1
|
Xie H, Cao S, Chen Y, Wang Z, Chen X, Cui Z. The role of SYK phosphorylation in LPS-induced immunoglobulin responses of B cells in large yellow croaker (Larimichthys crocea). FISH & SHELLFISH IMMUNOLOGY 2025; 161:110283. [PMID: 40107331 DOI: 10.1016/j.fsi.2025.110283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
Spleen tyrosine kinase (SYK), a non-receptor protein tyrosine kinase, is a key component of B cell receptor signaling and can regulate multiple physiological functions of B cells in mammals. In this study, a SYK gene was cloned and characterized from large yellow croaker (Larimichthys crocea) (LcSYK), whose open reading frame consists of 1851 base pairs and encodes 616 amino acid residues. The predicted LcSYK protein contains two N-terminal tandem Src homology 2 domains and a C-terminal tyrosine kinase catalytic domain, and shares a high amino acid sequence identity with SYK sequences in other vertebrate species. LcSYK was mainly expressed in immune tissues, such as head kidney, trunk kidney, spleen, and gill. The mRNA expression of LcSYK in primary head kidney leukocytes was not changed at 12, 24, and 48 h after lipopolysaccharide (LPS) stimulation. LPS stimulation upregulated the mRNA expression and protein production of IgM in IgM+ B cells, accompanied by an increase in the phosphorylation level, but not the total protein level, of LcSYK. Moreover, when we used PRT062607 HCl to inhibit the phosphorylation of LcSYK, both mRNA expression and protein production of IgM in IgM+ B cells were significantly suppressed. These results suggest that SYK phosphorylation may play a role in LPS-induced IgM production by IgM+ B cells, improving our understanding of the role of SYK in immunoglobulin responses of B cells in fish.
Collapse
Affiliation(s)
- Hongjun Xie
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Shuangshuang Cao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Yueming Chen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Zhiqiang Wang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Xinhua Chen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China.
| | - Zhengwei Cui
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| |
Collapse
|
2
|
Watanabe T, Hayashi M, Arai M, Matsushita S, Handa H, Kawano M. Interferon-γ production in response to the cytotoxic T lymphocyte epitope within an antigen incorporated in simian virus 40 virus-like particles. Heliyon 2025; 11:e41729. [PMID: 39897799 PMCID: PMC11786666 DOI: 10.1016/j.heliyon.2025.e41729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 12/05/2024] [Accepted: 01/04/2025] [Indexed: 02/04/2025] Open
Abstract
In a previous study, we showed that administration of antigen-incorporated virus-like particles (VLPs) derived from simian virus 40 (SV40) induces production of cytotoxic T lymphocytes (CTL), as well as antibodies against the incorporated antigen(s), without the need for an adjuvant; however, it remains unclear how immune cells recognize and respond to the SV40 capsid because SV40 VLPs did not upregulate expression of maturation markers on dendritic cells. In this study, administration of chicken ovalbumin (OVA) incorporated within SV40 VLPs induced interferon (IFN)-γ production in response to the OVA CTL epitope. IFN-γ production in response to the OVA CTL epitope was not inhibited in B cell-depleted mice, but it was inhibited in cluster of differentiation (CD)4+ T cell-depleted mice. Administration of SV40 VLPs upregulated expression of CD63/CD68/CD83/CD86/CD196, and induced secretion of C-C chemokine ligand (CCL)3 and CCL4, by B cells, as well as secretion of CCL4 by T cells and tumor necrosis factor-α by bone marrow-derived dendritic cells.
Collapse
Affiliation(s)
- Takeharu Watanabe
- Vaccine Sohyaku Group Modality Laboratories Sohyaku. Innovation Research Division Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida, Aoba-ku, Yokohama, 227-0033, Japan
| | - Masayuki Hayashi
- Vaccine Sohyaku Group Modality Laboratories Sohyaku. Innovation Research Division Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida, Aoba-ku, Yokohama, 227-0033, Japan
| | - Masaaki Arai
- Vaccine Sohyaku Group Modality Laboratories Sohyaku. Innovation Research Division Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida, Aoba-ku, Yokohama, 227-0033, Japan
| | - Sho Matsushita
- Department of Allergy and Immunology, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama, Saitama, 350-0495, Japan
- Allergy Center, Saitama Medical University, 38 Morohongo, Moroyama, Saitama, 350-0495, Japan
| | - Hiroshi Handa
- Department of Molecular Pharmacology, Tokyo Medical University, Shinjuku, Tokyo, 160-8402, Japan
- Center for Future Medical Research, Institute of Medical Science, Tokyo Medical University, Shinjuku, Tokyo, 160-8402, Japan
| | - Masaaki Kawano
- Department of Allergy and Immunology, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama, Saitama, 350-0495, Japan
- Allergy Center, Saitama Medical University, 38 Morohongo, Moroyama, Saitama, 350-0495, Japan
- Division of Analytical Science, Biomedical Research Center, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama, Saitama, 350-0495, Japan
| |
Collapse
|
3
|
Ono C, Kochi Y, Baba Y, Tanaka S. Humoral responses are enhanced by facilitating B cell viability by Fcrl5 overexpression in B cells. Int Immunol 2024; 36:529-540. [PMID: 38738271 DOI: 10.1093/intimm/dxae028] [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: 02/28/2024] [Accepted: 05/11/2024] [Indexed: 05/14/2024] Open
Abstract
B cell initial activity is regulated through a balance of activation and suppression mediated by regulatory molecules expressed in B cells; however, the molecular mechanisms underlying this process remain incompletely understood. In this study, we investigated the function of the Fc receptor-like (Fcrl) family molecule Fcrl5, which is constitutively expressed in naive B cells, in humoral immune responses. Our study demonstrated that B cell-specific overexpression of Fcrl5 enhanced antibody (Ab) production in both T cell-independent type 1 (TI1) and T cell-dependent (TD) responses. Additionally, it promoted effector B cell formation under competitive conditions in TD responses. Mechanistically, in vitro ligation of Fcrl5 by agonistic Abs reduced cell death and enhanced proliferation in lipopolysaccharide-stimulated B cells. In the presence of anti-CD40 Abs and IL-5, the Fcrl5 ligation not only suppressed cell death but also enhanced differentiation into plasma cells. These findings reveal a novel role of Fcrl5 in promoting humoral immune responses by enhancing B cell viability and plasma cell differentiation.
Collapse
Affiliation(s)
- Chisato Ono
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yuta Kochi
- Department of Genomic Function and Diversity, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshihiro Baba
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Shinya Tanaka
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
4
|
Yang S, Wahab S, Almoyad MAA, Chen Y, Kalam N, Khalid M. Discovery of promising B lymphocyte kinase inhibitors using structure-guided virtual screening. J Biomol Struct Dyn 2024; 42:7054-7064. [PMID: 37688373 DOI: 10.1080/07391102.2023.2256397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/12/2023] [Indexed: 09/10/2023]
Abstract
Tyrosine-protein kinase BLK, also known as B-cell lymphocyte kinase (BLK), is a non-receptor tyrosine kinase that is primarily expressed in B-cells. BLK plays a key role in B-cell signaling, particularly in B-cell development and maturation. The increased expression of BLK has been linked to various complex diseases, including autoimmune disorders, and specific malignancies of B cells, such as lymphomas and leukemias. Due to its significant involvement in B-cell signaling, BLK has emerged as a promising target for drug development, offering the potential for developing novel therapeutics to combat these diseases. Small molecule inhibitors of BLK hold great potential for therapeutic intervention; however, discovering potent and selective inhibitors remains challenging. Within this context, natural compounds hold significant potential as a valuable resource for discovering novel inhibitors of BLK. In the current study, a structure-based virtual screening of the IMPPAT 2 library was employed to identify promising candidates with potential as inhibitors of BLK. The control molecule for this study was the known BLK inhibitor, Dasatinib. After a multi-step filtering process, two molecules (Withanolide I and Mexogenin) demonstrated potential against BLK based on their superior binding affinity, ligand efficiency, and specific interaction. Interaction analysis of these compounds revealed several significant interactions with the active site residues of BLK. Both proposed molecules remained bound to the binding pocket of BLK, as indicated by the molecular dynamics (MD) simulation study. Taken together, these findings provide valuable insights for guiding future research endeavors and translational efforts in developing therapeutics for different complex diseases, such as autoimmune disorders, lymphomas, and leukemias.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Song Yang
- Department of Wine, Food and Molecular Biosciences, Faculty of Agriculture & Life Sciences, Lincoln University, Christchurch, New Zealand
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Ali Abdullah Almoyad
- Department of Basic Medical Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Yanxin Chen
- Department of Anaesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Nida Kalam
- School of Pharmaceutical Education and Research (SPER), New Delhi, India
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| |
Collapse
|
5
|
Lorenzo EC, Torrance BL, Haynes L. Impact of senolytic treatment on immunity, aging, and disease. FRONTIERS IN AGING 2023; 4:1161799. [PMID: 37886012 PMCID: PMC10598643 DOI: 10.3389/fragi.2023.1161799] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 07/31/2023] [Indexed: 10/28/2023]
Abstract
Cellular senescence has been implicated in the pathophysiology of many age-related diseases. However, it also plays an important protective role in the context of tumor suppression and wound healing. Reducing senescence burden through treatment with senolytic drugs or the use of genetically targeted models of senescent cell elimination in animals has shown positive results in the context of mitigating disease and age-associated inflammation. Despite positive, albeit heterogenous, outcomes in clinical trials, very little is known about the short-term and long-term immunological consequences of using senolytics as a treatment for age-related conditions. Further, many studies examining cellular senescence and senolytic treatment have been demonstrated in non-infectious disease models. Several recent reports suggest that senescent cell elimination may have benefits in COVID-19 and influenza resolution and disease prognosis. In this review, we discuss the current clinical trials and pre-clinical studies that are exploring the impact of senolytics on cellular immunity. We propose that while eliminating senescent cells may have an acute beneficial impact on primary immune responses, immunological memory may be negatively impacted. Closer investigation of senolytics on immune function and memory generation would provide insight as to whether senolytics could be used to enhance the aging immune system and have potential to be used as therapeutics or prophylactics in populations that are severely and disproportionately affected by infections such as the elderly and immunocompromised.
Collapse
Affiliation(s)
- Erica C. Lorenzo
- UConn Health Center on Aging, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Blake L. Torrance
- UConn Health Center on Aging, University of Connecticut School of Medicine, Farmington, CT, United States
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Laura Haynes
- UConn Health Center on Aging, University of Connecticut School of Medicine, Farmington, CT, United States
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| |
Collapse
|
6
|
Mamidi MK, Huang J, Honjo K, Li R, Tabengwa EM, Neeli I, Randall NL, Ponnuchetty MV, Radic M, Leu CM, Davis RS. FCRL1 immunoregulation in B cell development and malignancy. Front Immunol 2023; 14:1251127. [PMID: 37822931 PMCID: PMC10562807 DOI: 10.3389/fimmu.2023.1251127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/01/2023] [Indexed: 10/13/2023] Open
Abstract
Immunotherapeutic targeting of surface regulatory proteins and pharmacologic inhibition of critical signaling pathways has dramatically shifted our approach to the care of individuals with B cell malignancies. This evolution in therapy reflects the central role of the B cell receptor (BCR) signaling complex and its co-receptors in the pathogenesis of B lineage leukemias and lymphomas. Members of the Fc receptor-like gene family (FCRL1-6) encode cell surface receptors with complex tyrosine-based regulation that are preferentially expressed by B cells. Among them, FCRL1 expression peaks on naïve and memory B cells and is unique in terms of its intracellular co-activation potential. Recent studies in human and mouse models indicate that FCRL1 contributes to the formation of the BCR signalosome, modulates B cell signaling, and promotes humoral responses. Progress in understanding its regulatory properties, along with evidence for its over-expression by mature B cell leukemias and lymphomas, collectively imply important yet unmet opportunities for FCRL1 in B cell development and transformation. Here we review recent advances in FCRL1 biology and highlight its emerging significance as a promising biomarker and therapeutic target in B cell lymphoproliferative disorders.
