1
|
Song ML, Sun YY, Yin HJ, Li Y, Yang H. p-Coumaric acid alleviates neuronal damage in ischemic stroke mice by promoting BACH1 nuclear export and degradation. Acta Pharmacol Sin 2025:10.1038/s41401-025-01510-0. [PMID: 40087473 DOI: 10.1038/s41401-025-01510-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 03/17/2025]
Abstract
Oxidative damage induced by glutamate triggers neuronal death in cerebral ischemic/reperfusion injury. BTB and CNC homology 1 (BACH1) is a major link between the cellular heme level, the redox state and the transcriptional response. p-Coumaric acid (p-CA) is a natural antioxidant that has been shown to ameliorate ischemic/reperfusion injury. In this study, we investigated whether and how p-CA regulated BACH1 in ischemic/reperfusion injury from the perspective of BACH1 subcellular localization and function. Middle cerebral artery occlusion (MCAO) model was established in male mice. MCAO mice were treated with p-CA (50, 100 mg/kg, ip) twice 5 min after MCAO and 5 h after reperfusion operation, respectively. We showed that p-CA treatment exerted dramatic neuroprotective effects, which were associated with the inhibition of BACH1. In HT22 cells, treatment with p-CA (20 μM) ameliorated OGD/R or glutamate-induced oxidative damage and mitochondrial dysfunction through decreasing the protein level of BACH1, the beneficial effect of p-CA was blocked by BACH1 overexpression. We demonstrated that BACH1 level was markedly elevated in the nucleus of HT22 cells under glutamate stimulation, and transcriptionally regulated NADPH oxidase 4 (NOX4) expression, thus mediating ROS outbreak. p-CA treatment activated the activated Cdc42-associated kinase 1 (ACK1)/protein kinase B (AKT) cascade to facilitate the phosphorylation of BACH1, augmented its interaction with chromosome region maintenance 1 (CRM1), thereby leading to the export of BACH1 from the nucleus and degradation mediated by heme-oxidized IRP2 ubiquitin ligase-1 (HOIL-1). In accord with this, administration of ACK1 inhibitor AIM-100 (20 mg/kg, ip) 5 min after MCAO significantly attenuated the neuroprotective effects of p-CA in MCAO mice. We concluded that ACK1/AKT/BACH1 axis may serve as a promising therapeutic approach for the management of ischemic stroke, thereby broadening the clinical utility of p-CA.Keywords: ischemic/reperfusion injury; p-Coumaric acid; BACH1; NOX4; ACK1/AKT; AIM-100.
Collapse
Affiliation(s)
- Meng-Lu Song
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yun-Yun Sun
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Hai-Jun Yin
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yi Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Hua Yang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
2
|
Sridaran D, Mahajan NP. ACK1/TNK2 kinase: molecular mechanisms and emerging cancer therapeutics. Trends Pharmacol Sci 2025; 46:62-77. [PMID: 39721828 DOI: 10.1016/j.tips.2024.11.006] [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: 09/23/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024]
Abstract
Activated CDC42-associated kinase 1 (ACK1), encoded by the TNK2 gene, is a cytoplasmic non-receptor tyrosine kinase whose aberrant activation correlates positively with cancer severity. Recent research has revealed the functional relevance of this oncokinase - it is an epigenetic regulator that drives cancer progression in multiple malignancies. Although ACK1 is an attractive target for therapeutic intervention, incomplete knowledge of its diverse signaling mechanisms and the lack of specific inhibitors have challenged its clinical success. We summarize recent breakthroughs in understanding ACK1 regulation and cellular signaling, and shed light on its immunomodulatory role in balancing T cell activation. We provide a comprehensive overview of preclinical, proof-of-concept studies of potent ACK1-targeting small-molecule inhibitors that are expected to enter clinical trials for cancer patients.
