1
|
Marques SI, Sá SI, Carmo H, Carvalho F, Silva JP. Pharmaceutical-mediated neuroimmune modulation in psychiatric/psychological adverse events. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111114. [PMID: 39111563 DOI: 10.1016/j.pnpbp.2024.111114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/21/2024] [Accepted: 08/03/2024] [Indexed: 08/13/2024]
Abstract
The therapeutic use of many pharmaceuticals, including small molecules and biological therapies, has been associated with the onset of psychiatric and psychological adverse events (PPAEs), posing substantial concerns to patients' health and safety. These events, which encompass mood (e.g., depression, schizophrenia, suicidal ideation) and cognitive changes (e.g., learning and memory impairment, dementia) often remain undetected until advanced stages of clinical trials or pharmacovigilance, mostly because the mechanisms underlying the onset of PPAEs remain poorly understood. In recent years, the role of neuroimmune modulation (comprising an intricate interplay between various cell types and signaling pathways) in PPAEs has garnered substantial interest. Indeed, understanding these complex interactions would substantially contribute to increase the ability to predict the potential onset of PPAEs during preclinical stages of a new drug's R&D. This review provides a comprehensive summary of the most recent advances in neuroimmune modulation-related mechanisms contributing to the onset of PPAEs and their association with specific pharmaceuticals. Reported data strongly support an association between neuroimmune modulation and the onset of PPAEs. Pharmaceuticals may target specific molecular pathways and pathway elements (e.g., cholinergic and serotonergic systems), which in turn may directly or indirectly impact the inflammatory status and the homeostasis of the brain, regulating inflammation and neuronal function. Also, modulation of the peripheral immune system by pharmaceuticals that do not permeate the blood-brain barrier (e.g., monoclonal antibodies) may alter the neuroimmunomodulatory status of the brain, leading to PPAEs. In summary, this review underscores the diverse pathways through which drugs can influence brain inflammation, shedding light on potential targeted interventions.
Collapse
Affiliation(s)
- Sandra I Marques
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Susana I Sá
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| | - Helena Carmo
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Félix Carvalho
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - João P Silva
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
2
|
González-Martín A, Moyano T, Gutiérrez DA, Carvajal FJ, Cerpa W, Hanley JG, Gutiérrez RA, Álvarez AR. c-Abl regulates a synaptic plasticity-related transcriptional program involved in memory and learning. Prog Neurobiol 2021; 205:102122. [PMID: 34284000 DOI: 10.1016/j.pneurobio.2021.102122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 11/20/2022]
Abstract
Memory consolidation requires activation of a gene expression program that allows de novo protein synthesis. But the molecular mechanisms that favour or restrict that program are poorly understood. The kinase c-Abl can modulate gene expression through transcription factors and chromatin modifiers. Here, we show that c-Abl ablation in the brain improves learning acquisition and memory consolidation in mice. Its absence also affects gene expression profiles in the mouse hippocampus. We found that genes involved in synaptic plasticity and actin cytoskeleton dynamics, such as Arp2 and Thorase, are up-regulated at the mRNA and protein levels in trained c-Abl KO mice and by a chemical-LTP stimulus. Trained c-Abl KO mice also show that dendritic spines are larger than in wild-type mice and present at a higher density. These results indicate that c-Abl kinase is an important part of the mechanism that limits or restricts signalling of relevant gene programs involved in morphological and functional spine changes upon neuronal stimulation.
Collapse
Affiliation(s)
- Adrián González-Martín
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile; Centre for Aging and Regeneration (CARE-UC), Chile
| | - Tomás Moyano
- FONDAP Center for Genome Regulation, Millennium Institute for Integrative Biology (iBio), Department of Molecular Genetics and Microbiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Avda. Libertador Bernardo O'Higgins 340, Santiago, 8331150, Chile
| | - Daniela A Gutiérrez
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile; Centre for Aging and Regeneration (CARE-UC), Chile
| | - Franciso J Carvajal
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile; Center of Excellence in Biomedicine of Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Waldo Cerpa
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile; Centre for Aging and Regeneration (CARE-UC), Chile; Center of Excellence in Biomedicine of Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Jonathan G Hanley
- Centre for Synaptic Plasticity and School of Biochemistry, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Rodrigo A Gutiérrez
- FONDAP Center for Genome Regulation, Millennium Institute for Integrative Biology (iBio), Department of Molecular Genetics and Microbiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Avda. Libertador Bernardo O'Higgins 340, Santiago, 8331150, Chile
| | - Alejandra R Álvarez
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile; Centre for Aging and Regeneration (CARE-UC), Chile.
| |
Collapse
|
3
|
Liu H, Cui Y, Bai Y, Fang Y, Gao T, Wang G, Zhu L, Dong Q, Zhang S, Yao Y, Song C, Niu X, Jin Y, Li P, Cao C, Liu X. The tyrosine kinase c-Abl potentiates interferon-mediated antiviral immunity by STAT1 phosphorylation. iScience 2021; 24:102078. [PMID: 33644712 PMCID: PMC7887405 DOI: 10.1016/j.isci.2021.102078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/28/2020] [Accepted: 01/15/2021] [Indexed: 01/02/2023] Open
Abstract
Interferon (IFN)-induced activation of the signal transducer and activator of transcription (STAT) family is an important event in antiviral immunity. Here, we show that the nonreceptor kinases c-Abl and Arg directly interact with STAT1 and potentiate the phosphorylation of STAT1 on Y701. c-Abl/Arg could mediate STAT1 phosphorylation independent of Janus kinases in the absence of IFNγ and potentiate IFNγ-mediated STAT1 phosphorylation. Moreover, STAT1 dimerization, nuclear translocation, and downstream gene transcription are regulated by c-Abl/Arg. c-Abl/Arg (abl1/abl2) deficiency significantly suppresses antiviral responses in vesicular stomatitis virus-infected cells. Compared to vehicle, administration of the c-Abl/Arg selective inhibitor AMN107 resulted in significantly increased mortality in mice infected with human influenza virus. Our study demonstrates that c-Abl plays an essential role in the STAT1 activation signaling pathway and provides an important approach for antiviral immunity regulation.