Collapse
Affiliation(s)
- Murali K. Mamidi
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jifeng Huang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kazuhito Honjo
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ran Li
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Edlue M. Tabengwa
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Indira Neeli
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Nar’asha L. Randall
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Manasa V. Ponnuchetty
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Marko Radic
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Chuen-Miin Leu
- Institute of Microbiology and Immunology, National Yang Ming ChiaoTung University, Taipei, Taiwan
| | - Randall S. Davis
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Departments of Microbiology, and Biochemistry & Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
7
|
Tsubata T. Siglec cis-ligands and their roles in the immune system. Glycobiology 2023; 33:532-544. [PMID: 37154567 DOI: 10.1093/glycob/cwad038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/14/2023] [Indexed: 05/10/2023] Open
Abstract
Sialic acid-binding immunoglobulin-like lectins are a family of membrane molecules primarily expressed in immune cells. Most of them are inhibitory receptors containing immunoreceptor tyrosine-based inhibition motifs in the cytoplasmic tail. On the cell surface, sialic acid-binding immunoglobulin-like lectins are mostly bound by sialylated glycans on membrane molecules expressed in the same cell (cis-ligands). Although ligands of sialic acid-binding immunoglobulin-like lectins are not efficiently identified by conventional methods such as immunoprecipitation, in situ labeling including proximity labeling is useful in identifying both cis-ligands and the sialylated ligands expressed by other cells (trans-ligands) of sialic acid-binding immunoglobulin-like lectins. Interaction of the inhibitory sialic acid-binding immunoglobulin-like lectins with cis-ligands including both those with and without signaling function modulates the inhibitory activity of sialic acid-binding immunoglobulin-like lectins by multiple different ways. This interaction also modulates signaling function of the cis-ligands. So far, little is known about the role of the interaction between sialic acid-binding immunoglobulin-like lectins and the cis-ligands. Nonetheless, recent studies showed that the inhibitory activity of CD22 (also known as Siglec-2) is regulated by endogenous ligands, most likely cis-ligands, differentially in resting B cells and those in which B-cell antigen receptor is ligated. This differential regulation plays a role in quality control of signaling-competent B cells and also partial restoration of B-cell antigen receptor signaling in immunodeficient B cells.
Collapse
Affiliation(s)
- Takeshi Tsubata
- Department of Pathology, Nihon University School of Dentistry, Tokyo 101-8310, Japan
| |
Collapse
|
8
|
Guo C, Mattingly RR, Stemmer PM, Rosenspire AJ. At low levels, inorganic mercury interference with antigen signaling is associated with modifications to a panel of novel phosphoserine sites in B cell receptor pathway proteins. Toxicol In Vitro 2023; 89:105564. [PMID: 36736710 DOI: 10.1016/j.tiv.2023.105564] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/23/2022] [Accepted: 01/29/2023] [Indexed: 02/04/2023]
Abstract
Epidemiological studies indicate that human and animal exposure to environmental mercury (Hg) disrupts normal immune system function, but the molecular mechanism responsible for this is still unresolved. We have previously utilized phospho-proteomic mass spectrometry to demonstrate that in the absence of B Cell Receptor (BCR) stimulation, exposure of B cells to Hg induces significant changes to a great many elements of the BCR signaling pathway in a concentration dependent manner. In this report, we have extended those initial findings by utilizing mass spectrometry to evaluate in detail the effect of low-level Hg exposure on BCR induced phospho-proteomic changes. Specifically, murine WEHI-231 B lymphoma cells were exposed to environmentally relevant levels of Hg with or without concomitant BCR stimulation. The cellular phospho-proteomes were then profiled by LC-MS/MS. We found that for low-level exposures, Hg interference with signal transduction across the BCR pathway was predominantly associated with modification of phosphorylation of 12 phosphosites located on seven different proteins. Nine sites were serine, two sites tyrosine and one site threonine. Most of these sites are novel, in the sense that only the two tyrosine and one of the serine sites have previously been reported to be associated with BCR signaling.
Collapse
Affiliation(s)
- Chunna Guo
- Department of Biochemistry, Microbiology and Immunology, Wayne State University, Detroit, MI, USA
| | - Raymond R Mattingly
- Department of Pharmacology & Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Paul M Stemmer
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA.
| | - Allen J Rosenspire
- Department of Biochemistry, Microbiology and Immunology, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
9
|
Mäkinen S, Datta N, Rangarajan S, Nguyen YH, Olkkonen VM, Latva-Rasku A, Nuutila P, Laakso M, Koistinen HA. Finnish-specific AKT2 gene variant leads to impaired insulin signalling in myotubes. J Mol Endocrinol 2023; 70:JME-21-0285. [PMID: 36409629 PMCID: PMC9874976 DOI: 10.1530/jme-21-0285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/21/2022] [Indexed: 11/22/2022]
Abstract
Finnish-specific gene variant p.P50T/AKT2 (minor allele frequency (MAF) = 1.1%) is associated with insulin resistance and increased predisposition to type 2 diabetes. Here, we have investigated in vitro the impact of the gene variant on glucose metabolism and intracellular signalling in human primary skeletal muscle cells, which were established from 14 male p.P50T/AKT2 variant carriers and 14 controls. Insulin-stimulated glucose uptake and glucose incorporation into glycogen were detected with 2-[1,2-3H]-deoxy-D-glucose and D-[14C]-glucose, respectively, and the rate of glycolysis was measured with a Seahorse XFe96 analyzer. Insulin signalling was investigated with Western blotting. The binding of variant and control AKT2-PH domains to phosphatidylinositol (3,4,5)-trisphosphate (PI(3,4,5)P3) was assayed using PIP StripsTM Membranes. Protein tyrosine kinase and serine-threonine kinase assays were performed using the PamGene® kinome profiling system. Insulin-stimulated glucose uptake and glycogen synthesis in myotubes in vitro were not significantly affected by the genotype. However, the insulin-stimulated glycolytic rate was impaired in variant myotubes. Western blot analysis showed that insulin-stimulated phosphorylation of AKT-Thr308, AS160-Thr642 and GSK3β-Ser9 was reduced in variant myotubes compared to controls. The binding of variant AKT2-PH domain to PI(3,4,5)P3 was reduced as compared to the control protein. PamGene® kinome profiling revealed multiple differentially phosphorylated kinase substrates, e.g. calmodulin, between the genotypes. Further in silico upstream kinase analysis predicted a large-scale impairment in activities of kinases participating, for example, in intracellular signal transduction, protein translation and cell cycle events. In conclusion, myotubes from p.P50T/AKT2 variant carriers show multiple signalling alterations which may contribute to predisposition to insulin resistance and T2D in the carriers of this signalling variant.
Collapse
Affiliation(s)
- Selina Mäkinen
- Minerva Foundation Institute for Medical Research, Tukholmankatu, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Haartmaninkatu, Helsinki, Finland
| | - Neeta Datta
- Minerva Foundation Institute for Medical Research, Tukholmankatu, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Haartmaninkatu, Helsinki, Finland
| | - Savithri Rangarajan
- Pam Gene International B.V., Wolvenhoek, BJ ´s-Hertogenbosch, The Netherlands
| | - Yen H Nguyen
- Minerva Foundation Institute for Medical Research, Tukholmankatu, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Haartmaninkatu, Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Tukholmankatu, Helsinki, Finland
- Department of Anatomy, Faculty of Medicine, Haartmaninkatu, University of Helsinki, Helsinki, Finland
| | - Aino Latva-Rasku
- Turku PET Centre, University of Turku, Kiinamyllynkatu, Turku, Finland
- Turku PET Centre, Turku University Hospital, Kiinamyllynkatu, Turku, Finland
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, Kiinamyllynkatu, Turku, Finland
- Turku PET Centre, Turku University Hospital, Kiinamyllynkatu, Turku, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Puijonlaaksontie, Kuopio, Finland
| | - Heikki A Koistinen
- Minerva Foundation Institute for Medical Research, Tukholmankatu, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Haartmaninkatu, Helsinki, Finland
- Correspondence should be addressed to H A Koistinen:
| |
Collapse
|
10
|
Kerketta R, Erasmus MF, Wilson BS, Halasz AM, Edwards JS. Spatial Stochastic Model of the Pre-B Cell Receptor. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:683-693. [PMID: 35482702 PMCID: PMC10123485 DOI: 10.1109/tcbb.2022.3166149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Survival and proliferation of immature B lymphocytes requires expression and tonic signaling of the pre-B cell receptor (pre-BCR). This low level, ligand-independent signaling is likely achieved through frequent, but short-lived, homo interactions. Tonic signaling is also central in the pathology of precursor B acute lymphoblastic leukemia (B-ALL). In order to understand how repeated, transient events can lead to sustained signaling and to assess the impact of receptor accumulation induced by the membrane landscape, we developed a spatial stochastic model of receptor aggregation and downstream signaling events. Our rule- and agent-based model builds on previous mature BCR signaling models and incorporates novel parameters derived from single particle tracking of pre-BCR on surfaces of two different B-ALL cell lines, 697 and Nalm6. Live cell tracking of receptors on the two cell lines revealed characteristic differences in their dimer dissociation rates and diffusion coefficients. We report here that these differences affect pre-BCR aggregation and consequent signal initiation events. Receptors on Nalm6 cells, which have a lower off-rate and lower diffusion coefficient, more frequently form higher order oligomers than pre-BCR on 697 cells, resulting in higher levels of downstream phosphorylation in the Nalm6 cell line.
Collapse
|
11
|
Integrin Signaling Shaping BTK-Inhibitor Resistance. Cells 2022; 11:cells11142235. [PMID: 35883678 PMCID: PMC9322986 DOI: 10.3390/cells11142235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
Integrins are adhesion molecules that function as anchors in retaining tumor cells in supportive tissues and facilitating metastasis. Beta1 integrins are known to contribute to cell adhesion-mediated drug resistance in cancer. Very late antigen-4 (VLA-4), a CD49d/CD29 heterodimer, is a beta1 integrin implicated in therapy resistance in both solid tumors and haematological malignancies such as chronic lymphocytic leukemia (CLL). A complex inside-out signaling mechanism activates VLA-4, which might include several therapeutic targets for CLL. Treatment regimens for this disease have recently shifted towards novel agents targeting BCR signaling. Bruton’s tyrosine kinase (BTK) is a component of B cell receptor signaling and BTK inhibitors such as ibrutinib are highly successful; however, their limitations include indefinite drug administration, the development of therapy resistance, and toxicities. VLA-4 might be activated independently of BTK, resulting in an ongoing interaction of CD49d-expressing leukemic cells with their surrounding tissue, which may reduce the success of therapy with BTK inhibitors and increases the need for alternative therapies. In this context, we discuss the inside-out signaling cascade culminating in VLA-4 activation, consider the advantages and disadvantages of BTK inhibitors in CLL and elucidate the mechanisms behind cell adhesion-mediated drug resistance.
Collapse
|
12
|
Chen R, Zhou D, Wang L, Zhu L, Ye X. MYD88L265P and CD79B double mutations type (MCD type) of diffuse large B-cell lymphoma: mechanism, clinical characteristics, and targeted therapy. Ther Adv Hematol 2022; 13:20406207211072839. [PMID: 35126963 PMCID: PMC8808040 DOI: 10.1177/20406207211072839] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/15/2021] [Indexed: 12/15/2022] Open
Abstract
MYD88/CD79B-mutated (MCD) genotype is a genetic subgroup of diffuse large B-cell lymphoma (DLBCL) with the co-occurrence of MYD88L265P and CD79B mutations. MCD genotype is characterized by poor prognosis and extranodal involvement especially in immune-privileged sites. MCD model is dominated by activated B-cell (ABC)-like subtype of DLBCLs. It is generally accepted that the pathogenesis of MCD DLBCL mainly includes chronic active B-cell receptor (BCR) signaling and oncogenic MYD88 mutations, which drives pathological nuclear factor kappa B (NF-κB) activation in MCD lymphoid malignancies. CD79B and MYD88L265P mutations are frequently and contemporaneously founded in B-cell malignancies. The collaboration of the two mutations may explain the unique biology of MCD. Meanwhile, standard immunochemotherapy combine with different targeted therapies worth further study to improve the prognosis of MCD, according to genetic, phenotypic, and clinical features of MCD type. In this review, we systematically described mechanism, clinical characteristics, and targeted therapy of MCD DLBCL.
