1
|
Zhuang R, Siebels B, Hoffer K, Worthmann A, Horn S, von Bubnoff NCC, Khandanpour C, Gebauer N, Gorantla SP, Voss H, Schlüter H, Kriegs M, Fiedler W, Bokemeyer C, Jücker M, Kebenko M. Functional Role of Fatty Acid Synthase for Signal Transduction in Core-Binding Factor Acute Myeloid Leukemia with an Activating c-Kit Mutation. Biomedicines 2025; 13:619. [PMID: 40149597 PMCID: PMC11939861 DOI: 10.3390/biomedicines13030619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Acute myeloid leukemia (AML) is a rare hematological malignancy with a poor prognosis. Activating c-Kit (CD117) mutations occur in 5% of de novo AML and 30% of core-binding factor (CBF) AML, leading to worse clinical outcomes. Posttranslational modifications, particularly with myristic and palmitic acid, are crucial for various cellular processes, including membrane organization, signal transduction, and apoptosis regulation. However, most research has focused on solid tumors, with limited understanding of these mechanisms in AML. Fatty acid synthase (FASN), a key palmitoyl-acyltransferase, regulates the subcellular localization, trafficking, and degradation of target proteins, such as H-Ras, N-Ras, and FLT3-ITDmut receptors in AML. Methods: In this study, we investigated the role of FASN in two c-Kit-N822K-mutated AML cell lines using FASN knockdown via shRNA and the FASN inhibitor TVB-3166. Functional implications, including cell proliferation, were assessed through Western blotting, mass spectrometry, and PamGene. Results: FASN inhibition led to an increased phosphorylation of c-Kit (p-c-Kit), Lyn kinase (pLyn), MAP kinase (pMAPK), and S6 kinase (pS6). Furthermore, we observed sustained high expression of Gli1 in Kasumi1 cells following FASN inhibition, which is well known to be mediated by the upregulation of pS6. Conclusions: The combination of TVB-3166 and the Gli inhibitor GANT61 resulted in a significant reduction in the survival of Kasumi1 cells.
Collapse
Affiliation(s)
- Ruimeng Zhuang
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (R.Z.); (M.J.)
| | - Bente Siebels
- Section Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (B.S.); (H.S.)
| | - Konstantin Hoffer
- Department of Radiobiology & Radiation Oncology, UCCH Kinomics Core Facility, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (K.H.); (M.K.)
| | - Anna Worthmann
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Stefan Horn
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | | | - Cyrus Khandanpour
- Clinic for Hematology and Oncology, University Hospital Schleswig-Holstein Campus, 23562 Lübeck, Germany; (N.C.C.v.B.); (C.K.); (N.G.); (S.P.G.)
| | - Niklas Gebauer
- Clinic for Hematology and Oncology, University Hospital Schleswig-Holstein Campus, 23562 Lübeck, Germany; (N.C.C.v.B.); (C.K.); (N.G.); (S.P.G.)
| | - Sivahari Prasad Gorantla
- Clinic for Hematology and Oncology, University Hospital Schleswig-Holstein Campus, 23562 Lübeck, Germany; (N.C.C.v.B.); (C.K.); (N.G.); (S.P.G.)
| | - Hanna Voss
- Section Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (B.S.); (H.S.)
| | - Hartmut Schlüter
- Section Mass Spectrometric Proteomics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (B.S.); (H.S.)
| | - Malte Kriegs
- Department of Radiobiology & Radiation Oncology, UCCH Kinomics Core Facility, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (K.H.); (M.K.)
| | - Walter Fiedler
- Hubertus Wald Tumorzentrum, Department of Oncology–Hematology, Bone Marrow Transplantation and Pneumology, University Cancer Center, 20251 Hamburg, Germany; (W.F.); (C.B.)
| | - Carsten Bokemeyer
- Hubertus Wald Tumorzentrum, Department of Oncology–Hematology, Bone Marrow Transplantation and Pneumology, University Cancer Center, 20251 Hamburg, Germany; (W.F.); (C.B.)
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (R.Z.); (M.J.)
| | - Maxim Kebenko
- Clinic for Hematology and Oncology, University Hospital Schleswig-Holstein Campus, 23562 Lübeck, Germany; (N.C.C.v.B.); (C.K.); (N.G.); (S.P.G.)
- Hubertus Wald Tumorzentrum, Department of Oncology–Hematology, Bone Marrow Transplantation and Pneumology, University Cancer Center, 20251 Hamburg, Germany; (W.F.); (C.B.)
| |
Collapse
|
2
|
Sá da Bandeira D, Nevitt CD, Segato Dezem F, Marção M, Liu Y, Kelley Z, DuBose H, Chabot A, Hall T, Caprio C, Okhomina V, Kang G, Plummer J, McKinney-Freeman S, Clements WK, Ganuza M. NR4A1 and NR4A2 orphan nuclear receptors regulate endothelial-to-hematopoietic transition in mouse hematopoietic stem cell specification. Development 2024; 151:dev201957. [PMID: 39589268 PMCID: PMC11634030 DOI: 10.1242/dev.201957] [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: 05/10/2023] [Accepted: 10/14/2024] [Indexed: 11/27/2024]
Abstract
Hematopoietic stem cells (HSCs) sustain life-long hematopoiesis and emerge during mid-gestation from hemogenic endothelial progenitors via an endothelial-to-hematopoietic transition (EHT). The full scope of molecular mechanisms governing this process remains unclear. The NR4A subfamily of orphan nuclear receptors act as tumor suppressors in myeloid leukemogenesis and have never been implicated in HSC specification. Here, we report that Nr4a1 and Nr4a2 expression is upregulated in hemogenic endothelium during EHT. Progressive genetic ablation of Nr4a gene dosage results in a gradual decrease in numbers of nascent c-Kit+ hematopoietic progenitors in developing embryos, c-Kit+ cell cluster size in the dorsal aorta, and a block in HSC maturation, revealed by an accumulation of pro-HSCs and pre-HSC-type I cells and decreased numbers of pre-HSC-type II cells. Consistent with these observations, cells isolated from embryonic day 11.5 Nr4a1-/-; Nr4a2-/- aorta-gonads-mesonephros are devoid of in vivo long-term hematopoietic repopulating potential. Molecularly, employing spatial transcriptomic analysis we determined that the genetic ablation of Nr4a1 and Nr4a2 prevents Notch signaling from being downregulated in intra-aortic clusters and thus for pro-HSCs to mature into HSCs. Interestingly, this defect is partially rescued by ex vivo culture of dissected aorta-gonads-mesonephros with SCF, IL3 and FLT3L, which may bypass Notch-dependent regulation. Overall, our data reveal a role for the NR4A family of orphan nuclear receptors in EHT.
Collapse
MESH Headings
- Animals
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/cytology
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Mice
- Hematopoiesis/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Cell Differentiation/genetics
- Gene Expression Regulation, Developmental
- Aorta/embryology
- Aorta/metabolism
- Gonads/metabolism
- Gonads/embryology
- Mice, Knockout
- Endothelial Cells/metabolism
- Mice, Inbred C57BL
- Mesonephros/embryology
- Mesonephros/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Diana Sá da Bandeira
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Chris D. Nevitt
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Felipe Segato Dezem
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Maycon Marção
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yutian Liu
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zakiya Kelley
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Hannah DuBose
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ashley Chabot
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Trent Hall
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Claire Caprio
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Victoria Okhomina
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Guolian Kang
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jasmine Plummer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | - Wilson K. Clements
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Miguel Ganuza
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
3
|
Stevenson M, Hebron ML, Liu X, Balaraman K, Wolf C, Moussa C. c-KIT inhibitors reduce pathology and improve behavior in the Tg(SwDI) model of Alzheimer's disease. Life Sci Alliance 2024; 7:e202402625. [PMID: 39009412 PMCID: PMC11249953 DOI: 10.26508/lsa.202402625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
Treatments for Alzheimer's disease have primarily focused on removing brain amyloid plaques to improve cognitive outcomes in patients. We developed small compounds, known as BK40143 and BK40197, and we hypothesize that these drugs alleviate microglial-mediated neuroinflammation and induce autophagic clearance of neurotoxic proteins to improve behavior in models of neurodegeneration. Specificity binding assays of BK40143 and BK40197 showed primary binding to c-KIT/Platelet Derived Growth Factor Receptors (PDGFR)α/β, whereas BK40197 also differentially binds to FYVE finger-containing phosphoinositide kinase (PIKFYVE). Both compounds penetrate the CNS, and treatment with these drugs inhibited the maturation of peripheral mast cells in transgenic mice, correlating with cognitive improvements on measures of memory and anxiety. In the brain, microglial activation was profoundly attenuated and amyloid-beta and tau were reduced via autophagy. Multi-kinase inhibition, including c-KIT, exerts multifunctional effects to reduce neurodegenerative pathology via autophagy and microglial activity and may represent a potential therapeutic option for neurodegeneration.
Collapse
Affiliation(s)
- Max Stevenson
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| | - Michaeline L Hebron
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| | - Xiaoguang Liu
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| | - Kaluvu Balaraman
- Medicinal Chemistry Shared Resource, Department of Chemistry, Georgetown University Medical Center, Washington DC, USA
| | - Christian Wolf
- Medicinal Chemistry Shared Resource, Department of Chemistry, Georgetown University Medical Center, Washington DC, USA
| | - Charbel Moussa
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| |
Collapse
|
4
|
Giorgioni L, Ambrosone A, Cometa MF, Salvati AL, Nisticò R, Magrelli A. Revolutionizing CAR T-Cell Therapies: Innovations in Genetic Engineering and Manufacturing to Enhance Efficacy and Accessibility. Int J Mol Sci 2024; 25:10365. [PMID: 39408696 PMCID: PMC11476879 DOI: 10.3390/ijms251910365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has achieved notable success in treating hematological cancers but faces significant challenges in solid-tumor treatment and overall efficacy. Key limitations include T-cell exhaustion, tumor relapse, immunosuppressive tumor microenvironments (TME), immunogenicity, and antigen heterogeneity. To address these issues, various genetic engineering strategies have been proposed. Approaches such as overexpression of transcription factors or metabolic armoring and dynamic CAR regulation are being explored to improve CAR T-cell function and safety. Other efforts to improve CAR T-cell efficacy in solid tumors include targeting novel antigens or developing alternative strategies to address antigen diversity. Despite the promising preclinical results of these solutions, challenges remain in translating CAR T-cell therapies to the clinic to enable economically viable access to these transformative medicines. The efficiency and scalability of autologous CAR T-cell therapy production are hindered by traditional, manual processes which are costly, time-consuming, and prone to variability and contamination. These high-cost, time-intensive processes have complex quality-control requirements. Recent advancements suggest that smaller, decentralized solutions such as microbioreactors and automated point-of-care systems could improve production efficiency, reduce costs, and shorten manufacturing timelines, especially when coupled with innovative manufacturing methods such as transposons and lipid nanoparticles. Future advancements may include harmonized consumables and AI-enabled technologies, which promise to streamline manufacturing, reduce costs, and enhance production quality.
Collapse
Affiliation(s)
- Lorenzo Giorgioni
- Faculty of Physiology and Pharmacology “V. Erspamer”, Sapienza Università di Roma, 00185 Rome, Italy;
| | - Alessandra Ambrosone
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.A.); (M.F.C.)
| | - Maria Francesca Cometa
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.A.); (M.F.C.)
| | - Anna Laura Salvati
- Faculty of Pharmacy, Tor Vergata University of Rome, 00133 Rome, Italy (R.N.)
| | - Robert Nisticò
- Faculty of Pharmacy, Tor Vergata University of Rome, 00133 Rome, Italy (R.N.)