Collapse
Affiliation(s)
- Hainan Liu
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Yan Cui
- Beijing Institute of Biotechnology, Beijing 100850, China.,Staidson Bio-pharmaceutics (Beijing) Co. Ltd, Beijing 100176, China
| | - Yu Bai
- Anhui University, Hefei 230601, China
| | - Yi Fang
- The Fifth MedicaI Centre, Chinese PLA GeneraI HospitaI, Beijing 100071, China
| | - Ting Gao
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Guangfei Wang
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Lin Zhu
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Qincai Dong
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Shuwei Zhang
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Yi Yao
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Caiwei Song
- Beijing Institute of Biotechnology, Beijing 100850, China
| | | | - Yanwen Jin
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Ping Li
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Cheng Cao
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Xuan Liu
- Beijing Institute of Biotechnology, Beijing 100850, China
| |
Collapse
|
4
|
Jacobs CF, Eldering E, Kater AP. Kinase inhibitors developed for treatment of hematologic malignancies: implications for immune modulation in COVID-19. Blood Adv 2021; 5:913-925. [PMID: 33560402 PMCID: PMC7871903 DOI: 10.1182/bloodadvances.2020003768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are used to target dysregulated signaling pathways in virtually all hematologic malignancies. Many of the targeted signaling pathways are also essential in nonmalignant immune cells. The current coronavirus severe acute respiratory syndrome coronavirus 2 pandemic catalyzed clinical exploration of TKIs in the treatment of the various stages of COVID-19, which are characterized by distinct immune-related complications. Most of the reported effects of TKIs on immune regulation have been explored in vitro, with different class-specific drugs having nonoverlapping target affinities. Moreover, many of the reported in vivo effects are based on artificial animal models or on observations made in symptomatic patients with a hematologic malignancy who often already suffer from disturbed immune regulation. Based on in vitro and clinical observations, we attempt to decipher the impact of the main TKIs approved or in late-stage development for the treatment of hematological malignancies, including inhibitors of Bruton's tyrosine kinase, spleen tyrosine kinase, BCR-Abl, phosphatidylinositol 3-kinase/ mammalian target of rapamycin, JAK/STAT, and FMS-like tyrosine kinase 3, to provide a rationale for how such inhibitors could modify clinical courses of diseases, such as COVID-19.
Collapse
Affiliation(s)
- Chaja F Jacobs
- Department of Experimental Immunology and
- Department of Hematology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection & Immunity, Amsterdam, The Netherlands; and
| | - Eric Eldering
- Department of Experimental Immunology and
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection & Immunity, Amsterdam, The Netherlands; and
- Lymphoma and Myeloma Center Amsterdam, Amsterdam, The Netherlands
| | - Arnon P Kater
- Department of Hematology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection & Immunity, Amsterdam, The Netherlands; and
- Lymphoma and Myeloma Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Yañez MJ, Marín T, Balboa E, Klein AD, Alvarez AR, Zanlungo S. Finding pathogenic commonalities between Niemann-Pick type C and other lysosomal storage disorders: Opportunities for shared therapeutic interventions. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165875. [PMID: 32522631 DOI: 10.1016/j.bbadis.2020.165875] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/06/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022]
Abstract
Lysosomal storage disorders (LSDs) are diseases characterized by the accumulation of macromolecules in the late endocytic system and are caused by inherited defects in genes that encode mainly lysosomal enzymes or transmembrane lysosomal proteins. Niemann-Pick type C disease (NPCD), a LSD characterized by liver damage and progressive neurodegeneration that leads to early death, is caused by mutations in the genes encoding the NPC1 or NPC2 proteins. Both proteins are involved in the transport of cholesterol from the late endosomal compartment to the rest of the cell. Loss of function of these proteins causes primary cholesterol accumulation, and secondary accumulation of other lipids, such as sphingolipids, in lysosomes. Despite years of studying the genetic and molecular bases of NPCD and related-lysosomal disorders, the pathogenic mechanisms involved in these diseases are not fully understood. In this review we will summarize the pathogenic mechanisms described for NPCD and we will discuss their relevance for other LSDs with neurological components such as Niemann- Pick type A and Gaucher diseases. We will particularly focus on the activation of signaling pathways that may be common to these three pathologies with emphasis on how the intra-lysosomal accumulation of lipids leads to pathology, specifically to neurological impairments. We will show that although the primary lipid storage defect is different in these three LSDs, there is a similar secondary accumulation of metabolites and activation of signaling pathways that can lead to common pathogenic mechanisms. This analysis might help to delineate common pathological mechanisms and therapeutic targets for lysosomal storage diseases.