Collapse
Affiliation(s)
- Dhivya Sridaran
- Division of Urologic Surgery, Department of Surgery, Washington University at St. Louis, St. Louis, MO 63110, USA
| | - Nupam P Mahajan
- Division of Urologic Surgery, Department of Surgery, Washington University at St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Cancer Research Building, Washington University at St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
3
|
Murata M, Kuwajima H, Tanaka J, Hasegawa N, Yuki R, Saito Y, Nakayama Y. TNK2 Inhibitor (R)-9bMS Causes Polyploidization Through Mitotic Failure by Targeting Aurora B. Cell Biochem Funct 2024; 42:e70022. [PMID: 39639371 DOI: 10.1002/cbf.70022] [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: 05/29/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
TNK2 is a ubiquitously expressed nonreceptor-type tyrosine kinase. TNK2 participates in tumorigenesis, and TNK2 activation has been found in various cancers; therefore, TNK2 is a promising target for cancer chemotherapy. While the TNK2 inhibitor XMD16-5 is highly selective, it inhibits cytokinesis at higher concentrations by targeting Aurora B kinase, a key enzyme for cell division. Cytokinesis failure frequently generates polyploid cells, and the surviving polyploid cells risk leading to cancer development and malignant progression via chromosome instability. In this study, to investigate the possibility that (R)-9bMS, a TNK2 inhibitor structurally related to XMD16-5, drives malignant progression by inducing abnormal cell division, we examined its effects on cell division, Aurora B autophosphorylation, and colony formation. Cell count results showed a reduction in the number of A431, HeLa S3, HCT116, and MCF7 cells upon TNK2 inhibitor treatment. Microscopic observation indicated the formation of multinucleated and nucleus-enlarged cells. An increase in DNA content was confirmed with flow cytometry, which was underpinned by an increased number of centrosomes. Time-lapse imaging revealed mitotic failure, such as mitotic slippage and cytokinesis failure, as a cause of polyploidization. Of note, TNK2 knockdown significantly increased multinucleated cells, but the effect was quite weak, suggesting that TNK2 inhibition may only partially contribute to mitotic failure and polyploidization. Expectedly, Aurora B phosphorylation was reduced by (R)-9bMS like XMD16-5, but not by TNK2 knockdown. Collectively, TNK2 inhibitors (R)-9bMS and XMD16-5 induce polyploidization via mitotic failure caused by the inhibition of Aurora B kinase rather than TNK2. Notably, (R)-9bMS treatment promoted anchorage-independent colony formation, a hallmark of cancer. Our findings suggest that (R)-9bMS at a high concentration risks promoting cancer development or malignant progression. Therefore, caution should be used when using TNK2 inhibitors for cancers where TNK2 activation is not the transforming mutation and higher concentrations of TNK2 inhibitors are required to slow proliferation.
Collapse
Affiliation(s)
- Mayu Murata
- Laboratory of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Hiroki Kuwajima
- Laboratory of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Junna Tanaka
- Laboratory of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Nanami Hasegawa
- Laboratory of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Ryuzaburo Yuki
- Laboratory of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Youhei Saito
- Laboratory of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yuji Nakayama
- Laboratory of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
4
|
Guillet S, Lazarov T, Jordan N, Boisson B, Tello M, Craddock B, Zhou T, Nishi C, Bareja R, Yang H, Rieux-Laucat F, Fregel Lorenzo RI, Dyall SD, Isenberg D, D'Cruz D, Lachmann N, Elemento O, Viale A, Socci ND, Abel L, Nagata S, Huse M, Miller WT, Casanova JL, Geissmann F. ACK1 and BRK non-receptor tyrosine kinase deficiencies are associated with familial systemic lupus and involved in efferocytosis. eLife 2024; 13:RP96085. [PMID: 39570652 PMCID: PMC11581429 DOI: 10.7554/elife.96085] [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] [Indexed: 11/22/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease, the pathophysiology and genetic basis of which are incompletely understood. Using a forward genetic screen in multiplex families with SLE, we identified an association between SLE and compound heterozygous deleterious variants in the non-receptor tyrosine kinases (NRTKs) ACK1 and BRK. Experimental blockade of ACK1 or BRK increased circulating autoantibodies in vivo in mice and exacerbated glomerular IgG deposits in an SLE mouse model. Mechanistically, NRTKs regulate activation, migration, and proliferation of immune cells. We found that the patients' ACK1 and BRK variants impair efferocytosis, the MERTK-mediated anti-inflammatory response to apoptotic cells, in human induced pluripotent stem cell (hiPSC)-derived macrophages, which may contribute to SLE pathogenesis. Overall, our data suggest that ACK1 and BRK deficiencies are associated with human SLE and impair efferocytosis in macrophages.