Collapse
Affiliation(s)
- Rongrong Chen
- Program in Clinical Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - De Zhou
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lulu Wang
- Program in Clinical Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lixia Zhu
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xiujin Ye
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China
| |
Collapse
|
13
|
Khan N, Hu Y, Lowell CA, Rothstein TL. Signal Integration by Translocation and Phosphorylation of PKCδ in the B Cell Alternate Pathway. THE JOURNAL OF IMMUNOLOGY 2021; 207:2288-2296. [PMID: 34588218 DOI: 10.4049/jimmunol.2100295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/30/2021] [Indexed: 11/19/2022]
Abstract
B cell signaling for activation via the BCR occurs as an isolated event only in vitro; in real life, BCR signaling takes place within a complex milieu that involves interactions with agents that trigger additional receptors. Chief among these is IL-4. We have shown that BCR signaling is reprogrammed by IL-4 receptor engagement and that this reprogramming involves creation of a new, signalosome-independent, Lyn-dependent alternate signaling pathway in B cells isolated from BALB/cByJ mice. A unique aspect of the alternate pathway is protein kinase Cδ (PKCδ) phosphorylation. In dissecting this pathway, we unexpectedly found that Lyn is associated with IL-4Rα, that IL-4 induces Lyn activation, and that Lyn immunoprecipitated from IL-4-treated B cells capably phosphorylates PKCδ in a cell-free system. However, PKCδ phosphorylation does not occur in the absence of BCR triggering in vivo. This raised the question of why IL-4 alone failed to produce PKCδ phosphorylation. We considered the possibility that Lyn and PKCδ may be spatially separated. As expected, before any treatment, Lyn is located primarily in the membrane fraction, whereas PKCδ is located mainly in the cytosol fraction. However, when anti-Ig follows IL-4 treatment, PKCδ is found in the membrane fraction and phosphorylated. This translocation of PKCδ to the membrane fraction is not affected by loss of Lyn, although PKCδ phosphorylation requires Lyn. Thus, PKCδ phosphorylation through the alternate pathway represents the result of signal integration, whereby neither IL-4 nor anti-Ig working alone produces this outcome, but together they achieve this result by Lyn activation (IL-4) and PKCδ translocation (IL-4 followed by anti-Ig).
Collapse
Affiliation(s)
- Naeem Khan
- Center for Immunobiology, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI
| | - Yongmei Hu
- Department of Laboratory Medicine, University of California at San Francisco School of Medicine, San Francisco, CA; and
| | - Clifford A Lowell
- Department of Laboratory Medicine, University of California at San Francisco School of Medicine, San Francisco, CA; and
| | - Thomas L Rothstein
- Center for Immunobiology, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI; .,Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI
| |
Collapse
|
14
|
Liang X, Cao Y, Li C, Yu H, Yang C, Liu H. MALT1 as a promising target to treat lymphoma and other diseases related to MALT1 anomalies. Med Res Rev 2021; 41:2388-2422. [PMID: 33763890 DOI: 10.1002/med.21799] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/23/2020] [Accepted: 03/03/2021] [Indexed: 12/25/2022]
Abstract
Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) is a key adaptor protein that regulates the NF-κB pathway, in which MALT1 functions as a scaffold protein and protease to trigger downstream signals. The abnormal expression of MALT1 is closely associated with lymphomagenesis and other diseases, including solid tumors and autoimmune diseases. MALT1 is the only protease in the underlying pathogenesis of these diseases, and its proteolytic activity can be pharmacologically regulated. Therefore, MALT1 is a potential and promising target for anti-lymphoma and other MALT1-related disease treatments. Currently, the development of MALT1 inhibitors is still in its early stages. This review presents an overview of MALT1, particularly its X-ray structures and biological functions, and elaborates on the pathogenesis of diseases associated with its dysregulation. We then summarize previously reported MALT1 inhibitors, focusing on their molecular structure, biological activity, structure-activity relationship, and limitations. Finally, we propose future research directions to accelerate the discovery of novel MALT1 inhibitors with clinical applications. Overall, this review provides a comprehensive and systematic overview of MALT1-related research advances and serves as a theoretical basis for drug discovery and research.
Collapse
Affiliation(s)
- Xuewu Liang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - YiChun Cao
- School of Pharmacy, Fudan University, Shanghai, China
| | - Chunpu Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Haolan Yu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Chenghua Yang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hong Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
15
|
Rudd CE. How the Discovery of the CD4/CD8-p56 lck Complexes Changed Immunology and Immunotherapy. Front Cell Dev Biol 2021; 9:626095. [PMID: 33791292 PMCID: PMC8005572 DOI: 10.3389/fcell.2021.626095] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/11/2021] [Indexed: 12/22/2022] Open
Abstract
The past 25 years have seen enormous progress in uncovering the receptors and signaling mechanisms on T-cells that activate their various effecter functions. Until the late 1980s, most studies on T-cells had focused on the influx of calcium and the levels of cAMP/GMP in T-cells. My laboratory then uncovered the interaction of CD4 and CD8 co-receptors with the protein-tyrosine kinase p56lck which are now widely accepted as the initiators of the tyrosine phosphorylation cascade leading to T-cell activation. The finding explained how immune recognition receptors expressed by many immune cells, which lack intrinsic catalytic activity, can transduce activation signals via non-covalent association with non-receptor tyrosine kinases. The discovery also established the concept that a protein tyrosine phosphorylation cascade operated in T-cells. In this vein, we and others then showed that the CD4- and CD8-p56lck complexes phosphorylate the TCR complexes which led to the identification of other protein-tyrosine kinases such as ZAP-70 and an array of substrates that are now central to studies in T-cell immunity. Other receptors such as B-cell receptor, Fc receptors and others were also subsequently found to use src kinases to control cell growth. In T-cells, p56lck driven phosphorylation targets include co-receptors such as CD28 and CTLA-4 and immune cell-specific adaptor proteins such as LAT and SLP-76 which act to integrate signals proximal to surface receptors. CD4/CD8-p56lck regulated events in T-cells include intracellular calcium mobilization, integrin activation and the induction of transcription factors for gene expression. Lastly, the identification of the targets of p56lck in the TCR and CD28 provided the framework for the development of chimeric antigen receptor (CAR) therapy in the treatment of cancer. In this review, I outline a history of the development of events that led to the development of the "TCR signaling paradigm" and its implications to immunology and immunotherapy.
Collapse
Affiliation(s)
- Christopher E. Rudd
- Division of Immunology-Oncology, Centre de Recherche Hôpital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Faculty of Medicine, Universite de Montreal, Montreal, QC, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University Health Center, McGill University, Montreal, QC, Canada
| |
Collapse
|
16
|
Pierce CF, Brown VR, Olsen SC, Boggiatto P, Pedersen K, Miller RS, Speidel SE, Smyser TJ. Loci Associated With Antibody Response in Feral Swine ( Sus scrofa) Infected With Brucella suis. Front Vet Sci 2020; 7:554674. [PMID: 33324693 PMCID: PMC7724110 DOI: 10.3389/fvets.2020.554674] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 10/23/2020] [Indexed: 11/13/2022] Open
Abstract
Feral swine (Sus scrofa) are a destructive invasive species widespread throughout the United States that disrupt ecosystems, damage crops, and carry pathogens of concern for the health of domestic stock and humans including Brucella suis-the causative organism for swine brucellosis. In domestic swine, brucellosis results in reproductive failure due to abortions and infertility. Contact with infected feral swine poses spillover risks to domestic pigs as well as humans, companion animals, wildlife, and other livestock. Genetic factors influence the outcome of infectious diseases; therefore, genome wide association studies (GWAS) of differential immune responses among feral swine can provide an understanding of disease dynamics and inform management to prevent the spillover of brucellosis from feral swine to domestic pigs. We sought to identify loci associated with differential antibody responses among feral swine naturally infected with B. suis using a case-control GWAS. Tissue, serum, and genotype data (68,516 bi-allelic single nucleotide polymorphisms) collected from 47 feral swine were analyzed in this study. The 47 feral swine were culture positive for Brucella spp. Of these 47, 16 were antibody positive (cases) whereas 31 were antibody negative (controls). Single-locus GWAS were performed using efficient mixed-model association eXpedited (EMMAX) methodology with three genetic models: additive, dominant, and recessive. Eight loci associated with seroconversion were identified on chromosome 4, 8, 9, 10, 12, and 18. Subsequent bioinformatic analyses revealed nine putative candidate genes related to immune function, most notably phagocytosis and induction of an inflammatory response. Identified loci and putative candidate genes may play an important role in host immune responses to B. suis infection, characterized by a detectable bacterial presence yet a differential antibody response. Given that antibody tests are used to evaluate brucellosis infection in domestic pigs and for disease surveillance in invasive feral swine, additional studies are needed to fully understand the genetic component of the response to B. suis infection and to more effectively translate estimates of Brucella spp. antibody prevalence among feral swine to disease control management action.
Collapse
Affiliation(s)
- Courtney F. Pierce
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Wildlife Research Center, Fort Collins, CO, United States
- Department of Animal Sciences, Colorado State University, Fort Collins, CO, United States
| | - Vienna R. Brown
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Feral Swine Damage Management Program, Fort Collins, CO, United States
| | - Steven C. Olsen
- United States Department of Agriculture, Agricultural Research Service, Infectious Bacterial Diseases, National Animal Disease Center, Ames, IA, United States
| | - Paola Boggiatto
- United States Department of Agriculture, Agricultural Research Service, Infectious Bacterial Diseases, National Animal Disease Center, Ames, IA, United States
| | - Kerri Pedersen
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, Raleigh, NC, United States
| | - Ryan S. Miller
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, Center for Epidemiology and Animal Health, Fort Collins, CO, United States
| | - Scott E. Speidel
- Department of Animal Sciences, Colorado State University, Fort Collins, CO, United States
| | - Timothy J. Smyser
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Wildlife Research Center, Fort Collins, CO, United States
| |
Collapse
|
17
|
Src Family Protein Kinase Controls the Fate of B Cells in Autoimmune Diseases. Inflammation 2020; 44:423-433. [PMID: 33037966 DOI: 10.1007/s10753-020-01355-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/07/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023]
Abstract
There are more than 80 kinds of autoimmune diseases known at present, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), systemic sclerosis (SSc), inflammatory bowel disease (IBD), as well as other disorders. Autoimmune diseases have a characteristic of immune responses directly attacking own tissues, leading to systematic inflammation and subsequent tissue damage. B cells play a vital role in the development of autoimmune diseases and differentiate into plasma cells or memory B cells to secrete high-affinity antibody or provide long-lasting function. Drugs targeting B cells show good therapeutic effects for the treatment of autoimmune diseases, such as rituximab (anti-CD20 antibody). Src family protein kinases (SFKs) are believed to play important roles in a variety of cellular functions such as growth, proliferation, and differentiation of B cell via B cell antigen receptor (BCR). Lck/Yes-related novel protein tyrosine kinase (LYN), BLK (B lymphocyte kinase), and Fyn are three different kinds of SFKs mainly expressed in B cells. LYN has a dual role in the BCR signal. On the one hand, positive signals are beneficial to the development and maturation of B cells. On the other hand, LYN can also inhibit excessively activated B cells. BLK is involved in the proliferation, differentiation, and immune tolerance of B lymphocytes, and further affects the function of B cells, which may lead to autoreactive or regulatory cellular responses, increasing the risk of autoimmune diseases. Fyn may affect the development of autoimmune disorders via the differentiation of B cells in the early stage of B cell development. This article reviews the recent advances of SFKs in B lymphocytes in autoimmune diseases.
Collapse
|
18
|
Visco C, Tanasi I, Quaglia FM, Ferrarini I, Fraenza C, Krampera M. Oncogenic Mutations of MYD88 and CD79B in Diffuse Large B-Cell Lymphoma and Implications for Clinical Practice. Cancers (Basel) 2020; 12:E2913. [PMID: 33050534 PMCID: PMC7600909 DOI: 10.3390/cancers12102913] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/01/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common non-Hodgkin's lymphoma in adults. Despite the recognition of transcriptional subtypes with distinct functional characteristics, patient outcomes have not been substantially altered since the advent of chemoimmunotherapy (CIT) twenty years ago. Recently, a few pivotal studies added to the disease heterogeneity by describing several activating mutations, which have been associated with disease presentation, B-cell function and behavior, and final outcome. DLBCL arises from antigen exposed B-cells, with the B-cell receptor (BCR) playing a central role. BCR-activity related mutations, such as CD79B and MYD88, are responsible for chronic activation of the BCR in a substantial subset of patients. These mutations, often coexisting in the same patient, have been found in a substantial subset of patients with immune-privileged (IP) sites DLBCLs, and are drivers of lymphoma development conferring tissue-specific homing properties. Both mutations have been associated with disease behavior, including tumor response either to CIT or to BCR-targeted therapy. The recognition of CD79B and MYD88 mutations will contribute to the heterogeneity of the disease, both in recognizing the BCR as a potential therapeutic target and in providing genetic tools for personalized treatment.