Collapse
Affiliation(s)
- M J Yañez
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - T Marín
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - E Balboa
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A D Klein
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - A R Alvarez
- Laboratory of Cell Signaling, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago, Chile; CARE UC, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - S Zanlungo
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
6
|
Chen SH, Hsieh YY, Tzeng HE, Lin CY, Hsu KW, Chiang YS, Lin SM, Su MJ, Hsieh WS, Lee CH. ABL Genomic Editing Sufficiently Abolishes Oncogenesis of Human Chronic Myeloid Leukemia Cells In Vitro and In Vivo. Cancers (Basel) 2020; 12:E1399. [PMID: 32485885 PMCID: PMC7352505 DOI: 10.3390/cancers12061399] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic myelogenous leukemia (CML) is the most common type of leukemia in adults, and more than 90% of CML patients harbor the abnormal Philadelphia chromosome (Ph) that encodes the BCR-ABL oncoprotein. Although the ABL kinase inhibitor (imatinib) has proven to be very effective in achieving high remission rates and improving prognosis, up to 33% of CML patients still cannot achieve an optimal response. Here, we used CRISPR/Cas9 to specifically target the BCR-ABL junction region in K562 cells, resulting in the inhibition of cancer cell growth and oncogenesis. Due to the variety of BCR-ABL junctions in CML patients, we utilized gene editing of the human ABL gene for clinical applications. Using the ABL gene-edited virus in K562 cells, we detected 41.2% indels in ABL sgRNA_2-infected cells. The ABL-edited cells reveled significant suppression of BCR-ABL protein expression and downstream signals, inhibiting cell growth and increasing cell apoptosis. Next, we introduced the ABL gene-edited virus into a systemic K562 leukemia xenograft mouse model, and bioluminescence imaging of the mice showed a significant reduction in the leukemia cell population in ABL-targeted mice, compared to the scramble sgRNA virus-injected mice. In CML cells from clinical samples, infection with the ABL gene-edited virus resulted in more than 30.9% indels and significant cancer cell death. Notably, no off-target effects or bone marrow cell suppression was found using the ABL gene-edited virus, ensuring both user safety and treatment efficacy. This study demonstrated the critical role of the ABL gene in maintaining CML cell survival and tumorigenicity in vitro and in vivo. ABL gene editing-based therapy might provide a potential strategy for imatinib-insensitive or resistant CML patients.
Collapse
Affiliation(s)
- Shu-Huey Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Yao-Yu Hsieh
- Division of Hematology and Oncology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Huey-En Tzeng
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Division of Hematology/Oncology, Department of Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Chun-Yu Lin
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu 30068, Taiwan;
- Center for Intelligent Drug Systems and Smart Bio-devices, National Chiao Tung University, Hsinchu 30068, Taiwan
| | - Kai-Wen Hsu
- Institute of New Drug Development, China Medical University, Taichung City 40402, Taiwan;
- Research Center for Cancer Biology, China Medical University, Taichung City 40402, Taiwan
| | - Yun-Shan Chiang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Su-Mei Lin
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 11101, Taiwan;
| | - Ming-Jang Su
- Department of Clinical Pathology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
- Department of Family Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Wen-Shyang Hsieh
- Department of Laboratory Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
| | - Chia-Hwa Lee
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Laboratory Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei 11031, Taiwan
| |
Collapse
|
7
|
ABL Genomic Editing Sufficiently Abolishes Oncogenesis of Human Chronic Myeloid Leukemia Cells In Vitro and In Vivo. Cancers (Basel) 2020. [PMID: 32485885 DOI: 10.3390/cancers12061399.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Chronic myelogenous leukemia (CML) is the most common type of leukemia in adults, and more than 90% of CML patients harbor the abnormal Philadelphia chromosome (Ph) that encodes the BCR-ABL oncoprotein. Although the ABL kinase inhibitor (imatinib) has proven to be very effective in achieving high remission rates and improving prognosis, up to 33% of CML patients still cannot achieve an optimal response. Here, we used CRISPR/Cas9 to specifically target the BCR-ABL junction region in K562 cells, resulting in the inhibition of cancer cell growth and oncogenesis. Due to the variety of BCR-ABL junctions in CML patients, we utilized gene editing of the human ABL gene for clinical applications. Using the ABL gene-edited virus in K562 cells, we detected 41.2% indels in ABL sgRNA_2-infected cells. The ABL-edited cells reveled significant suppression of BCR-ABL protein expression and downstream signals, inhibiting cell growth and increasing cell apoptosis. Next, we introduced the ABL gene-edited virus into a systemic K562 leukemia xenograft mouse model, and bioluminescence imaging of the mice showed a significant reduction in the leukemia cell population in ABL-targeted mice, compared to the scramble sgRNA virus-injected mice. In CML cells from clinical samples, infection with the ABL gene-edited virus resulted in more than 30.9% indels and significant cancer cell death. Notably, no off-target effects or bone marrow cell suppression was found using the ABL gene-edited virus, ensuring both user safety and treatment efficacy. This study demonstrated the critical role of the ABL gene in maintaining CML cell survival and tumorigenicity in vitro and in vivo. ABL gene editing-based therapy might provide a potential strategy for imatinib-insensitive or resistant CML patients.
