinberg WC, Chen Z, Van Waes C. TNF-α promotes c-REL/ΔNp63α interaction and TAp73 dissociation from key genes that mediate growth arrest and apoptosis in head and neck cancer. Cancer Res 2011; 71:6867-77. [PMID: 21933882 PMCID: PMC3206154 DOI: 10.1158/0008-5472.can-11-2460] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Inflammation-induced activation of proto-oncogenic NF-κB/REL and dysfunction of tumor suppressor TP53/p63/p73 family transcription factors are key events in cancer progression. How inflammatory signaling coordinates dysregulation of these two transcription factor families during oncogenesis remains incompletely understood. Here, we observed that oncoprotein c-REL and tumor suppressor TAp73 are coexpressed and complex with ΔNp63α in the nucleus of a subset of head and neck squamous cell carcinoma (HNSCC) cell lines with mutant (mt)TP53. TNF-α, a proinflammatory cytokine, promoted c-REL nuclear translocation, c-REL/ΔNp63α interaction, and dissociation of TAp73 from ΔNp63α and the nucleus to the cytoplasm, whereas c-REL siRNA knockdown attenuated this effect. Overexpression of c-REL or a c-REL κB-site DNA-binding mutant enhanced protein interaction with ΔNp63α and TAp73 dissociation, implicating c-REL/ΔNp63α-specific interactions in these effects. We discovered that TNF-α or genetic alteration of c-REL expression inversely modulates ΔNp63α/TAp73 interactions on distinct p63 DNA-binding sites, including those for key growth arrest and apoptotic genes p21WAF1, NOXA, and PUMA. Functionally, c-REL repressed these genes and the antiproliferative effects of TNF-α or TAp73. Conversely, c-REL siRNA depletion enhanced TAp73 promoter interaction and expression of genes mediating growth arrest and apoptosis. Similar to TNF-α-treated HNSCC lines, human HNSCC tumors and hyperplastic squamous epithelia of transgenic mice overexpressing ΔNp63α that exhibit inflammation also show increased nuclear c-REL/ΔNp63α and cytoplasmic TAp73 localization. These findings unveil a novel and reversible dynamic mechanism whereby proinflammatory cytokine TNF-α-induced c-REL/ΔNp63α interactions inactivate tumor suppressor TAp73 function, promoting TNF-α resistance and cell survival in cancers with mtTP53.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Apoptosis/drug effects
- Apoptosis/genetics
- Apoptosis Regulatory Proteins/biosynthesis
- Apoptosis Regulatory Proteins/genetics
- Binding Sites
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Cell Division/drug effects
- Cell Division/genetics
- Cell Line, Tumor/drug effects
- Cell Line, Tumor/metabolism
- Cell Nucleus/metabolism
- Cyclin-Dependent Kinase Inhibitor p21/biosynthesis
- Cyclin-Dependent Kinase Inhibitor p21/genetics
- DNA, Neoplasm/metabolism
- DNA-Binding Proteins/metabolism
- Drug Resistance/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Genes, p53
- Genes, rel
- Head and Neck Neoplasms/genetics
- Head and Neck Neoplasms/pathology
- Humans
- Multiprotein Complexes
- Neoplasm Proteins/metabolism
- Nuclear Proteins/metabolism
- Promoter Regions, Genetic/drug effects
- Protein Binding/drug effects
- Proto-Oncogene Proteins/biosynthesis
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins c-bcl-2/biosynthesis
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-rel
- RNA, Small Interfering/pharmacology
- Transcription Factors/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
- Tumor Suppressor Proteins/metabolism
Collapse
Affiliation(s)
- Hai Lu
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Valentín-Acevedo A, Sinquett FL, Covey LR. c-Rel deficiency increases caspase-4 expression and leads to ER stress and necrosis in EBV-transformed cells. PLoS One 2011; 6:e25467. [PMID: 21984918 PMCID: PMC3184984 DOI: 10.1371/journal.pone.0025467] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 09/05/2011] [Indexed: 01/03/2023] Open
Abstract
LMP1-mediated activation of nuclear factor of kappaB (NF-κB) is critical for the ligand independent proliferation and cell survival of in vitro EBV-transformed lymphoblastoid cell lines (LCLs). Previous experiments revealed that a majority of LMP1-dependent responses are regulated by NF-κB. However, the extent that individual NF-κB family members are required for these responses, in particular, c-Rel, whose expression is restricted to mature hematopoietic cells, remains unclear. Here we report that low c-Rel expression in LCLs derived from a patient with hyper-IgM syndrome (Pt1), resulted in defects in proliferation and cell survival. In contrast to studies that associated loss of NF-κB with increased apoptosis, Pt1 LCLs failed to initiate apoptosis and alternatively underwent autophagy and necrotic cell death. Whereas the proliferation defect appeared linked to a c-Rel-associated decrease in c-myc expression, identified pro-survival and pro-apoptotic targets were expressed at or near control levels consistent with the absence of apoptosis. Ultrastructural examination of Pt1 LCLs revealed a high level of cellular and ER stress that was further supported by gene expression profiling showing the upregulation of several genes involved in stress and inflammation. Apoptosis-independent cell death was accompanied by increased expression of the inflammatory marker, caspase-4. Using gene overexpression and siRNA knockdown we demonstrated that levels of c-Rel directly modulated expression of caspase-4 as well as other ER stress genes. Overall, these findings reveal the importance of c-Rel in maintaining LCL viability and that decreased expression results in ER stress and a default response leading to necrotic cell death.
Collapse
Affiliation(s)
- Aníbal Valentín-Acevedo
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, United States of America
| | - Frank L. Sinquett
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, United States of America
| | - Lori R. Covey
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, United States of America
- * E-mail:
| |
Collapse
|