Collapse
Affiliation(s)
- Carlo Visco
- Correspondence: (C.V.); (I.T.); Tel.: +39-0458124797 (C.V.); +39-0458128418 (I.T.)
| | - Ilaria Tanasi
- Correspondence: (C.V.); (I.T.); Tel.: +39-0458124797 (C.V.); +39-0458128418 (I.T.)
| | | | | | | | | |
Collapse
|
19
|
He S, Zheng G, Yang X, Dong J, Zhou D, Venugopal N, Yao Y, Cheng Z. Avian leukosis virus subgroup J induces B cell anergy mediated by Lyn inhibited BCR signal transduction. Vet Microbiol 2020; 247:108781. [PMID: 32768227 DOI: 10.1016/j.vetmic.2020.108781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/23/2020] [Accepted: 06/28/2020] [Indexed: 12/22/2022]
Abstract
Immune tolerance induced by avian leukosis virus subgroup J (ALV-J) is a prerequisite for tumorigenesis. Although we had reported that B cell anergy induced by ALV-J was the main reason for immune tolerance, the molecular mechanism still remains unclear. Here, we found SU protein of ALV-J interacted with tyrosine kinase Lyn (a key protein in BCR signaling pathway) by confocal laser scanning microscopy and co-immunoprecipitation test, which suggested that Lyn might play an important role in B cell anergy induced by ALV-J. Correspondingly, the mRNA and protein level of Lyn was significantly up-regulated in B cells after ALV-J infection. Subsequently, the phosphorylated protein levels of Lyn at Tyr507 site were significantly up-regulated in ALV-J-infected B cells after BCR signal activation, but the phosphorylated protein level of Syk (a direct substrate of Lyn) at Tyr525/526 site, Ca2+ flux, and NF-κB p65 protein level were significantly down-regulated. Interestingly, the phosphorylated protein level of Syk at Tyr525/526 site, Ca2+ flux, and NF-κB p65 protein level were both significantly retrieved after the shLyn treatment in B cells infected by ALV-J. In summary, these results indicated that ALV-J activated the negative regulatory effect of phosphorylated Lyn protein at 507 site in BCR signal transduction pathway and then mediated B cell anergy, which will provide a new insight for revealing the pathogenesis of immune tolerance induced by ALV-J.
Collapse
Affiliation(s)
- Shuhai He
- College of Veterinary Medicine, Shandong Agricultural University, No 61, Daizong Street, Tai'an City, Shandong Province, 271018, China; College of Husbandry and Veterinary, Xinyang Agriculture and Forestry University, No 1, North Ring Road, Xinyang City, Henan Province, 464000, China.
| | - Gaoying Zheng
- College of Veterinary Medicine, Shandong Agricultural University, No 61, Daizong Street, Tai'an City, Shandong Province, 271018, China.
| | - Xiaoxia Yang
- Hospital of Shandong Agricultural University, No 61, Daizong Street, Tai'an City, Shandong Province, 271018, China.
| | - Jianguo Dong
- College of Husbandry and Veterinary, Xinyang Agriculture and Forestry University, No 1, North Ring Road, Xinyang City, Henan Province, 464000, China.
| | - Defang Zhou
- College of Veterinary Medicine, Shandong Agricultural University, No 61, Daizong Street, Tai'an City, Shandong Province, 271018, China.
| | - Nair Venugopal
- The Pirbright Institute & UK-China Centre of Excellence on Avian Disease Research, Pirbright, Ash Road, Guildford, Surrey, GU24 0NF, UK.
| | - Yongxiu Yao
- The Pirbright Institute & UK-China Centre of Excellence on Avian Disease Research, Pirbright, Ash Road, Guildford, Surrey, GU24 0NF, UK.
| | - Ziqiang Cheng
- College of Veterinary Medicine, Shandong Agricultural University, No 61, Daizong Street, Tai'an City, Shandong Province, 271018, China.
| |
Collapse
|
20
|
Zhao T, Luo K, Liu X, Dong S, Han Y, Gou M, Su P, Li Q, Zhu T. A novel Lyn-like protein tyrosine kinase identified in lamprey and its role in immune response. Acta Biochim Biophys Sin (Shanghai) 2020; 52:573-575. [PMID: 32286610 DOI: 10.1093/abbs/gmaa022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 11/09/2018] [Indexed: 02/06/2023] Open
Affiliation(s)
- Tingting Zhao
- College of Life Science, Liaoning Normal University, Dalian 116081, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116081, China
| | - Kedi Luo
- College of Life Science, Liaoning Normal University, Dalian 116081, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116081, China
| | - Xin Liu
- College of Life Science, Liaoning Normal University, Dalian 116081, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116081, China
| | - Shanshan Dong
- College of Life Science, Liaoning Normal University, Dalian 116081, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116081, China
- Green Biomedical Technology (Tianjin) Co., Ltd., Tianjin 301700, China
| | - Yinglun Han
- College of Life Science, Liaoning Normal University, Dalian 116081, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116081, China
| | - Meng Gou
- College of Life Science, Liaoning Normal University, Dalian 116081, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116081, China
| | - Peng Su
- College of Life Science, Liaoning Normal University, Dalian 116081, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116081, China
| | - Qingwei Li
- College of Life Science, Liaoning Normal University, Dalian 116081, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116081, China
| | - Ting Zhu
- College of Life Science, Liaoning Normal University, Dalian 116081, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116081, China
| |
Collapse
|
21
|
Yamada K, Huang ZQ, Raska M, Reily C, Anderson JC, Suzuki H, Kiryluk K, Gharavi AG, Julian BA, Willey CD, Novak J. Leukemia Inhibitory Factor Signaling Enhances Production of Galactose-Deficient IgA1 in IgA Nephropathy. KIDNEY DISEASES 2020; 6:168-180. [PMID: 32523959 DOI: 10.1159/000505748] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022]
Abstract
Objectives IgA nephropathy (IgAN) is thought to involve an autoimmune process wherein galactose-deficient IgA1 (Gd-IgA1), recognized as autoantigen by autoantibodies, forms pathogenic immune complexes. Mounting evidence has implicated abnormal activation of some protein-tyrosine kinases (PTKs) in IgAN. Furthermore, genome-wide association studies (GWAS) of IgAN provided insight into disease pathobiology and genetics. A GWAS locus on chromosome 22q12 contains genes encoding leukemia inhibitory factor (LIF) and oncostatin M, interleukin (IL)-6-related cytokines implicated in mucosal immunity and inflammation. We have previously shown that IL-6 mediates overproduction of Gd-IgA1 through aberrant STAT3 activation. Here, we show that LIF enhanced production of Gd-IgA1 in IgA1-secreting cells of patients with IgAN and provide initial analyses of LIF signaling. Methods We characterized LIF signaling that is involved in the overproduction of Gd-IgA1, using IgA1-secreting cell lines derived from peripheral blood of patients with IgAN and healthy controls (HC). We used global PTK activity profiling, immunoblotting, lectin ELISA, and siRNA knock-down. Results LIF stimulation did not significantly affect production of total IgA1 in IgA1-secreting cells from patients with IgAN or HC. However, LIF increased production of Gd-IgA1, but only in the cells from patients with IgAN. LIF stimulation enhanced phosphorylation of STAT1 in IgA1-secreting cells from patients with IgAN to a higher degree than in the cells from HC. siRNA knock-down of STAT1 blocked LIF-mediated overproduction of Gd-IgA1. Unexpectedly, this abnormal phosphorylation of STAT1 in IgA1-secreting cells from patients with IgAN was not mediated by JAK, but rather involved activation of Src-family PTKs (SFKs). Conclusion Abnormal LIF/STAT1 signaling represents another pathway potentially leading to overproduction of Gd-IgA1 in IgAN, providing possible explanation for the phenotype associated with chromosome 22q12 GWAS locus. Abnormal LIF/STAT1 signaling and the associated SFKs may represent potential diagnostic and/or therapeutic targets in IgAN.
Collapse
Affiliation(s)
- Koshi Yamada
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Zhi Qiang Huang
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Milan Raska
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Immunology, Palacky University Olomouc, Olomouc, Czechia
| | - Colin Reily
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Joshua C Anderson
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hitoshi Suzuki
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Krzysztof Kiryluk
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Ali G Gharavi
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Bruce A Julian
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Christopher D Willey
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
22
|
Vairy S, Tran TH. IKZF1 alterations in acute lymphoblastic leukemia: The good, the bad and the ugly. Blood Rev 2020; 44:100677. [PMID: 32245541 DOI: 10.1016/j.blre.2020.100677] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 02/20/2020] [Accepted: 02/28/2020] [Indexed: 12/16/2022]
Abstract
Advances in genomics have deepened our understanding of the biology of acute lymphoblastic leukemia (ALL), defined novel molecular leukemia subtypes, discovered new prognostic biomarkers and paved the way to emerging molecularly targeted therapeutic avenues. Since its discovery, IKZF1 has generated significant interest within the leukemia scientific community.IKZF1 plays a critical role in lymphoid development and its alterations cooperate to mediate leukemogenesis. IKZF1 alterations are present in approximately 15% of childhood ALL, rise in prevalence among adults with ALL and become highly enriched within kinase-driven ALL. A cumulating body of literature has highlighted the adverse prognostic impact of IKZF1 alterations in both Philadelphia chromosome (Ph)-negative and Ph-driven ALL. IKZF1 alterations thus emerge as an important prognostic biomarker in ALL. This article aims to provide a state-of-the-art review focusing on the prognostic clinical relevance of IKZF1 alterations in ALL, as well as current and future therapeutic strategies targeting IKZF1-altered ALL.
Collapse
Affiliation(s)
- Stephanie Vairy
- Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Center, CHU Sainte-Justine, Montréal, Québec, Canada
| | - Thai Hoa Tran
- Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Center, CHU Sainte-Justine, Montréal, Québec, Canada.
| |
Collapse
|
23
|
Hou Y, Hu T, Wei D, Gao J, Che D, Wang X, Wang C, He H. (-)-Asarinin inhibits mast cells activation as a Src family kinase inhibitor. Int J Biochem Cell Biol 2020; 121:105701. [PMID: 32007602 DOI: 10.1016/j.biocel.2020.105701] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 01/17/2020] [Accepted: 01/27/2020] [Indexed: 01/17/2023]
Abstract
As one of the major global health issues, allergic disease represents a considerable burden both on individual patients and public health. (-)-Asarinin (Asa), a lignan isolated from the roots of Asiasari radix, was reported to be associated with anti-allergic effect, but its efficacy and mechanism of action remain unclear. This study investigated the inhibitory effect of Asa on allergic reaction and its mechanism of action. Asa significantly suppressed Ag-sensitized human mast cell line LAD2 calcium mobilization, degranulation, and secretion. It also could reduce OVA-induced local and system anaphylaxis of mice in vivo. Further experiments revealed that Asa inhibit the mast cell activation by preventing the phosphorylation of Src family kinases. Moreover, after the IgE-dependent murine model of allergic rhinitis was treated with Asa, not only the concentration of histamine, total IgE, and IL-4 decreased, but also the inflammatory infiltrates and nasal mucosa incrassation were attenuated significantly. Meanwhile, Asa also inhibited the activation of mast cells induced by Compound48/80 in vivo and in vitro. In conclusion, Asa may serve as a potential novel Src family kinase inhibitor to inhibit IgE-dependent andIgE-independent allergic reaction and treat anaphylactic disease.
Collapse
Affiliation(s)
- Yajing Hou
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061 China
| | - Tian Hu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061 China
| | - Di Wei
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061 China
| | - Jiapan Gao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061 China
| | - Delu Che
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061 China
| | - Xiangjun Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061 China
| | - Cheng Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061 China
| | - Huaizhen He
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061 China.
| |
Collapse
|
24
|
Epigenetics evaluation of the oncogenic mechanisms of two closely related bovine and human deltaretroviruses: A system biology study. Microb Pathog 2020; 139:103845. [DOI: 10.1016/j.micpath.2019.103845] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 10/26/2019] [Accepted: 11/03/2019] [Indexed: 12/12/2022]
|
25
|
'Piperazining' the catalytic gatekeepers: unraveling the pan-inhibition of SRC kinases; LYN, FYN and BLK by masitinib. Future Med Chem 2019; 11:2365-2380. [PMID: 31516031 DOI: 10.4155/fmc-2018-0354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: Blocking oncogenic signaling of B-cell receptor (BCR) has been explored as a viable strategy in the treatment of diffuse large B-cell lymphoma. Masitinib is shown to multitarget LYN, FYN and BLK kinases that propagate BCR signals to downstream effectors. However, the molecular mechanisms of its selectivity and pan-inhibition remain elusive. Materials & methods: This study therefore employed molecular dynamics simulations coupled with advanced post-molecular dynamics simulation techniques to unravel the structural mechanisms that inform the reported multitargeting ability of masitinib. Results: Molecular dynamics simulations revealed initial selective targeting of catalytic residues (Asp334/Glu335 - LYN; Asp130/Asp148/Glu54 - FYN; Asp89 - BLK) by masitinib, with high-affinity interactions via its piperazine ring at the entrance of the ATP-binding pockets, before systematic access into the hydrophobic deep pocket grooves. Conclusion: Identification of these 'gatekeeper' residues could open up a novel paradigm of structure-based design of highly selective pan-inhibitors of BCR signaling in the treatment of diffuse large B-cell lymphoma.