Collapse
|
8
|
Wetzel DM, Rhodes EL, Li S, McMahon-Pratt D, Koleske AJ. The Src kinases Hck, Fgr and Lyn activate Arg to facilitate IgG-mediated phagocytosis and Leishmania infection. J Cell Sci 2016; 129:3130-43. [PMID: 27358479 DOI: 10.1242/jcs.185595] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 06/23/2016] [Indexed: 12/19/2022] Open
Abstract
Leishmaniasis is a devastating disease that disfigures or kills nearly two million people each year. Establishment and persistence of infection by the obligate intracellular parasite Leishmania requires repeated uptake by macrophages and other phagocytes. Therefore, preventing uptake could be a novel therapeutic strategy for leishmaniasis. Amastigotes, the life cycle stage found in the human host, bind Fc receptors and enter macrophages primarily through immunoglobulin-mediated phagocytosis. However, the host machinery that mediates amastigote uptake is poorly understood. We have previously shown that the Arg (also known as Abl2) non-receptor tyrosine kinase facilitates L. amazonensis amastigote uptake by macrophages. Using small-molecule inhibitors and primary macrophages lacking specific Src family kinases, we now demonstrate that the Hck, Fgr and Lyn kinases are also necessary for amastigote uptake by macrophages. Src-mediated Arg activation is required for efficient uptake. Interestingly, the dual Arg and Src kinase inhibitor bosutinib, which is approved to treat cancer, not only decreases amastigote uptake, but also significantly reduces disease severity and parasite burden in Leishmania-infected mice. Our results suggest that leishmaniasis could potentially be treated with host-cell-active agents such as kinase inhibitors.
Collapse
Affiliation(s)
- Dawn M Wetzel
- Department of Pediatrics, Yale University, New Haven, CT 06520, USA
| | - Emma L Rhodes
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shaoguang Li
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Diane McMahon-Pratt
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, New Haven, CT 06520, USA
| | - Anthony J Koleske
- Department of Molecular Biochemistry and Biophysics, Yale University, CT 06520, USA Department of Neuroscience, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
9
|
Tang DD. Critical role of actin-associated proteins in smooth muscle contraction, cell proliferation, airway hyperresponsiveness and airway remodeling. Respir Res 2015; 16:134. [PMID: 26517982 PMCID: PMC4628321 DOI: 10.1186/s12931-015-0296-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 10/22/2015] [Indexed: 01/16/2023] Open
Abstract
Asthma is characterized by airway hyperresponsiveness and airway remodeling, which are largely attributed to increased airway smooth muscle contractility and cell proliferation. It is known that both chemical and mechanical stimulation regulates smooth muscle contraction. Recent studies suggest that contractile activation and mechanical stretch induce actin cytoskeletal remodeling in smooth muscle. However, the mechanisms that control actin cytoskeletal reorganization are not completely elucidated. This review summarizes our current understanding regarding how actin-associated proteins may regulate remodeling of the actin cytoskeleton in airway smooth muscle. In particular, there is accumulating evidence to suggest that Abelson tyrosine kinase (Abl) plays a critical role in regulating airway smooth muscle contraction and cell proliferation in vitro, and airway hyperresponsiveness and remodeling in vivo. These studies indicate that Abl may be a novel target for the development of new therapy to treat asthma.
Collapse
Affiliation(s)
- Dale D Tang
- Center for Cardiovascular Sciences, Albany Medical College, 47 New Scotland Avenue, MC-8, Albany, NY, 12208, USA.
| |
Collapse
|
10
|
Hrincius ER, Liedmann S, Anhlan D, Wolff T, Ludwig S, Ehrhardt C. Avian influenza viruses inhibit the major cellular signalling integrator c-Abl. Cell Microbiol 2014; 16:1854-74. [PMID: 25052580 DOI: 10.1111/cmi.12332] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 07/11/2014] [Accepted: 07/15/2014] [Indexed: 11/27/2022]
Abstract
The non-structural protein 1 (NS1) of influenza A viruses (IAV) encodes several src homology (SH) binding motifs (bm) (one SH2bm, up to two SH3bm), which mediate interactions with host cell proteins. In contrast to NS1 of human IAV, NS1 of avian strains possess the second SH3bm (SH3(II)bm) consensus sequence. Since our former studies demonstrated an NS1-CRK interaction, mediated by this motif, here, we addressed the regulatory properties of this SH3bm for cellular signalling. Initially, we observed a reduced basal CRK phosphorylation upon infection with avian IAV harbouring an NS1 with an SH3(II)bm in contrast to human IAV. Reduced activity of the tyrosine kinase c-Abl was identified to be responsible for reduced CRK phosphorylation. Further, binding of NS1 to c-Abl was determined, and mutational manipulation of the SH3(II)bm illustrated the necessity of this motif for c-Abl inhibition. Interestingly, Abl kinase inhibition resulted in impaired avian IAV propagation and pathogenicity and mutational analysis linked the pronounced inhibition of c-Abl to cytopathogenic cell alterations upon avian IAV infections. Taken together, NS1 proteins of avian IAV interfere with the kinase activity of c-Abl, a major cellular signalling integrator that controls multiple signalling processes and cell fate regulations apparently including IAV infections.