- Agenzia Italiana del Farmaco, Via del Tritone 181, 00187 Rome, Italy
| | - Armando Magrelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.A.); (M.F.C.)
| |
Collapse
|
5
|
Hailey DR, Kanjilal D, Koulen P. Differential Expression of Mitogen-Activated Protein Kinase Signaling Pathways in the Human Choroid-Retinal Pigment Epithelial Complex Indicates Regional Predisposition to Disease. Int J Mol Sci 2024; 25:10105. [PMID: 39337590 PMCID: PMC11432750 DOI: 10.3390/ijms251810105] [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: 08/17/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
The retina is composed of neuronal layers that include several types of interneurons and photoreceptor cells, and separate underlying retinal pigment epithelium (RPE), Bruch's membrane, and choroid. Different regions of the human retina include the fovea, macula, and periphery, which have unique physiological functions and anatomical features. These regions are also unique in their protein expression, and corresponding cellular and molecular responses to physiological and pathophysiological stimuli. Skeie and Mahajan analyzed regional protein expression in the human choroid-RPE complex. Mitogen-Activated Protein Kinase (MAPK) signaling pathways have been implicated in responses to stimuli such as oxidative stress and inflammation, which are critical factors in retina diseases including age-related macular degeneration. We, therefore, analyzed the Skeie and Mahajan, 2014, dataset for regional differences in the expression of MAPK-related proteins and discussed the potential implications in retinal diseases presenting with regional signs and symptoms. Regional protein expression data from the Skeie and Mahajan, 2014, study were analyzed for members of signaling networks involving MAPK and MAPK-related proteins, categorized by specific MAPK cascades, such as p38, ERK1/2, and JNK1/2, both upstream or downstream of the respective MAPK and MAPK-related proteins. We were able to identify 207 MAPK and MAPK-related proteins, 187 of which belonging to specific MAPK cascades. A total of 31 of these had been identified in the retina with two proteins, DLG2 and FLG downstream, and the other 29 upstream, of MAPK proteins. Our findings provide evidence for potential molecular substrates of retina region-specific disease manifestation and potential new targets for therapeutics development.
Collapse
Affiliation(s)
| | | | - Peter Koulen
- Vision Research Center, Department of Ophthalmology, School of Medicine, University of Missouri–Kansas City, Kansas City, MO 64108, USA
| |
Collapse
|
6
|
Trembath HE, Yeh JJ, Lopez NE. Gastrointestinal Malignancy: Genetic Implications to Clinical Applications. Cancer Treat Res 2024; 192:305-418. [PMID: 39212927 DOI: 10.1007/978-3-031-61238-1_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Advances in molecular genetics have revolutionized our understanding of the pathogenesis, progression, and therapeutic options for treating gastrointestinal (GI) cancers. This chapter provides a comprehensive overview of the molecular landscape of GI cancers, focusing on key genetic alterations implicated in tumorigenesis across various anatomical sites including GIST, colon and rectum, and pancreas. Emphasis is placed on critical oncogenic pathways, such as mutations in tumor suppressor genes, oncogenes, chromosomal instability, microsatellite instability, and epigenetic modifications. The role of molecular biomarkers in predicting prognosis, guiding treatment decisions, and monitoring therapeutic response is discussed, highlighting the integration of genomic profiling into clinical practice. Finally, we address the evolving landscape of precision oncology in GI cancers, considering targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Hannah E Trembath
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA
| | - Jen Jen Yeh
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA
| | - Nicole E Lopez
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA.
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA.
| |
Collapse
|
7
|
Rajan V, Prykhozhij SV, Pandey A, Cohen AM, Rainey JK, Berman JN. KIT D816V is dimerization-independent and activates downstream pathways frequently perturbed in mastocytosis. Br J Haematol 2023; 202:960-970. [PMID: 35245395 DOI: 10.1111/bjh.18116] [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: 12/12/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 11/30/2022]
Abstract
KIT, a type III tyrosine kinase receptor, plays a crucial role in haematopoietic development. The KIT receptor forms a dimer after ligand binding; this activates tyrosine kinase activity leading to downstream signal transduction. The D816V KIT mutation is extensively implicated in haematological malignancies, including mastocytosis and leukaemia. KIT D816V is constitutively active, but the molecular nuances that lead to constitutive tyrosine kinase activity are unclear. For the first time, we present experimental evidence that the KIT D816V mutant does not dimerize like KIT wild type. We further show evidence of decreased stabilization of the tyrosine kinase domain in the KIT D816V mutant, a phenomenon that might contribute to its constitutive activity. Since the mechanism of KIT D816V activation varies from that of the wild type, we explored downstream signal transduction events and found that even though KIT D816V targets similar signalling moieties, the signalling is amplified in the mutant compared to stem cell factor-activated wild type receptor. Uniquely, KIT D816V induces infection-related pathways and the spliceosome pathway, providing alternate options for selective as well as combinatorial therapeutic targeting.
Collapse
Affiliation(s)
- Vinothkumar Rajan
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Sergey V Prykhozhij
- Children's Hospital of Eastern Ontario (CHEO) Research Institute and University of Ottawa, Ottawa, ON, Canada
| | - Aditya Pandey
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Alejandro M Cohen
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Jan K Rainey
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, Canada
- Department of Chemistry, Dalhousie University, Halifax, NS, Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| | - Jason N Berman
- Children's Hospital of Eastern Ontario (CHEO) Research Institute and University of Ottawa, Ottawa, ON, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
8
|
Wu YN, Su X, Wang XQ, Liu NN, Xu ZW. The roles of phospholipase C-β related signals in the proliferation, metastasis and angiogenesis of malignant tumors, and the corresponding protective measures. Front Oncol 2023; 13:1231875. [PMID: 37576896 PMCID: PMC10419273 DOI: 10.3389/fonc.2023.1231875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
PLC-β is widely distributed in eukaryotic cells and is the key enzyme in phosphatidylinositol signal transduction pathway. The cellular functions regulated by its four subtypes (PLC-β1, PLC-β2, PLC-β3, PLC-β4) play an important role in maintaining homeostasis of organism. PLC-β and its related signals can promote or inhibit the occurrence and development of cancer by affecting the growth, differentiation and metastasis of cells, while targeted intervention of PLC-β1-PI3K-AKT, PLC-β2/CD133, CXCR2-NHERF1-PLC-β3, Gαq-PLC-β4-PKC-MAPK and so on can provide new strategies for the precise prevention and treatment of malignant tumors. This paper reviews the mechanism of PLC-β in various tumor cells from four aspects: proliferation and differentiation, invasion and metastasis, angiogenesis and protective measures.
Collapse
Affiliation(s)
- Yu-Nuo Wu
- Department of Clinical Medical, the First Clinical Medical College of Anhui Medical University, Hefei, Anhui, China
| | - Xing Su
- Department of Clinical Medical, the First Clinical Medical College of Anhui Medical University, Hefei, Anhui, China
| | - Xue-Qin Wang
- Department of Clinical Medical, the First Clinical Medical College of Anhui Medical University, Hefei, Anhui, China
| | - Na-Na Liu
- Department of Emergency Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhou-Wei Xu
- Department of Emergency Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui, China
| |
Collapse
|
9
|
Xiong Y, Taleb M, Misawa K, Hou Z, Banerjee S, Amador-Molina A, Jones DR, Chintala NK, Adusumilli PS. c-Kit signaling potentiates CAR T cell efficacy in solid tumors by CD28- and IL-2-independent co-stimulation. NATURE CANCER 2023; 4:1001-1015. [PMID: 37336986 PMCID: PMC10765546 DOI: 10.1038/s43018-023-00573-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 05/08/2023] [Indexed: 06/21/2023]
Abstract
The limited efficacy of chimeric antigen receptor (CAR) T cell therapy for solid tumors necessitates engineering strategies that promote functional persistence in an immunosuppressive environment. Herein, we use c-Kit signaling, a physiological pathway associated with stemness in hematopoietic progenitor cells (T cells lose expression of c-Kit during differentiation). CAR T cells with intracellular expression, but no cell-surface receptor expression, of the c-Kit D816V mutation (KITv) have upregulated STAT phosphorylation, antigen activation-dependent proliferation and CD28- and interleukin-2-independent and interferon-γ-mediated co-stimulation, augmenting the cytotoxicity of first-generation CAR T cells. This translates to enhanced survival, including in transforming growth factor-β-rich and low-antigen-expressing solid tumor models. KITv CAR T cells have equivalent or better in vivo efficacy than second-generation CAR T cells and are susceptible to tyrosine kinase inhibitors (safety switch). When combined with CD28 co-stimulation, KITv co-stimulation functions as a third signal, enhancing efficacy and providing a potent approach to treat solid tumors.
Collapse
Affiliation(s)
- Yuquan Xiong
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Meriem Taleb
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kyohei Misawa
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhaohua Hou
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Srijita Banerjee
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alfredo Amador-Molina
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David R Jones
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Navin K Chintala
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Prasad S Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
10
|
Forester CM, Oses-Prieto JA, Phillips NJ, Miglani S, Pang X, Byeon GW, DeMarco R, Burlingame A, Barna M, Ruggero D. Regulation of eIF4E guides a unique translational program to control erythroid maturation. SCIENCE ADVANCES 2022; 8:eadd3942. [PMID: 36563140 PMCID: PMC9788769 DOI: 10.1126/sciadv.add3942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 11/22/2022] [Indexed: 05/22/2023]
Abstract
Translation control is essential in balancing hematopoietic precursors and differentiation; however, the mechanisms underlying this program are poorly understood. We found that the activity of the major cap-binding protein eIF4E is unexpectedly regulated in a dynamic manner throughout erythropoiesis that is uncoupled from global protein synthesis rates. Moreover, eIF4E activity directs erythroid maturation, and increased eIF4E expression maintains cells in an early erythroid state associated with a translation program driving the expression of PTPN6 and Igf2bp1. A cytosine-enriched motif in the 5' untranslated region is important for eIF4E-mediated translation specificity. Therefore, selective translation of key target genes necessary for the maintenance of early erythroid states by eIF4E highlights a unique mechanism used by hematopoietic precursors to rapidly elicit erythropoietic maturation upon need.
Collapse
Affiliation(s)
- Craig M. Forester
- Department of Pediatrics, University of Colorado, Denver, CO 80045, USA
- Division of Pediatric Hematology/Oncology/Bone Marrow Transplant, Children’s Hospital Colorado, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Juan A. Oses-Prieto
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nancy J. Phillips
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sohit Miglani
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Xiaming Pang
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Gun Woo Byeon
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94309, USA
| | - Rachel DeMarco
- Department of Pediatrics, University of Colorado, Denver, CO 80045, USA
| | - Al Burlingame
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Maria Barna
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94309, USA
| | - Davide Ruggero
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
11
|
Rix B, Maduro AH, Bridge KS, Grey W. Markers for human haematopoietic stem cells: The disconnect between an identification marker and its function. Front Physiol 2022; 13:1009160. [PMID: 36246104 PMCID: PMC9564379 DOI: 10.3389/fphys.2022.1009160] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
The haematopoietic system is a classical stem cell hierarchy that maintains all the blood cells in the body. Haematopoietic stem cells (HSCs) are rare, highly potent cells that reside at the apex of this hierarchy and are historically some of the most well studied stem cells in humans and laboratory models, with haematopoiesis being the original system to define functional cell types by cell surface markers. Whilst it is possible to isolate HSCs to near purity, we know very little about the functional activity of markers to purify HSCs. This review will focus on the historical efforts to purify HSCs in humans based on cell surface markers, their putative functions and recent advances in finding functional markers on HSCs.