Collapse
|
26
|
Abstract
Ebola virus (EBOV) is a single-stranded RNA virus that causes Ebola virus disease (EVD), characterized by excessive inflammation, lymphocyte apoptosis, hemorrhage, and coagulation defects leading to multiorgan failure and shock. Recombinant vesicular stomatitis virus expressing the EBOV glycoprotein (VSV-EBOV), which is highly efficacious against lethal challenge in nonhuman primates, is the only vaccine that successfully completed a phase III clinical trial. Additional studies showed VSV-EBOV provides complete and partial protection to macaques immunized 7 and 3 days before EBOV challenge, respectively. However, the mechanisms by which this live-attenuated vaccine elicits rapid protection are only partially understood. To address this, we carried out a longitudinal transcriptome analysis of host responses in whole-blood samples collected from cynomolgus macaques vaccinated with VSV-EBOV 28, 21, 14, 7, and 3 days before EBOV challenge. Our findings indicate the transcriptional response to the vaccine peaks 7 days following vaccination and contains signatures of both innate antiviral immunity as well as B-cell activation. EBOV challenge 1 week after vaccination resulted in large gene expression changes suggestive of a recall adaptive immune response 14 days postchallenge. Lastly, the timing and magnitude of innate immunity and interferon-stimulated gene expression correlated with viral burden and disease outcome in animals vaccinated 3 days before challenge.IMPORTANCE Ebola virus (EBOV) is the causative agent of Ebola virus disease (EVD), a deadly disease and major public health threat worldwide. A safe and highly efficacious vesicular stomatitis virus-based vaccine against EBOV is the only platform that has successfully completed phase III clinical trials and has been used in recent and ongoing outbreaks. Earlier studies showed that antibodies are the main mode of protection when this vaccine is administered 28 days before EBOV challenge. Recently, we showed this vaccine can provide protection when administered as early as 3 days before challenge and before antibodies are detected. This study seeks to identify the mechanisms of rapid protection, which in turn will pave the way for improved vaccines and therapeutics. Additionally, this study provides insight into host gene expression signatures that could provide early biomarkers to identify infected individuals who are at highest risk of poor outcomes.
Collapse
|
27
|
Pavlovic S, Kotur N, Stankovic B, Zukic B, Gasic V, Dokmanovic L. Pharmacogenomic and Pharmacotranscriptomic Profiling of Childhood Acute Lymphoblastic Leukemia: Paving the Way to Personalized Treatment. Genes (Basel) 2019; 10:E191. [PMID: 30832275 PMCID: PMC6471971 DOI: 10.3390/genes10030191] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/23/2019] [Accepted: 02/25/2019] [Indexed: 02/07/2023] Open
Abstract
Personalized medicine is focused on research disciplines which contribute to the individualization of therapy, like pharmacogenomics and pharmacotranscriptomics. Acute lymphoblastic leukemia (ALL) is the most common malignancy of childhood. It is one of the pediatric malignancies with the highest cure rate, but still a lethal outcome due to therapy accounts for 1%⁻3% of deaths. Further improvement of treatment protocols is needed through the implementation of pharmacogenomics and pharmacotranscriptomics. Emerging high-throughput technologies, including microarrays and next-generation sequencing, have provided an enormous amount of molecular data with the potential to be implemented in childhood ALL treatment protocols. In the current review, we summarized the contribution of these novel technologies to the pharmacogenomics and pharmacotranscriptomics of childhood ALL. We have presented data on molecular markers responsible for the efficacy, side effects, and toxicity of the drugs commonly used for childhood ALL treatment, i.e., glucocorticoids, vincristine, asparaginase, anthracyclines, thiopurines, and methotrexate. Big data was generated using high-throughput technologies, but their implementation in clinical practice is poor. Research efforts should be focused on data analysis and designing prediction models using machine learning algorithms. Bioinformatics tools and the implementation of artificial i Lack of association of the CEP72 rs924607 TT genotype with intelligence are expected to open the door wide for personalized medicine in the clinical practice of childhood ALL.
Collapse
Affiliation(s)
- Sonja Pavlovic
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia.
| | - Nikola Kotur
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia.
| | - Biljana Stankovic
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia.
| | - Branka Zukic
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia.
| | - Vladimir Gasic
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia.
| | - Lidija Dokmanovic
- University Children's Hospital, 11000 Belgrade, Serbia.
- University of Belgrade, Faculty of Medicine, 11000 Belgrade, Serbia.
| |
Collapse
|
28
|
Petrovic J, Zhou Y, Fasolino M, Goldman N, Schwartz GW, Mumbach MR, Nguyen SC, Rome KS, Sela Y, Zapataro Z, Blacklow SC, Kruhlak MJ, Shi J, Aster JC, Joyce EF, Little SC, Vahedi G, Pear WS, Faryabi RB. Oncogenic Notch Promotes Long-Range Regulatory Interactions within Hyperconnected 3D Cliques. Mol Cell 2019; 73:1174-1190.e12. [PMID: 30745086 DOI: 10.1016/j.molcel.2019.01.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/21/2018] [Accepted: 01/03/2019] [Indexed: 01/10/2023]
Abstract
Chromatin loops enable transcription-factor-bound distal enhancers to interact with their target promoters to regulate transcriptional programs. Although developmental transcription factors such as active forms of Notch can directly stimulate transcription by activating enhancers, the effect of their oncogenic subversion on the 3D organization of cancer genomes is largely undetermined. By mapping chromatin looping genome-wide in Notch-dependent triple-negative breast cancer and B cell lymphoma, we show that beyond the well-characterized role of Notch as an activator of distal enhancers, Notch regulates its direct target genes by instructing enhancer repositioning. Moreover, a large fraction of Notch-instructed regulatory loops form highly interacting enhancer and promoter spatial clusters termed "3D cliques." Loss- and gain-of-function experiments show that Notch preferentially targets hyperconnected 3D cliques that regulate the expression of crucial proto-oncogenes. Our observations suggest that oncogenic hijacking of developmental transcription factors can dysregulate transcription through widespread effects on the spatial organization of cancer genomes.
Collapse
Affiliation(s)
- Jelena Petrovic
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yeqiao Zhou
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria Fasolino
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Naomi Goldman
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gregory W Schwartz
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maxwell R Mumbach
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Son C Nguyen
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kelly S Rome
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yogev Sela
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zachary Zapataro
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen C Blacklow
- Department of Biological Chemistry, Harvard Medical School, Boston, MA 02215, USA
| | | | - Junwei Shi
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jon C Aster
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Eric F Joyce
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shawn C Little
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Golnaz Vahedi
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Warren S Pear
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Robert B Faryabi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
29
|
Schroeder HW, Imboden JB, Torres RM. Antigen Receptor Genes, Gene Products, and Coreceptors. Clin Immunol 2019. [DOI: 10.1016/b978-0-7020-6896-6.00004-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
30
|
Martini V, Frezzato F, Severin F, Raggi F, Trimarco V, Martinello L, Molfetta R, Visentin A, Facco M, Semenzato G, Paolini R, Trentin L. Abnormal regulation of BCR signalling by c-Cbl in chronic lymphocytic leukaemia. Oncotarget 2018; 9:32219-32231. [PMID: 30181811 PMCID: PMC6114956 DOI: 10.18632/oncotarget.25951] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/21/2018] [Indexed: 11/25/2022] Open
Abstract
Abnormalities of molecules involved in signal transduction pathways are connected to Chronic Lymphocytic Leukemia (CLL) pathogenesis and a critical role has been already ascribed to B-Cell Receptor (BCR)-Lyn axis. E3 ubiquitin ligase c-Cbl, working together with adapter protein CIN85, controls the degradation of protein kinases involved in BCR signaling. To investigate cell homeostasis in CLL, we studied c-Cbl since in normal B cells it is involved in the ubiquitin-dependent Lyn degradation and in the down-regulation of BCR signaling. We found that c-Cbl is overexpressed and not ubiquitinated after BCR engagement. We observed that c-Cbl did not associate to CIN85 in CLL with respect to normal B cells at steady state, nor following BCR engagement. c-Cbl association to Lyn was not detectable in CLL after BCR stimulation, as it happens in normal B cells. In some CLL patients, c-Cbl is constitutively phosphorylated at Y731 and in the same subjects, it associated to regulatory subunit p85 of PI3K. Moreover, c-Cbl is constitutive associated to Cortactin in those CLL patients presenting Cortactin overexpression and bad prognosis. These results support the hypothesis that c-Cbl, rather than E3 ligase activity, could have an adaptor function in turn influencing cell homeostasis in CLL.
Collapse
Affiliation(s)
- Veronica Martini
- Department of Medicine, Hematology and Clinical Immunology Branch, University School of Medicine, Padua, Italy.,Venetian Institute of Molecular Medicine, VIMM, Padua, Italy
| | - Federica Frezzato
- Department of Medicine, Hematology and Clinical Immunology Branch, University School of Medicine, Padua, Italy.,Venetian Institute of Molecular Medicine, VIMM, Padua, Italy
| | - Filippo Severin
- Department of Medicine, Hematology and Clinical Immunology Branch, University School of Medicine, Padua, Italy.,Venetian Institute of Molecular Medicine, VIMM, Padua, Italy
| | - Flavia Raggi
- Department of Medicine, Hematology and Clinical Immunology Branch, University School of Medicine, Padua, Italy.,Venetian Institute of Molecular Medicine, VIMM, Padua, Italy
| | - Valentina Trimarco
- Department of Medicine, Hematology and Clinical Immunology Branch, University School of Medicine, Padua, Italy.,Venetian Institute of Molecular Medicine, VIMM, Padua, Italy
| | - Leonardo Martinello
- Department of Medicine, Hematology and Clinical Immunology Branch, University School of Medicine, Padua, Italy.,Venetian Institute of Molecular Medicine, VIMM, Padua, Italy
| | - Rosa Molfetta
- Department of Molecular Medicine, University of La Sapienza, Rome, Italy
| | - Andrea Visentin
- Department of Medicine, Hematology and Clinical Immunology Branch, University School of Medicine, Padua, Italy.,Venetian Institute of Molecular Medicine, VIMM, Padua, Italy
| | - Monica Facco
- Department of Medicine, Hematology and Clinical Immunology Branch, University School of Medicine, Padua, Italy.,Venetian Institute of Molecular Medicine, VIMM, Padua, Italy
| | - Gianpietro Semenzato
- Department of Medicine, Hematology and Clinical Immunology Branch, University School of Medicine, Padua, Italy.,Venetian Institute of Molecular Medicine, VIMM, Padua, Italy
| | - Rossella Paolini
- Department of Molecular Medicine, University of La Sapienza, Rome, Italy
| | - Livio Trentin
- Department of Medicine, Hematology and Clinical Immunology Branch, University School of Medicine, Padua, Italy.,Venetian Institute of Molecular Medicine, VIMM, Padua, Italy
| |
Collapse
|
31
|
Hawkins LJ, Al-Attar R, Storey KB. Transcriptional regulation of metabolism in disease: From transcription factors to epigenetics. PeerJ 2018; 6:e5062. [PMID: 29922517 PMCID: PMC6005171 DOI: 10.7717/peerj.5062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/04/2018] [Indexed: 12/13/2022] Open
Abstract
Every cell in an individual has largely the same genomic sequence and yet cells in different tissues can present widely different phenotypes. This variation arises because each cell expresses a specific subset of genomic instructions. Control over which instructions, or genes, are expressed is largely controlled by transcriptional regulatory pathways. Each cell must assimilate a huge amount of environmental input, and thus it is of no surprise that transcription is regulated by many intertwining mechanisms. This large regulatory landscape means there are ample possibilities for problems to arise, which in a medical context means the development of disease states. Metabolism within the cell, and more broadly, affects and is affected by transcriptional regulation. Metabolism can therefore contribute to improper transcriptional programming, or pathogenic metabolism can be the result of transcriptional dysregulation. Here, we discuss the established and emerging mechanisms for controling transcription and how they affect metabolism in the context of pathogenesis. Cis- and trans-regulatory elements, microRNA and epigenetic mechanisms such as DNA and histone methylation, all have input into what genes are transcribed. Each has also been implicated in diseases such as metabolic syndrome, various forms of diabetes, and cancer. In this review, we discuss the current understanding of these areas and highlight some natural models that may inspire future therapeutics.