Collapse
Affiliation(s)
- Eike R Hrincius
- Institute of Molecular Virology (IMV), Center of Molecular Biology of Inflammation (ZMBE), University of Muenster, Von Esmarch-Str. 56, D-48149, Muenster, Germany; Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, 38105-3678, USA
| | | | | | | | | | | |
Collapse
|
11
|
Nguyen TM, Arthur A, Hayball JD, Gronthos S. EphB and Ephrin-B interactions mediate human mesenchymal stem cell suppression of activated T-cells. Stem Cells Dev 2013; 22:2751-64. [PMID: 23711177 PMCID: PMC3787464 DOI: 10.1089/scd.2012.0676] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Accepted: 05/27/2013] [Indexed: 01/13/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSC) express the contact-dependent erythropoietin-producing hepatocellular (Eph) receptor tyrosine kinase family and their cognate ephrin ligands, which are known to regulate thymocyte maturation and selection, T-cell transendothelial migration, activation, co-stimulation, and proliferation. However, the contribution of Eph/ephrin molecules in mediating human MSC suppression of activated T-cells remains to be determined. In the present study, we showed that EphB2 and ephrin-B2 are expressed by ex vivo expanded MSC, while the corresponding ligands, ephrin-B1 and EphB4, respectively, are highly expressed by T-cells. Initial studies demonstrated that EphB2-Fc and ephrin-B2-Fc molecules suppressed T-cell proliferation in allogeneic mixed lymphocyte reaction (MLR) assays compared with human IgG-treated controls. While the addition of a third-party MSC population demonstrated dramatic suppression of T-cell proliferation responses in the MLR, blocking the function of EphB2 or EphB4 receptors using inhibitor binding peptides significantly increased T-cell proliferation. Consistent with these observations, shRNA EphB2 or ephrin-B2 knockdown expression in MSC reduced their ability to inhibit T-cell proliferation. Importantly, the expression of immunosuppressive factors, indoleamine 2, 3-dioxygenase, transforming growth factor-β1, and inducible nitric oxide synthase expressed by MSC, was up-regulated after stimulation with EphB4 and ephrin-B1 in the presence of interferon (IFN)-γ, compared with untreated controls. Conversely, key factors involved in T-cell activation and proliferation, such as interleukin (IL)-2, IFN-γ, tumor necrosis factor-α, and IL-17, were down-regulated by T-cells treated with EphB2 or ephrin-B2 compared with untreated controls. Studies utilizing signaling inhibitors revealed that inhibition of T-cell proliferation is partly mediated through EphB2-induced ephrin-B1 reverse signaling or ephrin-B2-mediated EphB4 forward signaling by activating Src, PI3Kinase, Abl, and JNK kinase pathways, activated by tyrosine phosphorylation. Taken together, these observations suggest that EphB/ephrin-B interactions play an important role in mediating human MSC inhibition of activated T cells.
Collapse
MESH Headings
- Cell Proliferation
- Coculture Techniques
- Ephrin-B2/antagonists & inhibitors
- Ephrin-B2/genetics
- Ephrin-B2/metabolism
- Gene Expression Regulation
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Interferon-gamma/metabolism
- Interferon-gamma/pharmacology
- Interleukin-17/genetics
- Interleukin-17/metabolism
- Interleukin-2/genetics
- Interleukin-2/metabolism
- Lymphocyte Activation
- Lymphocyte Culture Test, Mixed
- Mesenchymal Stem Cells/cytology
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Nitric Oxide Synthase Type II/genetics
- Nitric Oxide Synthase Type II/metabolism
- Phosphorylation
- Primary Cell Culture
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Receptor, EphB2/antagonists & inhibitors
- Receptor, EphB2/genetics
- Receptor, EphB2/metabolism
- Receptor, EphB4/genetics
- Receptor, EphB4/metabolism
- Signal Transduction
- T-Lymphocytes/cytology
- T-Lymphocytes/drug effects
- T-Lymphocytes/metabolism
- Transforming Growth Factor beta1/genetics
- Transforming Growth Factor beta1/metabolism
Collapse
Affiliation(s)
- Thao M. Nguyen
- Mesenchymal Stem Cell Laboratory, School of Medical Sciences, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia
- School of Pharmacy and Medical Sciences and Sansom Institute, University of South Australia, Adelaide, SA, Australia
| | - Agnes Arthur
- Mesenchymal Stem Cell Laboratory, School of Medical Sciences, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia
| | - John D. Hayball
- School of Pharmacy and Medical Sciences and Sansom Institute, University of South Australia, Adelaide, SA, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, School of Medical Sciences, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia
- Centre for Stem Cell Research and Robinson Institute, School of Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
12
|
Cleary RA, Wang R, Wang T, Tang DD. Role of Abl in airway hyperresponsiveness and airway remodeling. Respir Res 2013; 14:105. [PMID: 24112389 PMCID: PMC3852349 DOI: 10.1186/1465-9921-14-105] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 10/09/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Asthma is a chronic disease that is characterized by airway hyperresponsiveness and airway remodeling. The underlying mechanisms that mediate the pathological processes are not fully understood. Abl is a non-receptor protein tyrosine kinase that has a role in the regulation of smooth muscle contraction and smooth muscle cell proliferation in vitro. The role of Abl in airway hyperresponsiveness and airway remodeling in vivo is largely unknown. METHODS To evaluate the role of Abl in asthma pathology, we assessed the expression of Abl in airway tissues from the ovalbumin sensitized and challenged mouse model, and human asthmatic airway smooth muscle cells. In addition, we generated conditional knockout mice in which Abl expression in smooth muscle was disrupted, and then evaluated the effects of Abl conditional knockout on airway resistance, smooth muscle mass, cell proliferation, IL-13 and CCL2 in the mouse model of asthma. Furthermore, we determined the effects of the Abl pharmacological inhibitors imatinib and GNF-5 on these processes in the animal model of asthma. RESULTS The expression of Abl was upregulated in airway tissues of the animal model of asthma and in airway smooth muscle cells of patients with severe asthma. Conditional knockout of Abl attenuated airway resistance, smooth muscle mass and staining of proliferating cell nuclear antigen in the airway of mice sensitized and challenged with ovalbumin. Interestingly, conditional knockout of Abl did not affect the levels of IL-13 and CCL2 in bronchoalveolar lavage fluid of animals treated with ovalbumin. However, treatment with imatinib and GNF-5 inhibited the ovalbumin-induced increase in IL-13 and CCL2 as well as airway resistance and smooth muscle growth in animals. CONCLUSIONS These results suggest that the altered expression of Abl in airway smooth muscle may play a critical role in the development of airway hyperresponsiveness and airway remodeling in asthma. Our findings support the concept that Abl may be a novel target for the development of new therapy to treat asthma.