Collapse
Affiliation(s)
| | | | | | - William Grey
- *Correspondence: Katherine S. Bridge, ; William Grey,
| |
Collapse
|
12
|
Miao R, Chun H, Feng X, Gomes AC, Choi J, Pereira JP. Competition between hematopoietic stem and progenitor cells controls hematopoietic stem cell compartment size. Nat Commun 2022; 13:4611. [PMID: 35941168 PMCID: PMC9360400 DOI: 10.1038/s41467-022-32228-w] [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: 10/26/2021] [Accepted: 07/21/2022] [Indexed: 11/14/2022] Open
Abstract
Cellular competition for limiting hematopoietic factors is a physiologically regulated but poorly understood process. Here, we studied this phenomenon by hampering hematopoietic progenitor access to Leptin receptor+ mesenchymal stem/progenitor cells (MSPCs) and endothelial cells (ECs). We show that HSC numbers increase by 2-fold when multipotent and lineage-restricted progenitors fail to respond to CXCL12 produced by MSPCs and ECs. HSCs are qualitatively normal, and HSC expansion only occurs when early hematopoietic progenitors but not differentiated hematopoietic cells lack CXCR4. Furthermore, the MSPC and EC transcriptomic heterogeneity is stable, suggesting that it is impervious to major changes in hematopoietic progenitor interactions. Instead, HSC expansion correlates with increased availability of membrane-bound stem cell factor (mSCF) on MSPCs and ECs presumably due to reduced consumption by cKit-expressing hematopoietic progenitors. These studies suggest that an intricate homeostatic balance between HSCs and proximal hematopoietic progenitors is regulated by cell competition for limited amounts of mSCF. Hematopoietic stem cells (HSCs) rely on a combination of paracrine signals produced by their niche, including SCF. Here the authors show that HSCs and hematopoietic progenitors compete for limited amounts of membrane-bound SCF.
Collapse
Affiliation(s)
- Runfeng Miao
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, 06519, USA
| | - Harim Chun
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Xing Feng
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, 06519, USA
| | - Ana Cordeiro Gomes
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, 06519, USA.,i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
| | - Jungmin Choi
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea. .,Department of Genetics, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, 06519, USA.
| | - João P Pereira
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, 06519, USA.
| |
Collapse
|
13
|
Lummertz da Rocha E, Kubaczka C, Sugden WW, Najia MA, Jing R, Markel A, LeBlanc ZC, Dos Santos Peixoto R, Falchetti M, Collins JJ, North TE, Daley GQ. CellComm infers cellular crosstalk that drives haematopoietic stem and progenitor cell development. Nat Cell Biol 2022; 24:579-589. [PMID: 35414020 PMCID: PMC10123873 DOI: 10.1038/s41556-022-00884-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/01/2022] [Indexed: 01/05/2023]
Abstract
Intercellular communication orchestrates a multitude of physiologic and pathologic conditions. Algorithms to infer cell-cell communication and predict downstream signalling and regulatory networks are needed to illuminate mechanisms of stem cell differentiation and tissue development. Here, to fill this gap, we developed and applied CellComm to investigate how the aorta-gonad-mesonephros microenvironment dictates haematopoietic stem and progenitor cell emergence. We identified key microenvironmental signals and transcriptional networks that regulate haematopoietic development, including Stat3, Nr0b2, Ybx1 and App, and confirmed their roles using zebrafish, mouse and human models. Notably, CellComm revealed extensive crosstalk among signalling pathways and convergence on common transcriptional regulators, indicating a resilient developmental programme that ensures dynamic adaptation to changes in the embryonic environment. Our work provides an algorithm and data resource for the scientific community.
Collapse
Affiliation(s)
- Edroaldo Lummertz da Rocha
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Caroline Kubaczka
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Wade W Sugden
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA
| | - Mohamad Ali Najia
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard-MIT Health Sciences & Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ran Jing
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Arianna Markel
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Zachary C LeBlanc
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA
| | - Rafael Dos Santos Peixoto
- Undergraduate program in Automation and Control Engineering, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Marcelo Falchetti
- Graduate Program of Pharmacology, Center for Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - James J Collins
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard-MIT Health Sciences & Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Trista E North
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA.
- Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA.
| | - George Q Daley
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA.
- Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA.
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Khaddour K, Maahs L, Avila-Rodriguez AM, Maamar Y, Samaan S, Ansstas G. Melanoma Targeted Therapies beyond BRAF-Mutant Melanoma: Potential Druggable Mutations and Novel Treatment Approaches. Cancers (Basel) 2021; 13:5847. [PMID: 34831002 PMCID: PMC8616477 DOI: 10.3390/cancers13225847] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/24/2022] Open
Abstract
Melanomas exhibit the highest rate of somatic mutations among all different types of cancers (with the exception of BCC and SCC). The accumulation of a multimode of mutations in the driver oncogenes are responsible for the proliferative, invasive, and aggressive nature of melanomas. High-resolution and high-throughput technology has led to the identification of distinct mutational signatures and their downstream alterations in several key pathways that contribute to melanomagenesis. This has enabled the development of individualized treatments by targeting specific molecular alterations that are vital for cancer cell survival, which has resulted in improved outcomes in several cancers, including melanomas. To date, BRAF and MEK inhibitors remain the only approved targeted therapy with a high level of evidence in BRAFV600E/K mutant melanomas. The lack of approved precision drugs in melanomas, relative to other cancers, despite harboring one of the highest rates of somatic mutations, advocates for further research to unveil effective therapeutics. In this review, we will discuss potential druggable mutations and the ongoing research of novel individualized treatment approaches targeting non-BRAF mutations in melanomas.
Collapse
Affiliation(s)
- Karam Khaddour
- Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (L.M.); (A.M.A.-R.)
| | - Lucas Maahs
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (L.M.); (A.M.A.-R.)
| | - Ana Maria Avila-Rodriguez
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (L.M.); (A.M.A.-R.)
| | - Yazan Maamar
- Division of Hematology and Oncology, Department of Medicine, University of Tishreen Lattakia, Lattakia 2217, Syria;
| | - Sami Samaan
- Department of Medicine, American University of Beirut, Beirut 1107, Lebanon;
| | - George Ansstas
- Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| |
Collapse
|
15
|
Georas SN, Donohue P, Connolly M, Wechsler ME. JAK inhibitors for asthma. J Allergy Clin Immunol 2021; 148:953-963. [PMID: 34625142 DOI: 10.1016/j.jaci.2021.08.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/20/2021] [Accepted: 08/21/2021] [Indexed: 02/06/2023]
Abstract
Asthma is an inflammatory disease of the airways characterized by intermittent episodes of wheezing, chest tightness, and cough. Many of the inflammatory pathways implicated in asthma involve cytokines and growth factors that activate Janus kinases (JAKs). The discovery of the JAK/signal transducer and activator of transcription (STAT) signaling pathway was a major breakthrough that revolutionized our understanding of cell growth and differentiation. JAK inhibitors are under active investigation for immune and inflammatory diseases, and they have demonstrated clinical efficacy in diseases such as rheumatoid arthritis and atopic dermatitis. Substantial preclinical data support the idea that inhibiting JAKs will ameliorate airway inflammation and hyperreactivity in asthma. Here, we review the rationale for use of JAK inhibitors in different asthma endotypes as well as the preclinical and early clinical evidence supporting such use. We review preclinical data from the use of systemic and inhaled JAK inhibitors in animal models of asthma and safety data based on the use of JAK inhibitors in other diseases. We conclude that JAK inhibitors have the potential to usher in a new era of anti-inflammatory treatment for asthma.
Collapse
Affiliation(s)
- Steve N Georas
- Division of Pulmonary and Critical Care Medicine, University of Rochester Medical Center, Rochester, NY.
| | | | - Margaret Connolly
- Division of Pulmonary and Critical Care Medicine, University of Rochester Medical Center, Rochester, NY
| | | |
Collapse
|
16
|
Chen T, Ni N, Yuan L, Xu L, Bahri N, Sun B, Wu Y, Ou WB. Proteasome Inhibition Suppresses KIT-Independent Gastrointestinal Stromal Tumors Via Targeting Hippo/YAP/Cyclin D1 Signaling. Front Pharmacol 2021; 12:686874. [PMID: 34025442 PMCID: PMC8134732 DOI: 10.3389/fphar.2021.686874] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 04/23/2021] [Indexed: 12/01/2022] Open
Abstract
Purpose: Gastrointestinal stromal tumors (GISTs) are the most common malignant tumor of mesenchymal origin of the digestive tract. A yet more challenging resistance mechanism involves transition from oncogenic KIT to a new imatinib-insensitive oncogenic driver, heralded by loss of KIT expression. Our recent studies have shown that inhibition of cyclin D1 and Hippo signaling, which are overexpressed in KIT-independent GIST, is accompanied by anti-proliferative and apoptosis-promoting effects. PRKCQ, JUN, and the Hippo/YAP pathway coordinately regulate GIST cyclin D1 expression. Thus, targeting of these pathways could be effective therapeutically for these now untreatable tumors. Methods: Targeting cyclin D1 expression of small molecular drugs was screened by a cell monolayer growth and western blotting. The biologic mechanisms of bortezomib to KIT-independent GISTs were assessed by immunoblotting, qRT-PCR, cell viability, colony growth, cell cycle analysis, apoptosis, migration and invasiveness. Results: In the initial small molecular inhibitor screening in KIT-independent GIST62, we found that bortezomib-mediated inhibition of the ubiquitin-proteasome machinery showed anti-proliferative effects of KIT-independent GIST cells via downregulation of cyclin D1 and induction of p53 and p21. Treatment with proteasome inhibitor, bortezomib, led to downregulation of cyclin D1 and YAP/TAZ and an increase in the cleaved PARP expression in three KIT-independent GIST cell lines (GIST48B, GIST54, and GIST226). Additionally, it induced p53 and p21 expression in GIST48B and GIST54, increased apoptosis, and led to cell cycle G1/G2-phase arrest, decreased cell viability, colony formation, as well as migration and invasiveness in all GIST cell lines. Conclusion: Although our findings are early proof-of-principle, there are signs of a potential effective treatment for KIT-independent GISTs, the data highlight that targeting of cyclin D1 and Hippo/YAP by bortezomib warrants evaluation as a novel therapeutic strategy in KIT-independent GISTs.
Collapse
Affiliation(s)
- Ting Chen
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Nan Ni
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Li Yuan
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Liangliang Xu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Nacef Bahri
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Boshu Sun
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yuehong Wu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Wen-Bin Ou
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China.,Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
17
|
Paivandy A, Pejler G. Novel Strategies to Target Mast Cells in Disease. J Innate Immun 2021; 13:131-147. [PMID: 33582673 DOI: 10.1159/000513582] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/03/2020] [Indexed: 12/12/2022] Open
Abstract
Mast cells (MCs) are versatile effector cells of the immune system, characterized by a large content of secretory granules containing a variety of inflammatory mediators. They are implicated in the host protection toward various external insults, but are mostly well known for their detrimental impact on a variety of pathological conditions, including allergic disorders such as asthma and a range of additional disease settings. Based on this, there is currently a large demand for therapeutic regimens that can dampen the detrimental impact of MCs in these respective pathological conditions. This can be accomplished by several strategies, including targeting of individual mediators released by MCs, blockade of receptors for MC-released compounds, inhibition of MC activation, limiting mast cell growth or by inducing mast cell apoptosis. Here, we review the currently available and emerging regimens to interfere with harmful mast cell activities in asthma and other pathological settings and discuss the advantages and limitations of such strategies.
Collapse
Affiliation(s)
- Aida Paivandy
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden,
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.,Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
18
|
Harris KS, Shi L, Foster BM, Mobley ME, Elliott PL, Song CJ, Watabe K, Langefeld CD, Kerr BA. CD117/c-kit defines a prostate CSC-like subpopulation driving progression and TKI resistance. Sci Rep 2021; 11:1465. [PMID: 33446896 PMCID: PMC7809150 DOI: 10.1038/s41598-021-81126-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer stem-like cells (CSCs) are associated with cancer progression, metastasis, and recurrence, and may also represent a subset of circulating tumor cells (CTCs). In our prior study, CTCs in advanced prostate cancer patients were found to express CD117/c-kit in a liquid biopsy. Whether CD117 expression played an active or passive role in the aggressiveness and migration of these CTCs remained an open question. In this study, we show that CD117 expression in prostate cancer patients is associated with decreased overall and progression-free survival and that activation and phosphorylation of CD117 increases in prostate cancer patients with higher Gleason grades. To determine how CD117 expression and activation by its ligand stem cell factor (SCF, kit ligand, steel factor) alter prostate cancer aggressiveness, we used C4-2 and PC3-mm human prostate cancer cells, which contain a CD117+ subpopulation. We demonstrate that CD117+ cells display increased proliferation and migration. In prostaspheres, CD117 expression enhances sphere formation. In both 2D and 3D cultures, stemness marker gene expression is higher in CD117+ cells. Using xenograft limiting dilution assays and serial tumor initiation assays, we show that CD117+ cells represent a CSC population. Combined, these data indicate that CD117 expression potentially promotes tumor initiation and metastasis. Further, in cell lines, CD117 activation by SCF promotes faster proliferation and invasiveness, while blocking CD117 activation with tyrosine kinase inhibitors (TKIs) decreased progression in a context-dependent manner. We demonstrate that CD117 expression and activation drives prostate cancer aggressiveness through the CSC phenotype and TKI resistance.