Collapse
Affiliation(s)
- Liam J Hawkins
- Institute of Biochemistry, Department of Biology, Carleton University, Ottawa, ON, Canada
| | - Rasha Al-Attar
- Institute of Biochemistry, Department of Biology, Carleton University, Ottawa, ON, Canada
| | - Kenneth B Storey
- Institute of Biochemistry, Department of Biology, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
32
|
Balaji S, Ahmed M, Lorence E, Yan F, Nomie K, Wang M. NF-κB signaling and its relevance to the treatment of mantle cell lymphoma. J Hematol Oncol 2018; 11:83. [PMID: 29907126 PMCID: PMC6002979 DOI: 10.1186/s13045-018-0621-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/28/2018] [Indexed: 02/07/2023] Open
Abstract
Mantle cell lymphoma is an aggressive subtype of non-Hodgkin B cell lymphoma that is characterized by a poor prognosis determined by Ki67 and Mantle Cell International Prognostic Index scores, but it is becoming increasingly treatable. The majority of patients, especially if young, achieve a progression-free survival of at least 5 years. Mantle cell lymphoma can initially be treated with an anti-CD20 antibody in combination with a chemotherapy backbone, such as VR-CAP (the anti-CD20 monoclonal antibody rituximab administered with cyclophosphamide, doxorubicin, and prednisone) or R-CHOP (the anti-CD20 monoclonal antibody rituximab administered with cyclophosphamide, doxorubicin, vincristine, and prednisone). While initial treatment can facilitate recovery and complete remission in a few patients, many patients experience relapsed or refractory mantle cell lymphoma within 2 to 3 years after initial treatment. Targeted agents such as ibrutinib, an inhibitor of Bruton’s tyrosine kinase, which has been approved only in the relapsed setting, can be used to treat patients with relapsed or refractory mantle cell lymphoma. However, mantle cell lymphoma cells often acquire resistance to such targeted agents and continue to survive by activating alternate signaling pathways such as the PI3K-Akt pathway or the NF-κB pathways. NF-κB is a transcription factor family that regulates the growth and survival of B cells; mantle cell lymphoma cells depend on NF-κB signaling for continued growth and proliferation. The NF-κB signaling pathways are categorized into canonical and non-canonical types, wherein the canonical pathway prompts inflammatory responses, immune regulation, and cell proliferation, while the non-canonical leads to B cell maturation and lymphoid organogenesis. Since these pathways upregulate survival genes and tumor-promoting cytokines, they can be activated to overcome the inhibitory effects of targeted agents, thereby having profound effects on tumorigenesis. The NF-κB pathways are also highly targetable in that they are interconnected with numerous other pathways, including B cell receptor signaling, PI3K/Akt/mTOR signaling, and toll-like receptor signaling pathways. Additionally, elements of the non-canonical NF- κB pathway, such as NF-κB-inducing kinase, can be targeted to overcome resistance to targeting of the canonical NF- κB pathway. Targeting the molecular mechanisms of the NF-κB pathways can facilitate the development of novel agents to treat malignancies and overcome drug resistance in patients with relapsed or refractory mantle cell lymphoma.
Collapse
Affiliation(s)
- Swathi Balaji
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 0429, Houston, TX, 77030-4009, USA
| | - Makhdum Ahmed
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 0429, Houston, TX, 77030-4009, USA
| | - Elizabeth Lorence
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 0429, Houston, TX, 77030-4009, USA
| | - Fangfang Yan
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 0429, Houston, TX, 77030-4009, USA
| | - Krystle Nomie
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 0429, Houston, TX, 77030-4009, USA
| | - Michael Wang
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 0429, Houston, TX, 77030-4009, USA.
| |
Collapse
|
33
|
Novel role of prostate apoptosis response-4 tumor suppressor in B-cell chronic lymphocytic leukemia. Blood 2018; 131:2943-2954. [PMID: 29695515 DOI: 10.1182/blood-2017-10-813931] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 04/08/2018] [Indexed: 01/04/2023] Open
Abstract
Prostate apoptosis response-4 (Par-4), a proapoptotic tumor suppressor protein, is downregulated in many cancers including renal cell carcinoma, glioblastoma, endometrial, and breast cancer. Par-4 induces apoptosis selectively in various types of cancer cells but not normal cells. We found that chronic lymphocytic leukemia (CLL) cells from human patients and from Eµ-Tcl1 mice constitutively express Par-4 in greater amounts than normal B-1 or B-2 cells. Interestingly, knockdown of Par-4 in human CLL-derived Mec-1 cells results in a robust increase in p21/WAF1 expression and decreased growth due to delayed G1-to-S cell-cycle transition. Lack of Par-4 also increased the expression of p21 and delayed CLL growth in Eμ-Tcl1 mice. Par-4 expression in CLL cells required constitutively active B-cell receptor (BCR) signaling, as inhibition of BCR signaling with US Food and Drug Administration (FDA)-approved drugs caused a decrease in Par-4 messenger RNA and protein, and an increase in apoptosis. In particular, activities of Lyn, a Src family kinase, spleen tyrosine kinase, and Bruton tyrosine kinase are required for Par-4 expression in CLL cells, suggesting a novel regulation of Par-4 through BCR signaling. Together, these results suggest that Par-4 may play a novel progrowth rather than proapoptotic role in CLL and could be targeted to enhance the therapeutic effects of BCR-signaling inhibitors.
Collapse
|
34
|
Pan-SRC kinase inhibition blocks B-cell receptor oncogenic signaling in non-Hodgkin lymphoma. Blood 2018; 131:2345-2356. [PMID: 29567799 DOI: 10.1182/blood-2017-10-809210] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 03/13/2018] [Indexed: 12/14/2022] Open
Abstract
In diffuse large B-cell lymphoma (DLBCL), activation of the B-cell receptor (BCR) promotes multiple oncogenic signals, which are essential for tumor proliferation. Inhibition of the Bruton's tyrosine kinase (BTK), a BCR downstream target, is therapeutically effective only in a subgroup of patients with DLBCL. Here, we used lymphoma cells isolated from patients with DLBCL to measure the effects of targeted therapies on BCR signaling and to anticipate response. In lymphomas resistant to BTK inhibition, we show that blocking BTK activity enhanced tumor dependencies from alternative oncogenic signals downstream of the BCR, converging on MYC upregulation. To completely ablate the activity of the BCR, we genetically and pharmacologically repressed the activity of the SRC kinases LYN, FYN, and BLK, which are responsible for the propagation of the BCR signal. Inhibition of these kinases strongly reduced tumor growth in xenografts and cell lines derived from patients with DLBCL independent of their molecular subtype, advancing the possibility to be relevant therapeutic targets in broad and diverse groups of DLBCL patients.
Collapse
|
35
|
Siveen KS, Prabhu KS, Achkar IW, Kuttikrishnan S, Shyam S, Khan AQ, Merhi M, Dermime S, Uddin S. Role of Non Receptor Tyrosine Kinases in Hematological Malignances and its Targeting by Natural Products. Mol Cancer 2018; 17:31. [PMID: 29455667 PMCID: PMC5817858 DOI: 10.1186/s12943-018-0788-y] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/01/2018] [Indexed: 12/12/2022] Open
Abstract
Tyrosine kinases belong to a family of enzymes that mediate the movement of the phosphate group to tyrosine residues of target protein, thus transmitting signals from the cell surface to cytoplasmic proteins and the nucleus to regulate physiological processes. Non-receptor tyrosine kinases (NRTK) are a sub-group of tyrosine kinases, which can relay intracellular signals originating from extracellular receptor. NRTKs can regulate a huge array of cellular functions such as cell survival, division/propagation and adhesion, gene expression, immune response, etc. NRTKs exhibit considerable variability in their structural make up, having a shared kinase domain and commonly possessing many other domains such as SH2, SH3 which are protein-protein interacting domains. Recent studies show that NRTKs are mutated in several hematological malignancies, including lymphomas, leukemias and myelomas, leading to aberrant activation. It can be due to point mutations which are intragenic changes or by fusion of genes leading to chromosome translocation. Mutations that lead to constitutive kinase activity result in the formation of oncogenes, such as Abl, Fes, Src, etc. Therefore, specific kinase inhibitors have been sought after to target mutated kinases. A number of compounds have since been discovered, which have shown to inhibit the activity of NRTKs, which are remarkably well tolerated. This review covers the role of various NRTKs in the development of hematological cancers, including their deregulation, genetic alterations, aberrant activation and associated mutations. In addition, it also looks at the recent advances in the development of novel natural compounds that can target NRTKs and perhaps in combination with other forms of therapy can show great promise for the treatment of hematological malignancies.
Collapse
Affiliation(s)
- Kodappully S. Siveen
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| | - Kirti S. Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| | - Iman W. Achkar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| | - Sunitha Shyam
- Medical Research Center, Hamad Medical Corporation, Doha, State of Qatar
| | - Abdul Q. Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| | - Maysaloun Merhi
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, State of Qatar
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, State of Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| |
Collapse
|
36
|
Korbee CJ, Heemskerk MT, Kocev D, van Strijen E, Rabiee O, Franken KLMC, Wilson L, Savage NDL, Džeroski S, Haks MC, Ottenhoff THM. Combined chemical genetics and data-driven bioinformatics approach identifies receptor tyrosine kinase inhibitors as host-directed antimicrobials. Nat Commun 2018; 9:358. [PMID: 29367740 PMCID: PMC5783939 DOI: 10.1038/s41467-017-02777-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/23/2017] [Indexed: 01/01/2023] Open
Abstract
Antibiotic resistance poses rapidly increasing global problems in combatting multidrug-resistant (MDR) infectious diseases like MDR tuberculosis, prompting for novel approaches including host-directed therapies (HDT). Intracellular pathogens like Salmonellae and Mycobacterium tuberculosis (Mtb) exploit host pathways to survive. Only very few HDT compounds targeting host pathways are currently known. In a library of pharmacologically active compounds (LOPAC)-based drug-repurposing screen, we identify multiple compounds, which target receptor tyrosine kinases (RTKs) and inhibit intracellular Mtb and Salmonellae more potently than currently known HDT compounds. By developing a data-driven in silico model based on confirmed targets from public databases, we successfully predict additional efficacious HDT compounds. These compounds target host RTK signaling and inhibit intracellular (MDR) Mtb. A complementary human kinome siRNA screen independently confirms the role of RTK signaling and kinases (BLK, ABL1, and NTRK1) in host control of Mtb. These approaches validate RTK signaling as a drugable host pathway for HDT against intracellular bacteria.
Collapse
Affiliation(s)
- Cornelis J Korbee
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - Matthias T Heemskerk
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - Dragi Kocev
- Department of Knowledge Technologies, Jožef Stefan Institute, Jamova Cesta 39, Ljubljana, 1000, Slovenia
| | - Elisabeth van Strijen
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - Omid Rabiee
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - Kees L M C Franken
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - Louis Wilson
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - Nigel D L Savage
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - Sašo Džeroski
- Department of Knowledge Technologies, Jožef Stefan Institute, Jamova Cesta 39, Ljubljana, 1000, Slovenia
| | - Mariëlle C Haks
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands.
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands.
| |
Collapse
|
37
|
Berman-Booty LD, Eraslan R, Hanumegowda U, Cantor GH, Bounous DI, Janovitz EB, Jones BK, Buiakova O, Hayward M, Wee S. Systemic Loss of C-terminal Src Kinase Expression Elicits Spontaneous Suppurative Inflammation in Conditional Knockout Mice. Vet Pathol 2018; 55:331-340. [DOI: 10.1177/0300985817747330] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
C-terminal Src kinase (Csk) is one of the critical negative regulators of the Src family of kinases. The Src family of kinases are nonreceptor tyrosine kinases that regulate inflammation, cell proliferation, motility, and adhesion. To investigate potential histologic lesions associated with systemic loss of Csk gene activity in adult mice, conditional Csk-knockout mice were examined. Cre-mediated systemic excision of Csk induced by tamoxifen treatment resulted in multiorgan inflammation. Specifically, induction of Csk gene excision with three days of tamoxifen treatment resulted in greater than 90% gene excision. Strikingly, these mice developed enteritis that ranged from minimal and suppurative to severe, fibrinonecrosuppurative and hemorrhagic. Other inflammatory lesions included suppurative pneumonia, gastritis, and myocarditis, and increased numbers of inflammatory cells within the hepatic parenchyma. When tamoxifen treatment was reduced from three days to one day in an effort to lower the level of Csk gene excision and limit lesion development, the mice developed severe suppurative to pyogranulomatous pneumonia and minimal to mild suppurative enteritis. Lesions observed secondary to Csk gene excision suggest important roles for Csk in downregulating the proinflammatory activity of the Src family of kinases and limiting neutrophil-mediated inflammation.