Collapse
Affiliation(s)
- Rachel A Cleary
- Center for Cardiovascular Sciences, Albany Medical College, 47 New Scotland Avenue MC-8, Albany, NY 12208, USA.
| | | | | | | |
Collapse
|
13
|
Mahurkar S, Moldovan M, Suppiah V, O’Doherty C. Identification of shared genes and pathways: a comparative study of multiple sclerosis susceptibility, severity and response to interferon beta treatment. PLoS One 2013; 8:e57655. [PMID: 23469041 PMCID: PMC3585216 DOI: 10.1371/journal.pone.0057655] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 01/24/2013] [Indexed: 12/30/2022] Open
Abstract
Recent genome-wide association studies (GWAS) have successfully identified several gene loci associated with multiple sclerosis (MS) susceptibility, severity or interferon-beta (IFN-ß) response. However, due to the nature of these studies, the functional relevance of these loci is not yet fully understood. We have utilized a systems biology based approach to explore the genetic interactomes of these MS related traits. We hypothesised that genes and pathways associated with the 3 MS related phenotypes might interact collectively to influence the heterogeneity and unpredictable clinical outcomes observed. Individual genetic interactomes for each trait were constructed and compared, followed by prioritization of common interactors based on their frequencies. Pathway enrichment analyses were performed to highlight shared functional pathways. Biologically relevant genes ABL1, GRB2, INPP5D, KIF1B, PIK3R1, PLCG1, PRKCD, SRC, TUBA1A and TUBA4A were identified as common to all 3 MS phenotypes. We observed that the highest number of first degree interactors were shared between MS susceptibility and MS severity (p = 1.34×10−79) with UBC as the most prominent first degree interactor for this phenotype pair from the prioritisation analysis. As expected, pairwise comparisons showed that MS susceptibility and severity interactomes shared the highest number of pathways. Pathways from signalling molecules and interaction, and signal transduction categories were found to be highest shared pathways between 3 phenotypes. Finally, FYN was the most common first degree interactor in the MS drugs-gene network. By applying the systems biology based approach, additional significant information can be extracted from GWAS. Results of our interactome analyses are complementary to what is already known in the literature and also highlight some novel interactions which await further experimental validation. Overall, this study illustrates the potential of using a systems biology based approach in an attempt to unravel the biological significance of gene loci identified in large GWAS.
Collapse
Affiliation(s)
- Sunil Mahurkar
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Max Moldovan
- Australian Institute of Health Innovation, University of New South Wales, Sydney, Australia
| | - Vijayaprakash Suppiah
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
- * E-mail:
| | - Catherine O’Doherty
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
14
|
Claudin-1 induces epithelial-mesenchymal transition through activation of the c-Abl-ERK signaling pathway in human liver cells. Oncogene 2012; 32:4873-82. [PMID: 23160379 DOI: 10.1038/onc.2012.505] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 09/13/2012] [Accepted: 09/21/2012] [Indexed: 12/13/2022]
Abstract
Claudins (CLDNs) are a family of integral membrane proteins central to the formation of tight junctions, structures that are involved in paracellular transport and cellular growth and differentiation, and are critical for the maintenance of cellular polarity. Recent studies have provided evidence that CLDNs are aberrantly expressed in diverse types of human cancers, including hepatocellular carcinomas (HCCs). However, little is known about how CLDN expression is involved in cancer progression. In this study, we show that CLDN1 has a causal role in the epithelial-mesenchymal transition (EMT) in human liver cells, and that the c-Abl-Ras-Raf-1-ERK1/2 signaling axis is critical for the induction of malignant progression by CLDN1. Overexpression of CLDN1 induced expression of the EMT-regulating transcription factors Slug and Zeb1, and thereby led to repression of E-cadherin, β-catenin expression, enhanced expression of N-cadherin and Vimentin, a loss of cell adhesion, and increased cell motility in normal liver cells and HCC cells. In line with these findings, inhibition of either c-Abl or ERK clearly attenuated CLDN1-induced EMT, as evidenced by a reversal of N-cadherin and E-cadherin expression patterns, and restored normal motility. Collectively, these results indicate that CLDN1 is necessary for the induction of EMT in human liver cells, and that activation of the c-Abl-Ras-Raf-1-ERK1/2 signaling pathway is required for CLDN1-induced acquisition of the malignant phenotype. The present observations suggest that CLDN1 could be exploited as a biomarker for liver cancer metastasis and might provide a pivotal point for therapeutic intervention in HCC.
Collapse
|
15
|
Greuber EK, Pendergast AM. Abl family kinases regulate FcγR-mediated phagocytosis in murine macrophages. THE JOURNAL OF IMMUNOLOGY 2012; 189:5382-92. [PMID: 23100514 DOI: 10.4049/jimmunol.1200974] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Phagocytosis of Ab-coated pathogens is mediated through FcγRs, which activate intracellular signaling pathways to drive actin cytoskeletal rearrangements. Abl and Arg define a family of nonreceptor tyrosine kinases that regulate actin-dependent processes in a variety of cell types, including those important in the adaptive immune response. Using pharmacological inhibition as well as dominant negative and knockout approaches, we demonstrate a role for the Abl family kinases in phagocytosis by macrophages and define a mechanism whereby Abl kinases regulate this process. Bone marrow-derived macrophages from mice lacking Abl and Arg kinases exhibit inefficient phagocytosis of sheep erythrocytes and zymosan particles. Treatment with the Abl kinase inhibitors imatinib and GNF-2 or overexpression of kinase-inactive forms of the Abl family kinases also impairs particle internalization in murine macrophages, indicating Abl kinase activity is required for efficient phagocytosis. Further, Arg kinase is present at the phagocytic cup, and Abl family kinases are activated by FcγR engagement. The regulation of phagocytosis by Abl family kinases is mediated in part by the spleen tyrosine kinase (Syk). Loss of Abl and Arg expression or treatment with Abl inhibitors reduced Syk phosphorylation in response to FcγR ligation. The link between Abl family kinases and Syk may be direct, as purified Arg kinase phosphorylates Syk in vitro. Further, overexpression of membrane-targeted Syk in cells treated with Abl kinase inhibitors partially rescues the impairment in phagocytosis. Together, these findings reveal that Abl family kinases control the efficiency of phagocytosis in part through the regulation of Syk function.