Collapse
Affiliation(s)
- Koran S Harris
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Lihong Shi
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Brittni M Foster
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Mary E Mobley
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Phyllis L Elliott
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Conner J Song
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA.,Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, 27157, USA
| | - Carl D Langefeld
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, 27157, USA.,Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Bethany A Kerr
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA. .,Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, 27157, USA. .,Department of Urology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
19
|
Jobani BM, Mohebi E, Najafzadeh N. In Vitro Anticancer Effects of All-trans Retinoic Acid in Combination with Dacarbazine against CD117+ Melanoma Cells. Drug Res (Stuttg) 2020; 70:563-569. [PMID: 33022719 DOI: 10.1055/a-1240-0072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Malignant melanoma is a common form of skin cancer that contains different cell types recognized by various cell surface markers. Dacarbazine-based combination chemotherapy is frequently used for the treatment of melanoma. Despite its potent anticancer properties, resistance to dacarbazine develops in malignant melanoma. Here, we aim to improve response to dacarbazine therapy by pretreatment with all-trans retinoic acid (ATRA) in CD117+ melanoma cells. METHODS The CD117+ melanoma cells were sorted from A375 malignant melanoma cell line using magnetic-activated cell sorting (MACS). The cell viability was examined by cell proliferation assay (MTT). Apoptosis was determined by acridine orange/ ethidium bromide staining. Indeed, we performed flow cytometry to evaluate the cell cycle arrest. RESULTS Here, the CD117+ melanoma cells were incubated with various concentrations of ATRA, dacarbazine, and their combination to determine IC50 values. We found that 20 µM ATRA treatment followed by dacarbazine was found to be more effective than dacarbazine alone. There was an indication that the combination of ATRA with dacarbazine (ATRA/dacarbazine) caused more apoptosis and necrosis in the melanoma cells (P<0.05). Furthermore, ATRA/dacarbazine treatment inhibited the cell at the G0/G1 phase, while dacarbazine alone inhibited the cells at S phase. CONCLUSION Collectively, combined treatment with ATRA and dacarbazine induced more apoptosis and enhanced the cell cycle arrest of CD117+ melanoma cells. These results suggested that ATRA increased the sensitivity of melanoma cells to the effect of dacarbazine.
Collapse
Affiliation(s)
- Bahareh Mohammadi Jobani
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Mohebi
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nowruz Najafzadeh
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
20
|
PIM1 Promotes Survival of Cardiomyocytes by Upregulating c-Kit Protein Expression. Cells 2020; 9:cells9092001. [PMID: 32878131 PMCID: PMC7563506 DOI: 10.3390/cells9092001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/21/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022] Open
Abstract
Enhancing cardiomyocyte survival is crucial to blunt deterioration of myocardial structure and function following pathological damage. PIM1 (Proviral Insertion site in Murine leukemia virus (PIM) kinase 1) is a cardioprotective serine threonine kinase that promotes cardiomyocyte survival and antagonizes senescence through multiple concurrent molecular signaling cascades. In hematopoietic stem cells, PIM1 interacts with the receptor tyrosine kinase c-Kit upstream of the ERK (Extracellular signal-Regulated Kinase) and Akt signaling pathways involved in cell proliferation and survival. The relationship between PIM1 and c-Kit activity has not been explored in the myocardial context. This study delineates the interaction between PIM1 and c-Kit leading to enhanced protection of cardiomyocytes from stress. Elevated c-Kit expression is induced in isolated cardiomyocytes from mice with cardiac-specific overexpression of PIM1. Co-immunoprecipitation and proximity ligation assay reveal protein–protein interaction between PIM1 and c-Kit. Following treatment with Stem Cell Factor, PIM1-overexpressing cardiomyocytes display elevated ERK activity consistent with c-Kit receptor activation. Functionally, elevated c-Kit expression confers enhanced protection against oxidative stress in vitro. This study identifies the mechanistic relationship between PIM1 and c-Kit in cardiomyocytes, demonstrating another facet of cardioprotection regulated by PIM1 kinase.
Collapse
|
21
|
Persistent Human KIT Receptor Signaling Disposes Murine Placenta to Premature Differentiation Resulting in Severely Disrupted Placental Structure and Functionality. Int J Mol Sci 2020; 21:ijms21155503. [PMID: 32752102 PMCID: PMC7432075 DOI: 10.3390/ijms21155503] [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: 07/08/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022] Open
Abstract
Activating mutations in the human KIT receptor is known to drive severe hematopoietic disorders and tumor formation spanning various entities. The most common mutation is the substitution of aspartic acid at position 816 to valine (D816V), rendering the receptor constitutively active independent of ligand binding. As the role of the KIT receptor in placental signaling cascades is poorly understood, we analyzed the impact of KITD816V expression on placental development using a humanized mouse model. Placentas from KITD816V animals present with a grossly changed morphology, displaying a reduction in labyrinth and spongiotrophoblast layer and an increase in the Parietal Trophoblast Giant Cell (P-TGC) layer. Elevated differentiation to P-TGCs was accompanied with reduced differentiation to other Trophoblast Giant Cell (TGC) subtypes and by severe decrease in proliferation. The embryos display growth retardation and die in utero. KITD816V-trophoblast stem cells (TSC) differentiate much faster compared to wild type (WT) controls. In undifferentiated KITD816V-TSCs, levels of Phosphorylated Extracellular-signal Regulated Kinase (P-ERK) and Phosphorylated Protein Kinase B (P-AKT) are comparable to wildtype cultures differentiating for 3–6 days. Accordingly, P-TGC markers Placental Lactogen 1 (PL1) and Proliferin (PLF) are upregulated as well. The results reveal that KIT signaling orchestrates the fine-tuned differentiation of the placenta, with special emphasis on P-TGC differentiation. Appropriate control of KIT receptor action is therefore essential for placental development and nourishment of the embryo.
Collapse
|
22
|
Ward RA, Fawell S, Floc'h N, Flemington V, McKerrecher D, Smith PD. Challenges and Opportunities in Cancer Drug Resistance. Chem Rev 2020; 121:3297-3351. [PMID: 32692162 DOI: 10.1021/acs.chemrev.0c00383] [Citation(s) in RCA: 237] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There has been huge progress in the discovery of targeted cancer therapies in recent years. However, even for the most successful and impactful cancer drugs which have been approved, both innate and acquired mechanisms of resistance are commonplace. These emerging mechanisms of resistance have been studied intensively, which has enabled drug discovery scientists to learn how it may be possible to overcome such resistance in subsequent generations of treatments. In some cases, novel drug candidates have been able to supersede previously approved agents; in other cases they have been used sequentially or in combinations with existing treatments. This review summarizes the current field in terms of the challenges and opportunities that cancer resistance presents to drug discovery scientists, with a focus on small molecule therapeutics. As part of this review, common themes and approaches have been identified which have been utilized to successfully target emerging mechanisms of resistance. This includes the increase in target potency and selectivity, alternative chemical scaffolds, change of mechanism of action (covalents, PROTACs), increases in blood-brain barrier permeability (BBBP), and the targeting of allosteric pockets. Finally, wider approaches are covered such as monoclonal antibodies (mAbs), bispecific antibodies, antibody drug conjugates (ADCs), and combination therapies.
Collapse
Affiliation(s)
- Richard A Ward
- Medicinal Chemistry, Oncology R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Stephen Fawell
- Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Nicolas Floc'h
- Bioscience, Oncology R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | | | | | - Paul D Smith
- Bioscience, Oncology R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| |
Collapse
|
23
|
Grahn THM, Niroula A, Végvári Á, Oburoglu L, Pertesi M, Warsi S, Safi F, Miharada N, Garcia SC, Siva K, Liu Y, Rörby E, Nilsson B, Zubarev RA, Karlsson S. S100A6 is a critical regulator of hematopoietic stem cells. Leukemia 2020; 34:3323-3337. [PMID: 32555370 PMCID: PMC7685984 DOI: 10.1038/s41375-020-0901-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 01/22/2023]
Abstract
The fate options of hematopoietic stem cells (HSCs) include self-renewal, differentiation, migration, and apoptosis. HSCs self-renewal divisions in stem cells are required for rapid regeneration during tissue damage and stress, but how precisely intracellular calcium signals are regulated to maintain fate options in normal hematopoiesis is unclear. S100A6 knockout (KO) HSCs have reduced total cell numbers in the HSC compartment, decreased myeloid output, and increased apoptotic HSC numbers in steady state. S100A6KO HSCs had impaired self-renewal and regenerative capacity, not responding to 5-Fluorouracil. Our transcriptomic and proteomic profiling suggested that S100A6 is a critical HSC regulator. Intriguingly, S100A6KO HSCs showed decreased levels of phosphorylated Akt (p-Akt) and Hsp90, with an impairment of mitochondrial respiratory capacity and a reduction of mitochondrial calcium levels. We showed that S100A6 regulates intracellular and mitochondria calcium buffering of HSC upon cytokine stimulation and have demonstrated that Akt activator SC79 reverts the levels of intracellular and mitochondrial calcium in HSC. Hematopoietic colony-forming activity and the Hsp90 activity of S100A6KO are restored through activation of the Akt pathway. We show that p-Akt is the prime downstream mechanism of S100A6 in the regulation of HSC self-renewal by specifically governing mitochondrial metabolic function and Hsp90 protein quality.
Collapse
Affiliation(s)
- Tan Hooi Min Grahn
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University Hospital, 22184, Lund, Sweden.
| | - Abhishek Niroula
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, BMC B13, SE-221 84, Lund, Sweden
| | - Ákos Végvári
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solnavägen 9, SE-171 65, Solna, Sweden
| | - Leal Oburoglu
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University Hospital, 22184, Lund, Sweden
| | - Maroulio Pertesi
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, BMC B13, SE-221 84, Lund, Sweden
| | - Sarah Warsi
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University Hospital, 22184, Lund, Sweden
| | - Fatemeh Safi
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University Hospital, 22184, Lund, Sweden
| | - Natsumi Miharada
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University Hospital, 22184, Lund, Sweden
| | - Sandra C Garcia
- Department of Molecular, Cell and Developmental Biology, Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Kavitha Siva
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University Hospital, 22184, Lund, Sweden
| | - Yang Liu
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University Hospital, 22184, Lund, Sweden
| | - Emma Rörby
- Experimental Hematology Unit, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Björn Nilsson
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, BMC B13, SE-221 84, Lund, Sweden
| | - Roman A Zubarev
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solnavägen 9, SE-171 65, Solna, Sweden
| | - Stefan Karlsson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University Hospital, 22184, Lund, Sweden.
| |
Collapse
|
24
|
The c-kit Receptor Tyrosine Kinase Marks Sweet or Umami Sensing T1R3 Positive Adult Taste Cells in Mice. CHEMOSENS PERCEPT 2020. [DOI: 10.1007/s12078-019-09277-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
25
|
Lee D, Kim DW, Cho JY. Role of growth factors in hematopoietic stem cell niche. Cell Biol Toxicol 2020; 36:131-144. [PMID: 31897822 DOI: 10.1007/s10565-019-09510-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 12/16/2019] [Indexed: 12/20/2022]
Abstract
Hematopoietic stem cells (HSCs) produce new blood cells everyday throughout life, which is maintained by the self-renewal and differentiation ability of HSCs. This is not controlled by the HSCs alone, but rather by the complex and exquisite microenvironment surrounding the HSCs, which is called the bone marrow niche and consists of various bone marrow cells, growth factors, and cytokines. It is essential to understand the characteristic role of the stem cell niche and the growth factors in the niche formation. In this review, we describe the role of the bone marrow niche and factors for niche homeostasis, and also summarize the latest research related to stem cell niche.