Collapse
Affiliation(s)
| | - Rukiye Eraslan
- Bristol-Myers Squibb, Princeton, NJ, USA
- Invivotek, Hamilton, NJ, USA
| | - Umesh Hanumegowda
- Bristol-Myers Squibb, Princeton, NJ, USA
- ViiV Healthcare, Wallingford, CT, USA
| | | | | | | | | | | | | | - Susan Wee
- Bristol-Myers Squibb, Princeton, NJ, USA
| |
Collapse
|
38
|
Afifiyan N, Tillman B, French BA, Sweeny O, Masouminia M, Samadzadeh S, French SW. The role of Tec kinase signaling pathways in the development of Mallory Denk Bodies in balloon cells in alcoholic hepatitis. Exp Mol Pathol 2017; 103:191-199. [PMID: 28935395 DOI: 10.1016/j.yexmp.2017.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 09/15/2017] [Indexed: 02/08/2023]
Abstract
Several research strategies have been used to study the pathogenesis of alcoholic hepatitis (AH). These strategies have shown that various signaling pathways are the target of alcohol in liver cells. However, few have provided specific mechanisms associated with Mallory-Denk Bodies (MDBs) formed in Balloon cells in AH. The formation of MDBs in these hepatocytes is an indication that the mechanisms of protein quality control have failed. The MDB is the result of aggregation and accumulation of proteins in the cytoplasm of balloon degenerated liver cells. To understand the mechanisms that failed to degrade and remove proteins in the hepatocyte from patients suffering from alcoholic hepatitis, we investigated the pathways that showed significant up regulation in the AH liver biopsies compared to normal control livers (Liu et al., 2015). Analysis of genomic profiles of AH liver biopsies and control livers by RNA-seq revealed different pathways that were up regulated significantly. In this study, the focus was on Tec kinase signaling pathways and the genes that significantly interrupt this pathway. Quantitative PCR and immunofluorescence staining results, indicated that several genes and proteins are significantly over expressed in the livers of AH patients that affect the Tec kinase signaling to PI3K which leads to activation of Akt and its downstream effectors.
Collapse
Affiliation(s)
- N Afifiyan
- Department of Pathology, Harbor UCLA Medical Center, Los Angeles BioMedical Institute, 1000W. Carson, Torrance, CA 90509, United States
| | - B Tillman
- Department of Pathology, Harbor UCLA Medical Center, Los Angeles BioMedical Institute, 1000W. Carson, Torrance, CA 90509, United States
| | - B A French
- Department of Pathology, Harbor UCLA Medical Center, Los Angeles BioMedical Institute, 1000W. Carson, Torrance, CA 90509, United States
| | - O Sweeny
- Department of Pathology, Harbor UCLA Medical Center, Los Angeles BioMedical Institute, 1000W. Carson, Torrance, CA 90509, United States
| | - M Masouminia
- Department of Pathology, Harbor UCLA Medical Center, Los Angeles BioMedical Institute, 1000W. Carson, Torrance, CA 90509, United States
| | - S Samadzadeh
- Department of Pathology, Harbor UCLA Medical Center, Los Angeles BioMedical Institute, 1000W. Carson, Torrance, CA 90509, United States
| | - S W French
- Department of Pathology, Harbor UCLA Medical Center, Los Angeles BioMedical Institute, 1000W. Carson, Torrance, CA 90509, United States.
| |
Collapse
|
39
|
Mkaddem SB, Murua A, Flament H, Titeca-Beauport D, Bounaix C, Danelli L, Launay P, Benhamou M, Blank U, Daugas E, Charles N, Monteiro RC. Lyn and Fyn function as molecular switches that control immunoreceptors to direct homeostasis or inflammation. Nat Commun 2017; 8:246. [PMID: 28811476 PMCID: PMC5557797 DOI: 10.1038/s41467-017-00294-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 06/12/2017] [Indexed: 01/01/2023] Open
Abstract
Immunoreceptors can transduce either inhibitory or activatory signals depending on ligand avidity and phosphorylation status, which is modulated by the protein kinases Lyn and Fyn. Here we show that Lyn and Fyn control immune receptor signaling status. SHP-1 tyrosine 536 phosphorylation by Lyn activates the phosphatase promoting inhibitory signaling through the immunoreceptor. By contrast, Fyn-dependent phosphorylation of SHP-1 serine 591 inactivates the phosphatase, enabling activatory immunoreceptor signaling. These SHP-1 signatures are relevant in vivo, as Lyn deficiency exacerbates nephritis and arthritis in mice, whereas Fyn deficiency is protective. Similarly, Fyn-activating signature is detected in patients with lupus nephritis, underlining the importance of this Lyn-Fyn balance. These data show how receptors discriminate negative from positive signals that respectively result in homeostatic or inflammatory conditions.Src-family kinases Fyn and Lyn are signaling components downstream of ITAM-bearing antigen receptors. Here the authors show that by phosphorylating SHP-1 at different residues, Lyn and Fyn can have opposing regulatory effects on ITAM receptors.
Collapse
Affiliation(s)
- Sanae Ben Mkaddem
- INSERM U1149, Centre de Recherche sur l'Inflammation, Paris, France.
- CNRS ERL8252, Paris, France.
- Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, Paris, France.
- Inflamex Laboratory of Excellence, Paris, France.
| | - Amaya Murua
- INSERM U1149, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Héloise Flament
- INSERM U1149, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
- Service d'Immunologie, DHU Fire, Hôpital Bichat-Claude Bernard, Assistance Publique de Paris, Paris, France
| | - Dimitri Titeca-Beauport
- INSERM U1149, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Carine Bounaix
- INSERM U1149, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Luca Danelli
- INSERM U1149, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Pierre Launay
- INSERM U1149, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Marc Benhamou
- INSERM U1149, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Ulrich Blank
- INSERM U1149, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Eric Daugas
- INSERM U1149, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
- Service de Néphrologie, DHU Fire, Hôpital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Nicolas Charles
- INSERM U1149, Centre de Recherche sur l'Inflammation, Paris, France
- CNRS ERL8252, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Renato C Monteiro
- INSERM U1149, Centre de Recherche sur l'Inflammation, Paris, France.
- CNRS ERL8252, Paris, France.
- Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, Paris, France.
- Inflamex Laboratory of Excellence, Paris, France.
- Service d'Immunologie, DHU Fire, Hôpital Bichat-Claude Bernard, Assistance Publique de Paris, Paris, France.
| |
Collapse
|
40
|
Petersen DL, Berthelsen J, Willerslev-Olsen A, Fredholm S, Dabelsteen S, Bonefeld CM, Geisler C, Woetmann A. A novel BLK-induced tumor model. Tumour Biol 2017; 39:1010428317714196. [PMID: 28670978 DOI: 10.1177/1010428317714196] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
B-lymphoid tyrosine kinase (BLK) is a non-receptor tyrosine kinase belonging to the SRC family kinases. BLK is known to be functionally involved in B-cell receptor signaling and B-cell development. New evidence suggests that B-lymphoid tyrosine kinase is ectopically expressed and is a putative oncogene in cutaneous T-cell lymphoma and other T-cell malignancies. However, little is known about the role of BLK in lymphomagenesis, and the oncogenic function seems to depend on the cellular context. Importantly, BLK is also ectopically expressed in other hematological and multiple non-hematological malignancies including breast, kidney, and lung cancers, suggesting that BLK could be a new potential target for therapy. Here, we studied the oncogenic potential of human BLK. We found that engrafted Ba/F3 cells stably expressing constitutive active human BLK formed tumors in mice, whereas neither Ba/F3 cells expressing wild type BLK nor non-transfected Ba/F3 cells did. Inhibition of BLK with the clinical grade and broadly reacting SRC family kinase inhibitor dasatinib inhibited growth of BLK-induced tumors. In conclusion, our study provides evidence that human BLK is a true proto-oncogene capable of inducing tumors, and we demonstrate a novel BLK activity-dependent tumor model suitable for studies of BLK-driven lymphomagenesis and screening of novel BLK inhibitors in vivo.
Collapse
Affiliation(s)
- David Leander Petersen
- 1 Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jens Berthelsen
- 1 Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | - Simon Fredholm
- 1 Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Sally Dabelsteen
- 2 Department of Odontology, University of Copenhagen, Copenhagen, Denmark
| | | | - Carsten Geisler
- 1 Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Anders Woetmann
- 1 Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
41
|
Amengual JE, Prabhu SA, Lombardo M, Zullo K, Johannet PM, Gonzalez Y, Scotto L, Serrano XJ, Wei Y, Duong J, Nandakumar R, Cremers S, Verma A, Elemento O, O'Connor OA. Mechanisms of Acquired Drug Resistance to the HDAC6 Selective Inhibitor Ricolinostat Reveals Rational Drug-Drug Combination with Ibrutinib. Clin Cancer Res 2017; 23:3084-3096. [PMID: 27993968 PMCID: PMC5474138 DOI: 10.1158/1078-0432.ccr-16-2022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/22/2016] [Accepted: 11/23/2016] [Indexed: 12/31/2022]
Abstract
Purpose: Pan-class I/II histone deacetylase (HDAC) inhibitors are effective treatments for select lymphomas. Isoform-selective HDAC inhibitors are emerging as potentially more targeted agents. ACY-1215 (ricolinostat) is a first-in-class selective HDAC6 inhibitor. To better understand the discrete function of HDAC6 and its role in lymphoma, we developed a lymphoma cell line resistant to ACY-1215.Experimental Design: The diffuse large B-cell lymphoma cell line OCI-Ly10 was exposed to increasing concentrations of ACY-1215 over an extended period of time, leading to the development of a resistant cell line. Gene expression profiling (GEP) was performed to investigate differentially expressed genes. Combination studies of ACY-1215 and ibrutinib were performed in cell lines, primary human lymphoma tissue, and a xenograft mouse model.Results: Systematic incremental increases in drug exposure led to the development of distinct resistant cell lines with IC50 values 10- to 20-fold greater than that for parental lines. GEP revealed upregulation of MAPK10, HELIOS, HDAC9, and FYN, as well as downregulation of SH3BP5 and LCK. Gene-set enrichment analysis (GSEA) revealed modulation of the BTK pathway. Ibrutinib was found to be synergistic with ACY-1215 in cell lines as well as in 3 primary patient samples of lymphoma. In vivo confirmation of antitumor synergy was demonstrated with a xenograft of DLBCL.Conclusions: The development of this ACY-1215-resistant cell line has provided valuable insights into the mechanistic role of HDAC6 in lymphoma and offered a novel method to identify rational synergistic drug combinations. Translation of these findings to the clinic is underway. Clin Cancer Res; 23(12); 3084-96. ©2016 AACR.
Collapse
Affiliation(s)
- Jennifer E Amengual
- Center for Lymphoid Malignancies, Columbia University Medical Center, New York, New York.
| | - Sathyen A Prabhu
- Center for Lymphoid Malignancies, Columbia University Medical Center, New York, New York
| | - Maximilian Lombardo
- Center for Lymphoid Malignancies, Columbia University Medical Center, New York, New York
| | - Kelly Zullo
- Center for Lymphoid Malignancies, Columbia University Medical Center, New York, New York
| | - Paul M Johannet
- Stanford University School of Medicine, Stanford, California
| | - Yulissa Gonzalez
- Center for Lymphoid Malignancies, Columbia University Medical Center, New York, New York
| | - Luigi Scotto
- Center for Lymphoid Malignancies, Columbia University Medical Center, New York, New York
| | - Xavier Jirau Serrano
- Center for Lymphoid Malignancies, Columbia University Medical Center, New York, New York
| | - Ying Wei
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York
| | - Jimmy Duong
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York
| | - Renu Nandakumar
- Division of Clinical Pathology, Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Serge Cremers
- Division of Clinical Pathology, Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Akanksha Verma
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York
| | - Olivier Elemento
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York
| | - Owen A O'Connor
- Center for Lymphoid Malignancies, Columbia University Medical Center, New York, New York
| |
Collapse
|
42
|
Du W, Goldstein R, Jiang Y, Aly O, Cerchietti L, Melnick A, Elemento O. Effective Combination Therapies for B-cell Lymphoma Predicted by a Virtual Disease Model. Cancer Res 2017; 77:1818-1830. [PMID: 28130226 DOI: 10.1158/0008-5472.can-16-0476] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 12/10/2016] [Accepted: 01/23/2017] [Indexed: 12/15/2022]
Abstract
The complexity of cancer signaling networks limits the efficacy of most single-agent treatments and brings about challenges in identifying effective combinatorial therapies. In this study, we used chronic active B-cell receptor (BCR) signaling in diffuse large B-cell lymphoma as a model system to establish a computational framework to optimize combinatorial therapy in silico We constructed a detailed kinetic model of the BCR signaling network, which captured the known complex cross-talk between the NFκB, ERK, and AKT pathways and multiple feedback loops. Combining this signaling model with a data-derived tumor growth model, we predicted viability responses of many single drug and drug combinations in agreement with experimental data. Under this framework, we exhaustively predicted and ranked the efficacy and synergism of all possible combinatorial inhibitions of eleven currently targetable kinases in the BCR signaling network. Ultimately, our work establishes a detailed kinetic model of the core BCR signaling network and provides the means to explore the large space of possible drug combinations. Cancer Res; 77(8); 1818-30. ©2017 AACR.