Collapse
Affiliation(s)
- Emileigh K Greuber
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | | |
Collapse
|
16
|
c-Abl promotes osteoblast expansion by differentially regulating canonical and non-canonical BMP pathways and p16INK4a expression. Nat Cell Biol 2012; 14:727-37. [PMID: 22729085 DOI: 10.1038/ncb2528] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 05/22/2012] [Indexed: 11/08/2022]
Abstract
Defects in stem cell renewal or progenitor cell expansion underlie ageing-related diseases such as osteoporosis. Yet much remains unclear about the mechanisms regulating progenitor expansion. Here we show that the tyrosine kinase c-Abl plays an important role in osteoprogenitor expansion. c-Abl interacts with and phosphorylates BMPRIA and the phosphorylation differentially influences the interaction of BMPRIA with BMPRII and the Tab1-Tak1 complex, leading to uneven activation of Smad1/5/8 and Erk1/2, the canonical and non-canonical BMP pathways that direct the expression of p16(INK4a). c-Abl deficiency shunts BMP signalling from Smad1/5/8 to Erk1/2, leading to p16(INK4a) upregulation and osteoblast senescence. Mouse genetic studies revealed that p16(INK4a) controls mesenchymal stem cell maintenance and osteoblast expansion and mediates the effects of c-Abl deficiency on osteoblast expansion and bone formation. These findings identify c-Abl as a regulator of BMP signalling pathways and uncover a role for c-Abl in p16(INK4a) expression and osteoprogenitor expansion.
Collapse
|
17
|
The Abl and Arg kinases mediate distinct modes of phagocytosis and are required for maximal Leishmania infection. Mol Cell Biol 2012; 32:3176-86. [PMID: 22665498 DOI: 10.1128/mcb.00086-12] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Leishmania, an obligate intracellular parasite, binds several receptors to trigger engulfment by phagocytes, leading to cutaneous or visceral disease. These receptors include complement receptor 3 (CR3), used by promastigotes, and the Fc receptor (FcR), used by amastigotes. The mechanisms mediating uptake are not well understood. Here we show that Abl family kinases mediate both phagocytosis and the uptake of Leishmania amazonensis by macrophages (Ms). Imatinib, an Abl/Arg kinase inhibitor, decreases opsonized polystyrene bead phagocytosis and Leishmania uptake. Interestingly, phagocytosis of IgG-coated beads is decreased in Arg-deficient Ms, while that of C3bi-coated beads is unaffected. Conversely, uptake of C3bi-coated beads is decreased in Abl-deficient Ms, but that of IgG-coated beads is unaffected. Consistent with these results, Abl-deficient Ms are inefficient at C3bi-opsonized promastigote uptake, and Arg-deficient Ms are defective in IgG1-opsonized amastigote uptake. Finally, genetic loss of Abl or Arg reduces infection severity in murine cutaneous leishmaniasis, and imatinib treatment results in smaller lesions with fewer parasites than in controls. Our studies are the first to demonstrate that efficient phagocytosis and maximal Leishmania infection require Abl family kinases. These results highlight Abl family kinase-mediated signaling pathways as potential therapeutic targets for leishmaniasis.
Collapse
|
18
|
Functional mechanisms and roles of adaptor proteins in abl-regulated cytoskeletal actin dynamics. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:414913. [PMID: 22675626 PMCID: PMC3362954 DOI: 10.1155/2012/414913] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 03/16/2012] [Indexed: 01/20/2023]
Abstract
Abl is a nonreceptor tyrosine kinase and plays an essential role in the modeling and remodeling of F-actin by transducing extracellular signals. Abl and its paralog, Arg, are unique among the tyrosine kinase family in that they contain an unusual extended C-terminal half consisting of multiple functional domains. This structural characteristic may underlie the role of Abl as a mediator of upstream signals to downstream signaling machineries involved in actin dynamics. Indeed, a group of SH3-containing accessory proteins, or adaptor proteins, have been identified that bind to a proline-rich domain of the C-terminal portion of Abl and modulate its kinase activity, substrate recognition, and intracellular localization. Moreover, the existence of signaling cascade and biological outcomes unique to each adaptor protein has been demonstrated. In this paper, we summarize functional roles and mechanisms of adaptor proteins in Abl-regulated actin dynamics, mainly focusing on a family of adaptor proteins, Abi. The mechanism of Abl's activation and downstream signaling mediated by Abi is described in comparison with those by another adaptor protein, Crk.