Collapse
Affiliation(s)
- Dabin Lee
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742, South Korea
| | - Dong Wook Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742, South Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742, South Korea.
| |
Collapse
|
26
|
Raeisi M, Nikhanfar AR, Nejate B, Movassaghpour Akbari AA, Dolatkhah R, Roosta Y, Sanaat Z. Role of CD135/CD117 on Prognosis and Overall Survival of Acute Myeloid Leukemia. Asian Pac J Cancer Prev 2019; 20:2625-2631. [PMID: 31554356 PMCID: PMC6976829 DOI: 10.31557/apjcp.2019.20.9.2625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Indexed: 11/25/2022] Open
Abstract
Background: The key proliferative RTKs for AML include c-KIT receptor (CD117) and FLT-3 receptor (CD135). The aim of this study was to evaluate the CD135 and CD117 expression, co-expression of CD135 and CD117 (CD135+CD117), and the association of that co-expression with Event Free Survival (DFS) and Overall Survival (OS) rates. Material and Methods: We analyzed CD117 and CD135 expression on AML blasts by flow cytometry and its association with Event Free Survival (DFS) and Overall Survival (OS) in 66 AML treated on Hematology-Oncology Research Center, Iran, Tabriz. Results: The overall OS and EFS were 50% and 80.3% respectively during our study. Cox-regression analysis revealed that a poor EFS was significantly associated a low CD135 (HR 0.34, 95% CI 0.13–0.88, P = 0.02). Conclusion: This is the first study from Iran to show that the expressions of CD135, CD117 is easily measurable by routine diagnostic flow-cytometry, and CD135+117 were not significantly associated with CR, EFS, or OS .
Collapse
Affiliation(s)
| | - Ali Reza Nikhanfar
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Babak Nejate
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Roya Dolatkhah
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Yousef Roosta
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Zohreh Sanaat
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
27
|
Signalling circuits that direct early B-cell development. Biochem J 2019; 476:769-778. [PMID: 30842310 DOI: 10.1042/bcj20180565] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/11/2019] [Accepted: 02/15/2019] [Indexed: 12/23/2022]
Abstract
In mammals, the B-cell lineage arises from pluripotent progenitors in the bone marrow. During their development, B-cells undergo lineage specification and commitment, followed by expansion and selection. These processes are mediated by regulated changes in gene expression programmes, rearrangements of immunoglobulin (Ig) genes, and well-timed rounds of proliferation and apoptosis. Many of these processes are initiated by environmental factors including cytokines, chemokines, and cell-cell contacts. Developing B-cells process these environmental cues into stage-specific functions via signalling pathways including the PI3K, MAPK, or JAK-STAT pathway. The cytokines FLT3-Ligand and c-Kit-Ligand are important for the early expansion of the B-cell precursors at different developmental stages and conditions. Interleukin 7 is essential for commitment to the B-cell lineage and for orchestrating the Ig recombination machinery. After rearrangement of the immunoglobulin heavy chain, proliferation and apoptosis, and thus selection, are mediated by the clonal pre-B-cell receptor, and, following light chain rearrangement, by the B-cell receptor.
Collapse
|
28
|
Abstract
KIT is a receptor tyrosine kinase that after binding to its ligand stem cell factor activates signaling cascades linked to biological processes such as proliferation, differentiation, migration and cell survival. Based on studies performed on SCF and/or KIT mutant animals that presented anemia, sterility, and/or pigmentation disorders, KIT signaling was mainly considered to be involved in the regulation of hematopoiesis, gametogenesis, and melanogenesis. More recently, novel animal models and ameliorated cellular and molecular techniques have led to the discovery of a widen repertoire of tissue compartments and functions that are being modulated by KIT. This is the case for the lung, heart, nervous system, gastrointestinal tract, pancreas, kidney, liver, and bone. For this reason, the tyrosine kinase inhibitors that were originally developed for the treatment of hemato-oncological diseases are being currently investigated for the treatment of non-oncological disorders such as asthma, rheumatoid arthritis, and alzheimer's disease, among others. The beneficial effects of some of these tyrosine kinase inhibitors have been proven to depend on KIT inhibition. This review will focus on KIT expression and regulation in healthy and pathologic conditions other than cancer. Moreover, advances in the development of anti-KIT therapies, including tyrosine kinase inhibitors, and their application will be discussed.
Collapse
|
29
|
Caslin HL, Kiwanuka KN, Haque TT, Taruselli MT, MacKnight HP, Paranjape A, Ryan JJ. Controlling Mast Cell Activation and Homeostasis: Work Influenced by Bill Paul That Continues Today. Front Immunol 2018; 9:868. [PMID: 29755466 PMCID: PMC5932183 DOI: 10.3389/fimmu.2018.00868] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/09/2018] [Indexed: 01/13/2023] Open
Abstract
Mast cells are tissue resident, innate immune cells with heterogenous phenotypes tuned by cytokines and other microenvironmental stimuli. Playing a protective role in parasitic, bacterial, and viral infections, mast cells are also known for their role in the pathogenesis of allergy, asthma, and autoimmune diseases. Here, we review factors controlling mast cell activation, with a focus on receptor signaling and potential therapies for allergic disease. Specifically, we will discuss our work with FcεRI and FγR signaling, IL-4, IL-10, and TGF-β1 treatment, and Stat5. We conclude with potential therapeutics for allergic disease. Much of these efforts have been influenced by the work of Bill Paul. With many mechanistic targets for mast cell activation and different classes of therapeutics being studied, there is reason to be hopeful for continued clinical progress in this area.
Collapse
Affiliation(s)
- Heather L Caslin
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Kasalina N Kiwanuka
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Tamara T Haque
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Marcela T Taruselli
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - H Patrick MacKnight
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Anuya Paranjape
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - John J Ryan
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
30
|
Li G, Zhang Y, Cai S, Sun M, Wang J, Li S, Li X, Tighe S, Chen S, Xie H, Zhu Y. Human limbal niche cells are a powerful regenerative source for the prevention of limbal stem cell deficiency in a rabbit model. Sci Rep 2018; 8:6566. [PMID: 29700361 PMCID: PMC5919904 DOI: 10.1038/s41598-018-24862-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 04/09/2018] [Indexed: 12/13/2022] Open
Abstract
In this article, human limbal niche cells (LNC) or bone marrow derived mesenchymal stem cells (BMMSC) were used to prevent limbal stem cell deficiency (LSCD) in an alkali burn rabbit model and their results were compared. The epithelial cell defect area, corneal neovascularization, and the print cell cytometry were quantified to grade the severity of LSCD. Three months after the alkali burn, a partial LSCD was observed in the control group (no treatment) indicated by chronic corneal epithelial defects, positive corneal fluorescein staining, neovascularization and goblet cell migration. In contrast, the severity of LSCD in both the LNC and BMMSC transplantation groups was dramatically reduced as shown by smaller epithelial cell defects, decreased fluorescein sodium staining, decreased neovascularization and decreased goblet cell density. Interestingly, the LNC group was shown to more effectively prevent LSCD than the BMMSC group. Further analysis indicated subconjunctivally transplanted LNCs were more powerful than BMMSCs to prevent LSCD, at least partially, due to increased activation of SCF-c-Kit signal. We conclude that LNCs are a more powerful resource than BMMSCs to prevent LSCD in an alkali burn rabbit model, at least partially due to increased activation of SCF signaling.
Collapse
Affiliation(s)
- Guigang Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Research and Development Department, Tissue Tech, Inc, Miami, FL, 33126, USA
| | - Yuan Zhang
- Research and Development Department, Tissue Tech, Inc, Miami, FL, 33126, USA
| | - Subo Cai
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ming Sun
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Juan Wang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shen Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xinyu Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sean Tighe
- Research and Development Department, Tissue Tech, Inc, Miami, FL, 33126, USA
| | - Shuangling Chen
- Research and Development Department, Tissue Tech, Inc, Miami, FL, 33126, USA
| | - Huatao Xie
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yingting Zhu
- Research and Development Department, Tissue Tech, Inc, Miami, FL, 33126, USA.
| |
Collapse
|
31
|
S1P₄ Regulates Passive Systemic Anaphylaxis in Mice but Is Dispensable for Canonical IgE-Mediated Responses in Mast Cells. Int J Mol Sci 2018; 19:ijms19051279. [PMID: 29693558 PMCID: PMC5983835 DOI: 10.3390/ijms19051279] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/18/2018] [Accepted: 04/18/2018] [Indexed: 02/07/2023] Open
Abstract
Mast cells are key players in the development of inflammatory allergic reactions. Cross-linking of the high-affinity receptor for IgE (FcεRI) on mast cells leads to the generation and secretion of the sphingolipid mediator, sphingosine-1-phosphate (S1P) which is able, in turn, to transactivate its receptors on mast cells. Previous reports have identified the expression of two of the five receptors for S1P on mast cells, S1P1 and S1P2, with functions in FcεRI-mediated chemotaxis and degranulation, respectively. Here, we show that cultured mouse mast cells also express abundant message for S1P4. Genetic deletion of S1pr4 did not affect the differentiation of bone marrow progenitors into mast cells or the proliferation of mast cells in culture. A comprehensive characterization of IgE-mediated responses in S1P4-deficient bone marrow-derived and peritoneal mouse mast cells indicated that this receptor is dispensable for mast cell degranulation, cytokine/chemokine production and FcεRI-mediated chemotaxis in vitro. However, interleukin-33 (IL-33)-mediated enhancement of IgE-induced degranulation was reduced in S1P4-deficient peritoneal mast cells, revealing a potential negative regulatory role for S1P4 in an IL-33-rich environment. Surprisingly, genetic deletion of S1pr4 resulted in exacerbation of passive systemic anaphylaxis to IgE/anti-IgE in mice, a phenotype likely related to mast cell-extrinsic influences, such as the high circulating levels of IgE in these mice which increases FcεRI expression and consequently the extent of the response to FcεRI engagement. Thus, we provide evidence that S1P4 modulates anaphylaxis in an unexpected manner that does not involve regulation of mast cell responsiveness to IgE stimulation.