Collapse
Affiliation(s)
- Wei Du
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - Rebecca Goldstein
- Hematology/Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Yanwen Jiang
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York.,Hematology/Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Omar Aly
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - Leandro Cerchietti
- Hematology/Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York.,Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Ari Melnick
- Hematology/Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York.,Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Olivier Elemento
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York. .,Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York.,Institute for Precision Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
43
|
Skrzypczynska KM, Zhu JW, Weiss A. Positive Regulation of Lyn Kinase by CD148 Is Required for B Cell Receptor Signaling in B1 but Not B2 B Cells. Immunity 2016; 45:1232-1244. [PMID: 27889108 DOI: 10.1016/j.immuni.2016.10.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 08/09/2016] [Accepted: 09/01/2016] [Indexed: 12/31/2022]
Abstract
B1 and B2 B cells differ in their ability to respond to T-cell-independent (TI) antigens. Here we report that the Src-family kinase (SFK) regulator CD148 has a unique and critical role in the initiation of B1 but not B2 cell antigen receptor signaling. CD148 loss-of-function mice were found to have defective B1 B-cell-mediated antibody responses against the T-cell-independent antigens NP-ficoll and Pneumovax 23 and had impaired selection of the B1 B cell receptor (BCR) repertoire. These deficiencies were associated with a decreased ability of B1 B cells to induce BCR signaling downstream of the SFK Lyn. Notably, Lyn appeared to be selectively regulated by CD148 and loss of this SFK resulted in opposite signaling phenotypes in B1 and B2 B cells. These findings reveal that the function and regulation of Lyn during B1 cell BCR signaling is distinct from other B cell subsets.
Collapse
Affiliation(s)
- Katarzyna M Skrzypczynska
- Howard Hughes Medical Institute, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Departments of Medicine and of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143-0795, USA
| | - Jing W Zhu
- Howard Hughes Medical Institute, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Departments of Medicine and of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143-0795, USA
| | - Arthur Weiss
- Howard Hughes Medical Institute, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Departments of Medicine and of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143-0795, USA.
| |
Collapse
|
44
|
McKenna MK, Gachuki BW, Alhakeem SS, Oben KN, Rangnekar VM, Gupta RC, Bondada S. Anti-cancer activity of withaferin A in B-cell lymphoma. Cancer Biol Ther 2016; 16:1088-98. [PMID: 26020511 DOI: 10.1080/15384047.2015.1046651] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Withaferin A (WA), a withanolide from the plant, Ashwagandha (Withania somnifera) used in Ayurvedic medicine, has been found to be valuable in the treatment of several medical ailments. WA has been found to have anticancer activity against various solid tumors, but its effects on hematological malignancies have not been studied in detail. WA strongly inhibited the survival of several human and murine B cell lymphoma cell lines. Additionally, in vivo studies with syngeneic-graft lymphoma cells suggest that WA inhibits the growth of tumor but does not affect other proliferative tissues. We demonstrate that WA inhibits the efficiency of NF-κB nuclear translocation in diffuse large B cell lymphomas and found that WA treatment resulted in a significant decrease in protein levels involved in B cell receptor signaling and cell cycle regulation. WA inhibited the activity of heat shock protein (Hsp) 90 as reflected by a sharp increase in Hsp70 expression levels. Hence, we propose that the anti-cancer effects of WA in lymphomas are likely due to its ability to inhibit Hsp90 function and subsequent reduction of critical kinases and cell cycle regulators that are clients of Hsp90.
Collapse
Affiliation(s)
- M K McKenna
- a Department of Microbiology, Immunology and Molecular Genetics; Markey Cancer Center; University of Kentucky ; Lexington , KY , USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Weir ME, Mann JE, Corwin T, Fulton ZW, Hao JM, Maniscalco JF, Kenney MC, Roman Roque KM, Chapdelaine EF, Stelzl U, Deming PB, Ballif BA, Hinkle KL. Novel autophosphorylation sites of Src family kinases regulate kinase activity and SH2 domain-binding capacity. FEBS Lett 2016; 590:1042-52. [PMID: 27001024 DOI: 10.1002/1873-3468.12144] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 02/18/2016] [Accepted: 03/17/2016] [Indexed: 01/10/2023]
Abstract
Src family tyrosine kinases (SFKs) are critical players in normal and aberrant biological processes. While phosphorylation importantly regulates SFKs at two known tyrosines, large-scale phosphoproteomics have revealed four additional tyrosines commonly phosphorylated in SFKs. We found these novel tyrosines to be autophosphorylation sites. Mimicking phosphorylation at the C-terminal site to the activation loop decreased Fyn activity. Phosphomimetics and direct phosphorylation at the three SH2 domain sites increased Fyn activity while reducing phosphotyrosine-dependent interactions. While 68% of human SH2 domains exhibit conservation of at least one of these tyrosines, few have been found phosphorylated except when found in cis to a kinase domain.
Collapse
Affiliation(s)
- Marion E Weir
- Department of Biology, University of Vermont, Burlington, VT, USA
| | - Jacqueline E Mann
- Department of Medical Laboratory and Radiation Sciences, University of Vermont, Burlington, VT, USA
| | - Thomas Corwin
- Otto-Warburg Laboratory, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Zachary W Fulton
- Department of Biology, University of Vermont, Burlington, VT, USA.,Department of Biology and Physical Education, Norwich University, Northfield, VT, USA
| | - Jennifer M Hao
- Department of Biology, University of Vermont, Burlington, VT, USA
| | | | - Marie C Kenney
- Department of Biology, University of Vermont, Burlington, VT, USA
| | | | - Elizabeth F Chapdelaine
- Department of Biology, University of Vermont, Burlington, VT, USA.,Department of Biology and Physical Education, Norwich University, Northfield, VT, USA
| | - Ulrich Stelzl
- Otto-Warburg Laboratory, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Paula B Deming
- Department of Medical Laboratory and Radiation Sciences, University of Vermont, Burlington, VT, USA
| | - Bryan A Ballif
- Department of Biology, University of Vermont, Burlington, VT, USA
| | - Karen L Hinkle
- Department of Biology, University of Vermont, Burlington, VT, USA.,Department of Biology and Physical Education, Norwich University, Northfield, VT, USA
| |
Collapse
|
46
|
Sakaguchi N, Maeda K. Germinal Center B-Cell-Associated Nuclear Protein (GANP) Involved in RNA Metabolism for B Cell Maturation. Adv Immunol 2016; 131:135-86. [PMID: 27235683 DOI: 10.1016/bs.ai.2016.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Germinal center B-cell-associated nuclear protein (GANP) is upregulated in germinal center B cells against T-cell-dependent antigens in mice and humans. In mice, GANP depletion in B cells impairs antibody affinity maturation. Conversely, its transgenic overexpression augments the generation of high-affinity antigen-specific B cells. GANP associates with AID in the cytoplasm, shepherds AID into the nucleus, and augments its access to the rearranged immunoglobulin (Ig) variable (V) region of the genome in B cells, thereby precipitating the somatic hypermutation of V region genes. GANP is also upregulated in human CD4(+) T cells and is associated with APOBEC3G (A3G). GANP interacts with A3G and escorts it to the virion cores to potentiate its antiretroviral activity by inactivating HIV-1 genomic cDNA. Thus, GANP is characterized as a cofactor associated with AID/APOBEC cytidine deaminase family molecules in generating diversity of the IgV region of the genome and genetic alterations of exogenously introduced viral targets. GANP, encoded by human chromosome 21, as well as its mouse equivalent on chromosome 10, contains a region homologous to Saccharomyces Sac3 that was characterized as a component of the transcription/export 2 (TREX-2) complex and was predicted to be involved in RNA export and metabolism in mammalian cells. The metabolism of RNA during its maturation, from the transcription site at the chromosome within the nucleus to the cytoplasmic translation apparatus, needs to be elaborated with regard to acquired and innate immunity. In this review, we summarize the current knowledge on GANP as a component of TREX-2 in mammalian cells.
Collapse
Affiliation(s)
- N Sakaguchi
- WPI Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan; Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| | - K Maeda
- WPI Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan; Laboratory of Host Defense, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| |
Collapse
|
47
|
Goldstein RL, Yang SN, Taldone T, Chang B, Gerecitano J, Elenitoba-Johnson K, Shaknovich R, Tam W, Leonard JP, Chiosis G, Cerchietti L, Melnick A. Pharmacoproteomics identifies combinatorial therapy targets for diffuse large B cell lymphoma. J Clin Invest 2015; 125:4559-71. [PMID: 26529251 DOI: 10.1172/jci80714] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 09/21/2015] [Indexed: 01/22/2023] Open
Abstract
Rationally designed combinations of targeted therapies for refractory cancers, such as activated B cell-like diffuse large B cell lymphoma (ABC DLBCL), are likely required to achieve potent, durable responses. Here, we used a pharmacoproteomics approach to map the interactome of a tumor-enriched isoform of HSP90 (teHSP90). Specifically, we chemically precipitated teHSP90-client complexes from DLBCL cell lines with the small molecule PU-H71 and found that components of the proximal B cell receptor (BCR) signalosome were enriched within teHSP90 complexes. Functional assays revealed that teHSP90 facilitates BCR signaling dynamics by enabling phosphorylation of key BCR signalosome components, including the kinases SYK and BTK. Consequently, treatment of BCR-dependent ABC DLBCL cells with PU-H71 attenuated BCR signaling, calcium flux, and NF-κB signaling, ultimately leading to growth arrest. Combined exposure of ABC DLBCL cell lines to PU-H71 and ibrutinib, a BCR pathway inhibitor, more potently suppressed BCR signaling than either drug alone. Correspondingly, PU-H71 combined with ibrutinib induced synergistic killing of lymphoma cell lines, primary human lymphoma specimens ex vivo, and lymphoma xenografts in vivo, without notable toxicity. Together, our results demonstrate that a pharmacoproteome-driven rational combination therapy has potential to provide more potent BCR-directed therapy for ABC DLCBL patients.
Collapse
|
48
|
Carneiro BA, Kaplan JB, Giles FJ. Tyrosine kinase inhibitor therapy in chronic myeloid leukemia: update on key adverse events. Expert Rev Hematol 2015; 8:457-79. [DOI: 10.1586/17474086.2015.1041910] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
49
|
Oksvold MP, Duyvestyn JM, Dagger SA, Taylor SJ, Forfang L, Myklebust JH, Smeland EB, Langdon WY. The targeting of human and mouse B lymphocytes by dasatinib. Exp Hematol 2015; 43:352-363.e4. [DOI: 10.1016/j.exphem.2015.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 01/19/2015] [Accepted: 01/21/2015] [Indexed: 10/24/2022]
|
50
|
Bernard S, Danglade D, Gardano L, Laguillier C, Lazarian G, Roger C, Thieblemont C, Marzec J, Gribben J, Cymbalista F, Varin-Blank N, Ledoux D, Baran-Marszak F. Inhibitors of BCR signalling interrupt the survival signal mediated by the micro-environment in mantle cell lymphoma. Int J Cancer 2014; 136:2761-74. [DOI: 10.1002/ijc.29326] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 10/27/2014] [Indexed: 02/01/2023]
Affiliation(s)
- Sophie Bernard
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| | - Damien Danglade
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| | - Laura Gardano
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
| | - Christelle Laguillier
- Service de Biochimie, Hôpital Jean Verdier, Assistance Publique-Hôpitaux de Paris; Bondy France
| | - Gregory Lazarian
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| | - Claudine Roger
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| | - Catherine Thieblemont
- Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris; Paris France
| | - Jacek Marzec
- Barts Cancer Institute, Queen Mary, University of London; London United Kingdom
| | - John Gribben
- Barts Cancer Institute, Queen Mary, University of London; London United Kingdom
| | - Florence Cymbalista
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| | - Nadine Varin-Blank
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
| | - Dominique Ledoux
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
| | - Fanny Baran-Marszak
- U978 Institut National de la Santé et de la Recherche Médicale; Bobigny France
- Labex Inflamex, Université Paris 13; Sorbonne Paris Cité Bobigny France
- Service d'Hématologie Biologique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris; Bobigny France
| |
Collapse
|