Collapse
|
19
|
Schlatterer SD, Acker CM, Davies P. c-Abl in neurodegenerative disease. J Mol Neurosci 2011; 45:445-52. [PMID: 21728062 DOI: 10.1007/s12031-011-9588-1] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 06/21/2011] [Indexed: 12/30/2022]
Abstract
The c-Abl tyrosine kinase participates in a variety of cellular functions, including regulation of the actin cytoskeleton, regulation of the cell cycle, and the apoptotic/cell cycle arrest response to stress, and the Abl family of kinases has been shown to play a crucial role in development of the central nervous system. Recent studies have shown c-Abl activation in human Alzheimer's and Parkinson's diseases and c-Abl activation in mouse models and neuronal culture in response to amyloid beta fibrils and oxidative stress. Overexpression of active c-Abl in adult mouse neurons results in neurodegeneration and neuroinflammation. Based on this evidence, a potential role for c-Abl in the pathogenesis of neurodegenerative disease is discussed, and we attempt to place activation of c-Abl in context with other known contributors to neurodegenerative pathology.
Collapse
Affiliation(s)
- Sarah D Schlatterer
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | |
Collapse
|
20
|
Morris CM. Chronic myeloid leukemia: cytogenetic methods and applications for diagnosis and treatment. Methods Mol Biol 2011; 730:33-61. [PMID: 21431633 DOI: 10.1007/978-1-61779-074-4_4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Chronic myeloid leukemia (CML) is a clonal myeloproliferative disease caused by recombination between the BCR gene on chromosome 22 and the ABL1 gene on chromosome 9. This rearrangement generates the BCR-ABL1 fusion gene that characterizes leukemic cells in all CML cases. In about 90% of cases, the BCR-ABL1 rearrangement is manifest cytogenetically by the Philadelphia (Ph) chromosome, a derivative of the reciprocal translocation t(9;22)(q34;q11.2). For the remaining cases, recombination may be more complex, involving BCR, ABL1, and genomic sites on one or more other chromosomal regions, or it may occur cryptically within an apparently normal karyotype. Detection of the Ph and associated t(9;22) translocation is a recognized clinical hallmark for CML diagnosis. The disease has a natural multistep pathogenesis, and during chronic phase CML, the t(9;22) or complex variant is usually the sole abnormality. In 60-80% of cases, additional cytogenetic changes appear and often forecast progression to an accelerated disease phase or a terminal blast crisis. Because new frontline therapies such as imatinib specifically target the abnormal protein product of the BCR-ABL1 fusion gene to eliminate BCR-ABL1 positive cells, there is a new reliance on the cytogenetic evaluation of bone marrow cells at diagnosis, then at regular posttreatment intervals. Combined with other parameters, presence or absence of Ph-positive cells in the bone marrow is a powerful early indicator for clinical risk stratification. Cytogenetic changes detected at any stage during treatment, including in the BCR-ABL1-negative cells, may also provide useful prognostic information. Laboratory methods detailed here extend from initial collection of peripheral blood or bone marrow samples through cell culture with or without synchronization, metaphase or interphase harvest, hypotonic treatment and fixation, slide preparation for G-banding or fluorescent in situ hybridization (FISH), and final interpretation.
Collapse
Affiliation(s)
- Christine M Morris
- Cancer Genetics Research Group, Department of Pathology, University of Otago Christchurch School of Medicine and Health Services, Christchurch, New Zealand.
| |
Collapse
|
21
|
Abstract
ABL-family proteins comprise one of the best conserved branches of the tyrosine kinases. Each ABL protein contains an SH3-SH2-TK (Src homology 3-Src homology 2-tyrosine kinase) domain cassette, which confers autoregulated kinase activity and is common among nonreceptor tyrosine kinases. This cassette is coupled to an actin-binding and -bundling domain, which makes ABL proteins capable of connecting phosphoregulation with actin-filament reorganization. Two vertebrate paralogs, ABL1 and ABL2, have evolved to perform specialized functions. ABL1 includes nuclear localization signals and a DNA binding domain through which it mediates DNA damage-repair functions, whereas ABL2 has additional binding capacity for actin and for microtubules to enhance its cytoskeletal remodeling functions. Several types of posttranslational modifications control ABL catalytic activity, subcellular localization, and stability, with consequences for both cytoplasmic and nuclear ABL functions. Binding partners provide additional regulation of ABL catalytic activity, substrate specificity, and downstream signaling. Information on ABL regulatory mechanisms is being mined to provide new therapeutic strategies against hematopoietic malignancies caused by BCR-ABL1 and related leukemogenic proteins.
Collapse
Affiliation(s)
- John Colicelli
- Department of Biological Chemistry, Molecular Biology Institute and Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
22
|
Bradley WD, Koleske AJ. Regulation of cell migration and morphogenesis by Abl-family kinases: emerging mechanisms and physiological contexts. J Cell Sci 2009; 122:3441-54. [PMID: 19759284 DOI: 10.1242/jcs.039859] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The Abl-family non-receptor tyrosine kinases are essential regulators of the cytoskeleton. They transduce diverse extracellular cues into cytoskeletal rearrangements that have dramatic effects on cell motility and morphogenesis. Recent biochemical and genetic studies have revealed several mechanisms that Abl-family kinases use to mediate these effects. Abl-family kinases stimulate actin polymerization through the activation of cortactin, hematopoietic lineage cell-specific protein (HS1), WASp- and WAVE-family proteins, and Rac1. They also attenuate cell contractility by inhibiting RhoA and altering adhesion dynamics. These pathways impinge on several physiological processes, including development and maintenance of the nervous and immune systems, and epithelial morphogenesis. Elucidating how Abl-family kinases are regulated, and where and when they coordinate cytoskeletal changes, is essential for garnering a better understanding of these complex processes.
Collapse
Affiliation(s)
- William D Bradley
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA.
| | | |
Collapse
|