Collapse
|
32
|
Foster BM, Zaidi D, Young TR, Mobley ME, Kerr BA. CD117/c-kit in Cancer Stem Cell-Mediated Progression and Therapeutic Resistance. Biomedicines 2018. [PMID: 29518044 PMCID: PMC5874688 DOI: 10.3390/biomedicines6010031] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Metastasis is the primary cause of cancer patient morbidity and mortality, but due to persisting gaps in our knowledge, it remains untreatable. Metastases often occur as patient tumors progress or recur after initial therapy. Tumor recurrence at the primary site may be driven by a cancer stem-like cell or tumor progenitor cell, while recurrence at a secondary site is driven by metastatic cancer stem cells or metastasis-initiating cells. Ongoing efforts are aimed at identifying and characterizing these stem-like cells driving recurrence and metastasis. One potential marker for the cancer stem-like cell subpopulation is CD117/c-kit, a tyrosine kinase receptor associated with cancer progression and normal stem cell maintenance. Further, activation of CD117 by its ligand stem cell factor (SCF; kit ligand) in the progenitor cell niche stimulates several signaling pathways driving proliferation, survival, and migration. This review examines evidence that the SCF/CD117 signaling axis may contribute to the control of cancer progression through the regulation of stemness and resistance to tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Brittni M Foster
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | - Danish Zaidi
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | - Tyler R Young
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | - Mary E Mobley
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | - Bethany A Kerr
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA.
| |
Collapse
|
33
|
Wilhelm T, Bick F, Peters K, Mohta V, Tirosh B, Patterson JB, Kharabi-Masouleh B, Huber M. Infliction of proteotoxic stresses by impairment of the unfolded protein response or proteasomal inhibition as a therapeutic strategy for mast cell leukemia. Oncotarget 2017; 9:2984-3000. [PMID: 29423023 PMCID: PMC5790440 DOI: 10.18632/oncotarget.23354] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 12/03/2017] [Indexed: 01/05/2023] Open
Abstract
The intensity and duration of endoplasmic reticulum (ER) stress converts the unfolded protein response (UPR) from an adaptive into a terminal response. The first regulates homeostasis, the latter triggers apoptosis. Cells that rapidly proliferate and possess developed secretory capabilities, such as leukemia cells, depend on an efficiently operating UPR to maintain proteostasis. Activation of terminal UPR by either blockade of adaptive UPR or exaggeration of ER stress has been explored as a novel approach in cancer therapy. For mast cell leukemia (MCL) the efficacy of both approaches, by utilizing the KITV560G,D816V-positive MCL cell line HMC-1.2, was investigated. We show that HMC-1.2 cells display a tonic activation of the IRE1α arm of the UPR, which constitutively generates spliced XBP1. Inhibition of IRE1α by different types of inhibitors (MKC-8866, STF-083010, and KIRA6) suppressed proliferation at concentrations needed for blockade of IRE1α-mediated XBP1 splicing. At higher concentrations, these inhibitors triggered an apoptotic response. Blocking the proteasome by bortezomib, which confers an exaggerated UPR, resulted in a marked cytotoxic response. Bortezomib treatment also caused activation of the kinase JNK, which played a pro-proliferative and anti-apoptotic role. Hence, the combination of bortezomib with a JNK inhibitor synergized to induce cell death. In summary, the UPR can be addressed as an effective therapeutic target against KITD816V-positive MCL.
Collapse
Affiliation(s)
- Thomas Wilhelm
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Fabian Bick
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Kerstin Peters
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Vrinda Mohta
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Boaz Tirosh
- The Institute of Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Behzad Kharabi-Masouleh
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
34
|
Dufresne S, Rébillard A, Muti P, Friedenreich CM, Brenner DR. A Review of Physical Activity and Circulating miRNA Expression: Implications in Cancer Risk and Progression. Cancer Epidemiol Biomarkers Prev 2017; 27:11-24. [DOI: 10.1158/1055-9965.epi-16-0969] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 03/17/2017] [Accepted: 10/26/2017] [Indexed: 11/16/2022] Open
|
35
|
Kaitsiotou H, Keul M, Hardick J, Mühlenberg T, Ketzer J, Ehrt C, Krüll J, Medda F, Koch O, Giordanetto F, Bauer S, Rauh D. Inhibitors to Overcome Secondary Mutations in the Stem Cell Factor Receptor KIT. J Med Chem 2017; 60:8801-8815. [DOI: 10.1021/acs.jmedchem.7b00841] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Helena Kaitsiotou
- Faculty
of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Straße 4a, D-44227 Dortmund, Germany
| | - Marina Keul
- Faculty
of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Straße 4a, D-44227 Dortmund, Germany
| | - Julia Hardick
- Faculty
of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Straße 4a, D-44227 Dortmund, Germany
| | - Thomas Mühlenberg
- Department
of Medical Oncology, Sarcoma Centre West German Cancer Centre University Duisburg−Essen, Medical School, Hufelandstraße 55, D-45122 Essen, Germany
- Germany
and German Cancer Consortium (DKTK), Partner Site University Hospital Essen, D-45147 Essen, Germany
| | - Julia Ketzer
- Department
of Medical Oncology, Sarcoma Centre West German Cancer Centre University Duisburg−Essen, Medical School, Hufelandstraße 55, D-45122 Essen, Germany
- Germany
and German Cancer Consortium (DKTK), Partner Site University Hospital Essen, D-45147 Essen, Germany
| | - Christiane Ehrt
- Faculty
of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Straße 4a, D-44227 Dortmund, Germany
| | - Jasmin Krüll
- Faculty
of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Straße 4a, D-44227 Dortmund, Germany
| | - Federico Medda
- Taros Chemicals GmbH & Co. KG, Emil-Figge-Straße 76a, D-44227 Dortmund, Germany
| | - Oliver Koch
- Faculty
of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Straße 4a, D-44227 Dortmund, Germany
| | | | - Sebastian Bauer
- Department
of Medical Oncology, Sarcoma Centre West German Cancer Centre University Duisburg−Essen, Medical School, Hufelandstraße 55, D-45122 Essen, Germany
- Germany
and German Cancer Consortium (DKTK), Partner Site University Hospital Essen, D-45147 Essen, Germany
| | - Daniel Rauh
- Faculty
of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Straße 4a, D-44227 Dortmund, Germany
| |
Collapse
|
36
|
Jeong W, Jung S, Bazer FW, Kim J. Stem cell factor-induced AKT cell signaling pathway: Effects on porcine trophectoderm and uterine luminal epithelial cells. Gen Comp Endocrinol 2017; 250:113-121. [PMID: 28551414 DOI: 10.1016/j.ygcen.2017.05.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/19/2017] [Accepted: 05/23/2017] [Indexed: 01/25/2023]
Abstract
Stem cell factor (SCF) is a multipotent growth factor that elicits diverse biological actions in various aspects of embryogenesis and animal development. The aim of the present study was to assess SCF-induced intracellular signaling and cellular activities in porcine trophectoderm (pTr) and uterine luminal epithelial (pLE) cells which are well known as useful to elucidate developmental events. SCF induced abundances of p-AKT, p-P70RSK and RPS6 proteins in pTr cells reached to their maximum, and then returned to basal levels by 120min. In pLE cells, SCF induced protracted effect to increase AKT phosphorylation which was well correlated with the time course for P70RSK and RPS6 phosphorylation. LY294002 (an inhibitor of AKT) decreased SCF-induced p-AKT, p-P70RSK and p-RPS6 proteins. Also, immunofluorescence analyses revealed that p-RPS6 was abundant within the cytoplasm of SCF-treated cells, but p-RPS6 was present only at basal levels in cells treated with LY294002. In the presence of LY294002, both SCF-stimulated transient and sustained AKT phosphorylation were inhibited in pLE cells. Furthermore, SCF increased migration of pTr and pLE cells, but LY294002 significantly reduced this effect of SCF. In conclusion, results of the present study suggest that SCF secreted by the endometrium induces autocrine/paracrine signaling responses that stimulate migration of pTr and pLE cells through activation of the AKT cell signaling pathway. Those results support the hypothesis that SCF is a critical regulatory factor for conceptus development and implantation during pregnancy in pigs.
Collapse
Affiliation(s)
- Wooyoung Jeong
- Department of Animal Resources Science, Dankook University, Cheonan, Republic of Korea
| | - Seoungo Jung
- Department of Animal Resources Science, Dankook University, Cheonan, Republic of Korea
| | - Fuller W Bazer
- Center for Animal Biotechnology and Genomics and Department of Animal Science, Texas A&M University, College Station 77843-2471, TX, USA
| | - Jinyoung Kim
- Department of Animal Resources Science, Dankook University, Cheonan, Republic of Korea.
| |
Collapse
|
37
|
Embryonic exposure to the widely-used herbicide atrazine disrupts meiosis and normal follicle formation in female mice. Sci Rep 2017; 7:3526. [PMID: 28615648 PMCID: PMC5471253 DOI: 10.1038/s41598-017-03738-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/03/2017] [Indexed: 01/28/2023] Open
Abstract
The widely-used herbicide atrazine (ATZ) is detected in ground and surface water in many countries. Several studies in animals have demonstrated that ATZ has endocrine-disrupting effects on male and female reproduction in many vertebrate species. In this study, we investigated the effects of ATZ exposure on meiosis, a key step in gametogenesis in mammals. The treatment was initiated before oocyte entry into meiosis, which occurs during the embryonic period in females. We found that embryonic exposure to ATZ increases the level of 8-oxo-guanine in the nucleus of meiotic cells, reflecting oxidative stress and affecting meiotic double-strand break repair, chromosome synapsis and crossover numbers. Finally, embryonic exposure to ATZ reduces the number of primordial follicles and increases the incidence of multi-oocyte follicles in adult mice. Our data demonstrate that embryonic exposure to ATZ disrupts prophase I of meiosis and affects normal follicle formation in female mice.
Collapse
|
38
|
Singh J, Shah R, Singh D. Targeting mast cells: Uncovering prolific therapeutic role in myriad diseases. Int Immunopharmacol 2016; 40:362-384. [PMID: 27694038 DOI: 10.1016/j.intimp.2016.09.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 09/16/2016] [Accepted: 09/22/2016] [Indexed: 01/08/2023]
Abstract
The mast cells are integral part of immune system and they have pleiotropic physiological functions in our body. Any type of abnormal stimuli causes the mast cells receptors to spur the otherwise innocuous mast cells to degranulate and release inflammatory mediators like histamine, cytokines, chemokines and prostaglandins. These mediators are involved in various diseases like allergy, asthma, mastocytosis, cardiovascular disorders, etc. Herein, we describe the receptors involved in degranulation of mast cells and are broadly divided into four categories: G-protein coupled receptors, ligand gated ion channels, immunoreceptors and pattern recognition receptors. Although, activation of pattern recognition receptors do not cause mast cell degranulation, but result in cytokines production. Degranulation itself is a complex process involving cascade of events like membrane fusion events and various proteins like VAMP, Syntaxins, DOCK5, SNAP-23, MARCKS. Furthermore, we described these mast cell receptors antagonists or agonists useful in treatment of myriad diseases. Like, omalizumab anti-IgE antibody is highly effective in asthma, allergic disorders treatment and recently mechanistic insight of IgE uncovered; matrix mettaloprotease inhibitor marimistat is under phase III trial for inflammation, muscular dystrophy diseases; ZPL-389 (H4 receptor antagonist) is in Phase 2a Clinical Trial for atopic dermatitis and psoriasis; JNJ3851868 an oral H4 receptor antagonist is in phase II clinical development for asthma, rheumatoid arthritis. Therefore, research is still in inchoate stage to uncover mast cell biology, mast cell receptors, their therapeutic role in myriad diseases.
Collapse
Affiliation(s)
- Jatinder Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, Punjab, India
| | - Ramanpreet Shah
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, Punjab, India
| | - Dhandeep Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, Punjab, India.
| |
Collapse
|
39
|
Li B, Wang A, Liu J, Qi Z, Liu X, Yu K, Wu H, Chen C, Hu C, Wang W, Wu J, Hu Z, Ye L, Zou F, Liu F, Wang B, Wang L, Ren T, Zhang S, Bai M, Zhang S, Liu J, Liu Q. Discovery of N-((1-(4-(3-(3-((6,7-Dimethoxyquinolin-3-yl)oxy)phenyl)ureido)-2-(trifluoromethyl)phenyl)piperidin-4-yl)methyl)propionamide (CHMFL-KIT-8140) as a Highly Potent Type II Inhibitor Capable of Inhibiting the T670I “Gatekeeper” Mutant of cKIT Kinase. J Med Chem 2016; 59:8456-72. [DOI: 10.1021/acs.jmedchem.6b00902] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Binhua Li
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- CHMFL-HCMTC Target Therapy Joint Laboratory, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
| | - Aoli Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- University of Science and Technology of China, Hefei, Anhui 230036, P. R. China
| | - Juan Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
| | - Ziping Qi
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- CHMFL-HCMTC Target Therapy Joint Laboratory, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
| | - Xiaochuan Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
| | - Kailin Yu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- University of Science and Technology of China, Hefei, Anhui 230036, P. R. China
| | - Hong Wu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
| | - Cheng Chen
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- CHMFL-HCMTC Target Therapy Joint Laboratory, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
| | - Chen Hu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- University of Science and Technology of China, Hefei, Anhui 230036, P. R. China
| | - Wenchao Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- CHMFL-HCMTC Target Therapy Joint Laboratory, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
| | - Jiaxin Wu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- University of Science and Technology of China, Hefei, Anhui 230036, P. R. China
| | - Zhenquan Hu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- CHMFL-HCMTC Target Therapy Joint Laboratory, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
| | - Ling Ye
- Precision
Targeted Therapy Discovery Center, Institute of Technology Innovation, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230088, P. R. China
| | - Fengming Zou
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- CHMFL-HCMTC Target Therapy Joint Laboratory, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
| | - Feiyang Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- University of Science and Technology of China, Hefei, Anhui 230036, P. R. China
| | - Beilei Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- CHMFL-HCMTC Target Therapy Joint Laboratory, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
| | - Li Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- CHMFL-HCMTC Target Therapy Joint Laboratory, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
| | - Tao Ren
- Precision
Targeted Therapy Discovery Center, Institute of Technology Innovation, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230088, P. R. China
| | - Shaojuan Zhang
- Molecular
Imaging Laboratory, Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15232, United States
| | - Mingfeng Bai
- Molecular
Imaging Laboratory, Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15232, United States
| | - Shanchun Zhang
- CHMFL-HCMTC Target Therapy Joint Laboratory, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- Hefei Cosource Medicine Technology Co. LTD., 358 Ganquan Road, Hefei, Anhui 230031, P. R. China
| | - Jing Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- CHMFL-HCMTC Target Therapy Joint Laboratory, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
| | - Qingsong Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Mailbox 1110, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- CHMFL-HCMTC Target Therapy Joint Laboratory, 350 Shushanhu Road, Hefei, Anhui 230031, P. R. China
- University of Science and Technology of China, Hefei, Anhui 230036, P. R. China
- Precision
Targeted Therapy Discovery Center, Institute of Technology Innovation, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230088, P. R. China
| |
Collapse
|
40
|
Qin S, Wang X, Wu H, Xiao P, Cheng H, Zhang X, Ke Y. Cell-based phenotypic screening of mast cell degranulation unveils kinetic perturbations of agents targeting phosphorylation. Sci Rep 2016; 6:31320. [PMID: 27502076 PMCID: PMC4977535 DOI: 10.1038/srep31320] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/18/2016] [Indexed: 02/08/2023] Open
Abstract
Mast cells play an essential role in initiating allergic diseases. The activation of mast cells are controlled by a complicated signal network of reversible phosphorylation, and finding the key regulators involved in this network has been the focus of the pharmaceutical industry. In this work, we used a method named Time-dependent cell responding profile (TCRP) to track the process of mast cell degranulation under various perturbations caused by agents targeting phosphorylation. To test the feasibility of this high-throughput cell-based phenotypic screening method, a variety of biological techniques were used. We further screened 145 inhibitors and clustered them based on the similarities of their TCRPs. Stat3 phosphorylation has been widely reported as a key step in mast cell degranulation. Interestingly, our TCRP results showed that a Stat3 inhibitor JSI124 did not inhibit degranulation like other Stat3 inhibitors, such as Stattic, clearly inhibited degranulation. Regular endpoint assays demonstrated that the distinctive TCRP of JSI124 potentially correlated with the ability to induce apoptosis. Consequently, different agents possibly have disparate functions, which can be conveniently detected by TCRP. From this perspective, our TCRP screening method is reliable and sensitive when it comes to discovering and selecting novel compounds for new drug developments.
Collapse
Affiliation(s)
- Shenlu Qin
- Program in Molecular Cell Biology, Department of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xumeng Wang
- Program in Molecular Cell Biology, Department of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Huanwen Wu
- Program in Molecular Cell Biology, Department of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Peng Xiao
- Program in Molecular Cell Biology, Department of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hongqiang Cheng
- Program in Molecular Cell Biology, Department of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xue Zhang
- Program in Molecular Cell Biology, Department of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yuehai Ke
- Program in Molecular Cell Biology, Department of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
41
|
Grünwald B, Vandooren J, Gerg M, Ahomaa K, Hunger A, Berchtold S, Akbareian S, Schaten S, Knolle P, Edwards DR, Opdenakker G, Krüger A. Systemic Ablation of MMP-9 Triggers Invasive Growth and Metastasis of Pancreatic Cancer via Deregulation of IL6 Expression in the Bone Marrow. Mol Cancer Res 2016; 14:1147-1158. [PMID: 27489361 DOI: 10.1158/1541-7786.mcr-16-0180] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 06/24/2016] [Indexed: 02/06/2023]
Abstract
Matrix metalloproteinase 9 (MMP-9/Gelatinase B) is overexpressed in pancreatic ductal adenocarcinoma (PDAC) and plays a central role in tumor cell invasion and metastasis. Here we complemented mechanistic insights in the cancer biology of MMP-9 and investigated the effects of specific long-term loss-of-function, by genetic ablation, of MMP-9 on PDAC initiation and progression in the well-established KPC mouse model of spontaneous PDAC. Tumor growth and progression were analyzed by histopathology and IHC. Invasive growth of PDAC cells was analyzed by both in vitro (proliferation, survival, migration, invasion assays) and in vivo (experimental metastasis assays) methods. Retroviral shRNAi was used to knockdown target genes (MMP-9, IL6R). Gene expression was analyzed by qRT-PCR, immunoblot, ELISA, in situ hybridization, and zymography. PDAC tumors from MMP-9-deficient mice were dramatically larger, more invasive, and contained more stroma. Yet, ablation of MMP-9 in PDAC cells did not directly promote invasive growth. Interestingly, systemic ablation of MMP-9 led to increased IL6 levels resulting from abrogation of MMP-9-dependent SCF signaling in the bone marrow. IL6 levels in MMP-9-/- mice were sufficient to induce invasive growth and STAT3 activation in PDAC cells via IL6 receptor (IL6R). Interference with IL6R blocked the increased invasion and metastasis of PDAC cells in MMP-9-deficient hosts. In conclusion, ablation of systemic MMP-9 initiated fatal communication between maintenance of physiological functions of MMP-9 in the bone marrow and invasive growth of PDAC via the IL6/IL6R/STAT3 axis. IMPLICATIONS Thus, the beneficial effects of host MMP-9 on PDAC are an important caveat for the use of systemic MMP-9 inhibitors in cancer. Mol Cancer Res; 14(11); 1147-58. ©2016 AACR.
Collapse
Affiliation(s)
- Barbara Grünwald
- Klinikum rechts der Isar der Technischen Universität München, Institut für Molekulare Immunologie und Experimentelle Onkologie, München, Germany
| | - Jennifer Vandooren
- KU Leuven, Rega Institute for Medical Research, Department of Microbiology and Immunology, Leuven, Belgium
| | - Michael Gerg
- Klinikum rechts der Isar der Technischen Universität München, Institut für Molekulare Immunologie und Experimentelle Onkologie, München, Germany
| | - Kaarin Ahomaa
- Klinikum rechts der Isar der Technischen Universität München, Institut für Molekulare Immunologie und Experimentelle Onkologie, München, Germany
| | - Annique Hunger
- Klinikum rechts der Isar der Technischen Universität München, Institut für Molekulare Immunologie und Experimentelle Onkologie, München, Germany
| | - Sonja Berchtold
- Klinikum rechts der Isar der Technischen Universität München, Institut für Molekulare Immunologie und Experimentelle Onkologie, München, Germany
| | - Sophia Akbareian
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, United Kingdom
| | - Susanne Schaten
- Klinikum rechts der Isar der Technischen Universität München, Institut für Molekulare Immunologie und Experimentelle Onkologie, München, Germany
| | - Percy Knolle
- Klinikum rechts der Isar der Technischen Universität München, Institut für Molekulare Immunologie und Experimentelle Onkologie, München, Germany
| | - Dylan R Edwards
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, United Kingdom
| | - Ghislain Opdenakker
- KU Leuven, Rega Institute for Medical Research, Department of Microbiology and Immunology, Leuven, Belgium
| | - Achim Krüger
- Klinikum rechts der Isar der Technischen Universität München, Institut für Molekulare Immunologie und Experimentelle Onkologie, München, Germany.
| |
Collapse
|
42
|
Liang L, Yan XE, Yin Y, Yun CH. Structural and biochemical studies of the PDGFRA kinase domain. Biochem Biophys Res Commun 2016; 477:667-672. [PMID: 27349873 DOI: 10.1016/j.bbrc.2016.06.117] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/10/2016] [Accepted: 06/23/2016] [Indexed: 12/28/2022]
Abstract
Platelet-derived growth factor receptor α (PDGFRA) is a Type III receptor tyrosine kinase, and this kinase is a target for treatment of gastrointestinal stromal tumors (GIST) as it is frequently mutated in these cancers. Most of the mutations that cause constitutive activation of PDGFRA occur in either the activation loop (A-loop) or in the juxtamembrane (JM) domain, such as the mutations D842V or V561D respectively. Treatment of PDGFRA-mutated GIST with imatinib is successful in some cases, but the D842V mutation is imatinib-resistant. To better understand the mechanism of PDGFRA drug-resistance, we have determined the crystal structure of the PDGFRA kinase domain in the auto-inhibited form, and studied the kinetics of the D842V mutation. Auto-inhibited PDGFRA is stabilized by the JM domain, which inserts into the active site of the kinase. The conserved residue Asp842 makes extensive contacts with several A-loop residues to maintain PDGFRA in the "DFG out" conformation, which stabilizes the kinase in the inactive state and facilitates the binding of imatinib. The D842V mutation would therefore be expected to activate the kinase and hinder the binding of drug through destabilizing the "DFG out" conformation. Furthermore, our kinetic data show that drug resistance in the D842V mutation may also in part result from its increased affinity for ATP. The PDGFRA kinase domain structure we report in this study has potential to facilitate development of new agents which can inhibit this kinase, including both its activating and drug-resistant mutations.
Collapse
Affiliation(s)
- Ling Liang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China; Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China
| | - Xiao-E Yan
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China; Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China
| | - Yuxin Yin
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China; Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China; Peking-Tsinghua Center for Life Sciences, Beijing 100871, PR China.
| | - Cai-Hong Yun
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China; Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China.
| |
Collapse
|
43
|
Gao J, Swaminathan S, Pai N, Johnson Z, Chen YH, Peterson L, Goolsby C. Flow cytometric detection of altered signaling in myelodysplastic syndrome and cytopenia. Leuk Res 2015; 39:1396-404. [PMID: 26410459 DOI: 10.1016/j.leukres.2015.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 08/17/2015] [Accepted: 09/06/2015] [Indexed: 10/23/2022]
Abstract
Multiparameter flow cytometric analysis allows for precise evaluation of growth factor stimulated intracellular signaling in distinct immunophenotype defined hematopoetic populations. Our analysis of intracellular phosphoprotein in response to major hematopoietic growth factors or cytokines showed several interesting findings. Although there was no characteristic signaling abnormality that was diagnostic for MDS, MDS cases were often associated with more signaling aberrancies involving more cellular populations. Higher than average response in the CD34(+)CD117(+) progenitor cells to Flt3 ligand and stem cell factor stimulation was frequently associated with high risk features or disease progression in MDS. Although preliminary results hint an adverse prognostic role of dysregulated FLT3 pathway in MDS cases, whether this observation adds independent prognostic value to the existing prognostic system needs to be further explored in future prospective studies.
Collapse
Affiliation(s)
- Juehua Gao
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Feinberg 7-209A, 251 E. Huron Street, Chicago, IL 60611, USA.
| | - Suchitra Swaminathan
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Feinberg 7-209A, 251 E. Huron Street, Chicago, IL 60611, USA
| | - Navin Pai
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Feinberg 7-209A, 251 E. Huron Street, Chicago, IL 60611, USA
| | - Zachary Johnson
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Feinberg 7-209A, 251 E. Huron Street, Chicago, IL 60611, USA
| | - Yi-Hua Chen
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Feinberg 7-209A, 251 E. Huron Street, Chicago, IL 60611, USA
| | - LoAnn Peterson
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Feinberg 7-209A, 251 E. Huron Street, Chicago, IL 60611, USA
| | - Charles Goolsby
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Feinberg 7-209A, 251 E. Huron Street, Chicago, IL 60611, USA
| |
Collapse
|
44
|
Weon JL, Potts PR. The MAGE protein family and cancer. Curr Opin Cell Biol 2015; 37:1-8. [PMID: 26342994 DOI: 10.1016/j.ceb.2015.08.002] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 08/17/2015] [Indexed: 12/21/2022]
Abstract
The Melanoma Antigen Gene (MAGE) protein family is a large, highly conserved group of proteins that share a common MAGE homology domain. Intriguingly, many MAGE proteins are restricted in expression to reproductive tissues, but are aberrantly expressed in a wide variety of cancer types. Originally discovered as antigens on tumor cells and developed as cancer immunotherapy targets, recent literature suggests a more prominent role for MAGEs in driving tumorigenesis. This review will highlight recent developments into the function of MAGEs as oncogenes, their mechanisms of action in regulation of ubiquitin ligases, and outstanding questions in the field.
Collapse
Affiliation(s)
- Jenny L Weon
- Departments of Physiology, Pharmacology, and Biochemistry, UT Southwestern Medical Center, Dallas, TX 75390, United States
| | - Patrick Ryan Potts
- Departments of Physiology, Pharmacology, and Biochemistry, UT Southwestern Medical Center, Dallas, TX 75390, United States.
| |
Collapse
|
45
|
Thys A, Vandenberghe P, Hague P, Klein OD, Erneux C, Vanderwinden JM. Hyperplasia of interstitial cells of cajal in sprouty homolog 4 deficient mice. PLoS One 2015; 10:e0124861. [PMID: 25923139 PMCID: PMC4414615 DOI: 10.1371/journal.pone.0124861] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 03/11/2015] [Indexed: 12/28/2022] Open
Abstract
Gastrointestinal stromal tumors, which are thought to derive from interstitial cells of Cajal or their precursors, often harbor an oncogenic mutation of the KIT receptor tyrosine kinase. Sprouty homolog 4, a known negative regulator of ERK pathway, has been identified in the interstitial cells of Cajal in the KitK641E murine model of gastrointestinal stromal tumors. Sprouty homolog 4 was upregulated both at the mRNA and protein level in these cells, suggesting that Sprouty homolog 4 is downstream of oncogenic KIT activation and potentially engaged in the negative feedback loop of ERK activation in this model. Here, we used KitK641E heterozygous and Sprouty homolog 4 knock out animals to quantify interstitial cells of Cajal in situ, using quantitative immunofluorescence for the receptor tyrosine kinase Kit and for phosphodiesterase 3a (PDE3A). In the antrum of Sprouty homolog 4 knock out mice, hyperplasia of interstitial cells of Cajal was reminiscent of the KitK641E heterozygous mice antrum. Additionally, the density of interstitial cells of Cajal was higher in the colon of adult Sprouty homolog 4 knock out mice than in WT littermates, although hyperplasia seemed more severe in KitK641E heterozygous mice. Functional transit studies also show similarities between Sprouty homolog 4 knock out and KitK641E heterozygous mice, as the total transit time in 9 month old animals was significantly increased in both genotypes compared to WT littermates. We concluded that the lack of Sprouty homolog 4 expression leads to hyperplasia of the interstitial cells of Cajal and is functionally associated with a delayed transit time.
Collapse
Affiliation(s)
- An Thys
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Pierre Vandenberghe
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Perrine Hague
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Ophir D. Klein
- Department of Orofacial Sciences and Program in Craniofacial and Mesenchymal Biology, University of California, San Francisco, California, United States of America
- Department of Pediatrics and Institute for Human genetics, University of California, San Francisco, California, United States of America
| | - Christophe Erneux
- IRIBHM, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Marie Vanderwinden
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
- * E-mail:
| |
Collapse
|
46
|
Abstract
Cancer is a disease of the genome with diverse aetiologies including the accumulation of acquired mutations throughout the genome. There has been a flood of knowledge improving our understanding of the biology and molecular genetics of melanoma, lung and colorectal cancer since the genomics era started. Translation of this knowledge into a better understanding of cell proliferation, survival and apoptosis has produced a paradigm shift in medical oncology enabling gene-based cancer treatment (called personalised or precision medicine). Somatic mutation analysis is crucial for a genomics approach since it can identify driver mutations-the "Achilles' heel" of cancer, and support clinical decision-making through targeted therapy. Nevertheless, the applications of somatic DNA testing in cancer face many challenges such as obtaining comprehensive coverage of the cancer genome with limited DNA being available, and delivering an accurate report in a timely fashion without false-negative and false-positive results. Further advances in DNA technologies and bioinformatics will overcome these issues and maximise opportunities for targeted therapy. Somatic mutation analysis will then become an integral part of cancer management for all malignancies.
Collapse
Affiliation(s)
- Bing Yu
- 1 Department of Medical Genomics, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia ; 2 Sydney Medical School (Central), the University of Sydney, NSW 2006, Australia ; 3 Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Sandra A O'Toole
- 1 Department of Medical Genomics, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia ; 2 Sydney Medical School (Central), the University of Sydney, NSW 2006, Australia ; 3 Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Ronald J Trent
- 1 Department of Medical Genomics, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia ; 2 Sydney Medical School (Central), the University of Sydney, NSW 2006, Australia ; 3 Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| |
Collapse
|
47
|
Ciccone M, Calin GA, Perrotti D. From the Biology of PP2A to the PADs for Therapy of Hematologic Malignancies. Front Oncol 2015; 5:21. [PMID: 25763353 PMCID: PMC4329809 DOI: 10.3389/fonc.2015.00021] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 01/16/2015] [Indexed: 11/13/2022] Open
Abstract
Over the past decades, an emerging role of phosphatases in the pathogenesis of hematologic malignancies and solid tumors has been established. The tumor-suppressor protein phosphatase 2A (PP2A) belongs to the serine-threonine phosphatases family and accounts for the majority of serine-threonine phosphatase activity in eukaryotic cells. Numerous studies have shown that inhibition of PP2A expression and/or function may contribute to leukemogenesis in several hematological malignancies. Likewise, overexpression or aberrant expression of physiologic PP2A inhibitory molecules (e.g., SET and its associated SETBP1 and CIP2A) may turn off PP2A function and participate to leukemic progression. The discovery of PP2A as tumor suppressor has prompted the evaluation of the safety and the efficacy of new compounds, which can restore PP2A activity in leukemic cells. Although further studies are needed to better understand how PP2A acts in the intricate phosphatases/kinases cancer network, the results reviewed herein strongly support the development on new PP2A-activating drugs and the immediate introduction of those available into clinical protocols for leukemia patients refractory or resistant to current available therapies.
Collapse
Affiliation(s)
- Maria Ciccone
- Department of Experimental Therapeutics, MD Anderson Cancer Center, The University of Texas , Houston, TX , USA
| | - George A Calin
- Department of Experimental Therapeutics, MD Anderson Cancer Center, The University of Texas , Houston, TX , USA
| | - Danilo Perrotti
- Department of Medicine, The Greenebaum Cancer Center, University of Maryland School of Medicine , Baltimore, MD , USA
| |
Collapse
|
48
|
Ku M, Wall M, MacKinnon RN, Walkley CR, Purton LE, Tam C, Izon D, Campbell L, Cheng HC, Nandurkar H. Src family kinases and their role in hematological malignancies. Leuk Lymphoma 2015; 56:577-86. [PMID: 24898666 DOI: 10.3109/10428194.2014.907897] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The Src family protein tyrosine kinases (SFKs) are non-receptor intracellular kinases that have important roles in both hematopoiesis and leukemogenesis. The derangement of their expression or activation has been demonstrated to contribute to hematological malignancies. This review first examines the mechanisms of SFK overexpression and hyperactivation, emphasizing the dysregulation of the upstream modulators. Subsequently, the role of SFK up-regulation in the initiation, progression and therapy resistance of many hematological malignancies is also analyzed. The presented evidence endeavors to highlight the influence of SFK up-regulation on an extensive number of hematological malignancies and the need to consider them as candidates in targeted anticancer therapy.
Collapse
Affiliation(s)
- Matthew Ku
- Haematology Department and Victorian Cancer Cytogenetics Service, St Vincent's Hospital , Fitzroy , Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Sharawat SK, Bakhshi R, Vishnubhatla S, Bakhshi S. High receptor tyrosine kinase (FLT3, KIT) transcript versus anti-apoptotic (BCL2) transcript ratio independently predicts inferior outcome in pediatric acute myeloid leukemia. Blood Cells Mol Dis 2015; 54:56-64. [PMID: 25216797 DOI: 10.1016/j.bcmd.2014.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 07/28/2014] [Indexed: 11/25/2022]
Abstract
OBJECTIVE In acute myeloid leukemia (AML), simultaneous expression of proliferative (FLT3, KIT) and anti-apoptotic genes (BCL2) is unknown. The aim of the study was to prospectively evaluate proliferative and anti-apoptotic gene transcripts, their interrelationship and impact on the outcome in pediatric AML patients. METHODS We assessed proliferative and anti-apoptotic gene transcripts by Q-polymerase chain reaction (TaqMan probe) in 64 consecutive pediatric AML patients. Survival data was analyzed by Kaplan-Meier curves followed by log rank test to compare statistical significance between groups. Stepwise multivariable Cox regression method was used to evaluate independent prognostic factors. RESULTS In univariate analysis, transcript ratio of FLT3/BCL2 and FLT3+KIT/BCL2 significantly predicted event free survival (EFS) (<0.01 and <0.01 respectively) and overall survival (OS) (<0.01 and<0.01 respectively). In stepwise Cox-regression model, high white blood cell count and high FLT3+KIT/BCL2 ratio predicted EFS (HR: 2.2 and 2.3); high hemoglobin and high FLT3+KIT/BCL2 ratio predicted OS (HR: 0.45 and 3.85). Prognostic index (PI) was calculated using the hazard coefficient of independent prognostic factors; at 57.3 months, predicted OS of patients with the highest PI of 1.8 was 8% versus 73% for the lowest PI of -0.3. The mean PI of patients who died was 1.8±0.72 versus 0.54±0.70 for those who are alive, P=0.004. CONCLUSIONS This first study showed that individual expression of proliferative and anti-apoptotic transcripts is not as important in AML patients, rather their interrelationship and relative level probably determines the outcome.
Collapse
Affiliation(s)
- Surender Kumar Sharawat
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Radhika Bakhshi
- Department of Biomedical Sciences, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Sreenivas Vishnubhatla
- Department of Biostatistics, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
50
|
da Silva EZM, Jamur MC, Oliver C. Mast cell function: a new vision of an old cell. J Histochem Cytochem 2014; 62:698-738. [PMID: 25062998 PMCID: PMC4230976 DOI: 10.1369/0022155414545334] [Citation(s) in RCA: 416] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/07/2014] [Indexed: 02/06/2023] Open
Abstract
Since first described by Paul Ehrlich in 1878, mast cells have been mostly viewed as effectors of allergy. It has been only in the past two decades that mast cells have gained recognition for their involvement in other physiological and pathological processes. Mast cells have a widespread distribution and are found predominantly at the interface between the host and the external environment. Mast cell maturation, phenotype and function are a direct consequence of the local microenvironment and have a marked influence on their ability to specifically recognize and respond to various stimuli through the release of an array of biologically active mediators. These features enable mast cells to act as both first responders in harmful situations as well as to respond to changes in their environment by communicating with a variety of other cells implicated in physiological and immunological responses. Therefore, the critical role of mast cells in both innate and adaptive immunity, including immune tolerance, has gained increased prominence. Conversely, mast cell dysfunction has pointed to these cells as the main offenders in several chronic allergic/inflammatory disorders, cancer and autoimmune diseases. This review summarizes the current knowledge of mast cell function in both normal and pathological conditions with regards to their regulation, phenotype and role.
Collapse
Affiliation(s)
- Elaine Zayas Marcelino da Silva
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil (EZMDS, MCJ, CO)
| | - Maria Célia Jamur
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil (EZMDS, MCJ, CO)
| | - Constance Oliver
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil (EZMDS, MCJ, CO)
| |
Collapse
|