Collapse
Affiliation(s)
- Stephanie Guillet
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Ecole doctorale Bio Sorbonne Paris Cité, Université Paris Descartes-Sorbonne Paris CitéParisFrance
| | - Tomi Lazarov
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Natasha Jordan
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King’s College London and Louise Coote Lupus Unit, Guy’s and Thomas’ HospitalsLondonUnited Kingdom
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller UniversityNew YorkUnited States
- University of Paris Cité, Imagine InstituteParisFrance
| | - Maria Tello
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Barbara Craddock
- SKI Stem Cell Research Core, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Ting Zhou
- SKI Stem Cell Research Core, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Chihiro Nishi
- Laboratory of Biochemistry & Immunology, World Premier International Immunology Frontier Research Center, Osaka UniversityOsakaJapan
| | - Rohan Bareja
- Cary and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Meyer Cancer Center Weill Cornell Medical CollegeNew YorkUnited States
| | - Hairu Yang
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | | | | | - Sabrina D Dyall
- Department of Biosciences and Ocean Studies, Faculty of Science, University of MauritiusReduitMauritius
| | - David Isenberg
- Bioinformatics Core, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - David D'Cruz
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King’s College London and Louise Coote Lupus Unit, Guy’s and Thomas’ HospitalsLondonUnited Kingdom
| | - Nico Lachmann
- Centre for Rheumatology, Division of Medicine, University College London, The Rayne BuildingLondonUnited Kingdom
| | - Olivier Elemento
- Cary and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Meyer Cancer Center Weill Cornell Medical CollegeNew YorkUnited States
| | - Agnes Viale
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical SchoolHannoverGermany
| | - Nicholas D Socci
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical SchoolHannoverGermany
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller UniversityNew YorkUnited States
- University of Paris Cité, Imagine InstituteParisFrance
| | - Shigekazu Nagata
- Laboratory of Biochemistry & Immunology, World Premier International Immunology Frontier Research Center, Osaka UniversityOsakaJapan
| | - Morgan Huse
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - W Todd Miller
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Department of Physiology and Biophysics, Stony Brook University School of MedicineStony BrookUnited States
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller UniversityNew YorkUnited States
- University of Paris Cité, Imagine InstituteParisFrance
- Howard Hughes Medical InstituteNew YorkUnited States
- Lab of Human Genetics of Infectious Diseases, INSERM, Necker Hospital for Sick ChildrenParisFrance
- Department of Pediatrics, Necker Hospital for Sick ChildrenParisFrance
| | - Frédéric Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King’s College London and Louise Coote Lupus Unit, Guy’s and Thomas’ HospitalsLondonUnited Kingdom
| |
Collapse
|
5
|
Tsai HJ, Chang YF, Hsieh YJ, Wang JD, Wu CC, Ho MY, Cheng JC, Chen DP, Liao HR, Tseng CP. Human Disabled-2 regulates thromboxane A 2 signaling for efficient hemostasis in thrombocytopenia. Nat Commun 2024; 15:9816. [PMID: 39537612 PMCID: PMC11561248 DOI: 10.1038/s41467-024-54093-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Understanding platelet protein functions facilitates better assessment of platelet disorders. Megakaryocyte lineage-restricted human Disabled-2 knock-in (hDAB2-KI) mice are generated to delineate the functions of hDab2, a regulator of platelet function, in the control of bleeding associated with thrombocytopenia. Here we show that hDab2-KI mice with thrombocytopenia display decreased bleeding time when compared to the control mice. hDab2 augments thromboxane A2 (TxA2) mimetic U46619- but not other agonists-stimulated granule secretion, integrin activation, and aggregation at a lower platelet concentration in vitro. Binding of hDab2 to phosphatidic acid (PA) facilitates formation of the PA-hDab2-AKT complex leading to an increase in U46619-stimulated AKT-Ser473 phosphorylation and the first wave of ADP/ATP release. Consistent with these findings, hDab2 expression in platelets from patients with immune thrombocytopenic purpura is positively correlated with U46619-stimulated ATP release, which in turn inversely correlated with their bleeding tendency. hDab2 appears crucial in regulating bleeding severity associated with thrombocytopenia by a functional interplay with ADP/ATP release underlying TxA2 signaling.
Collapse
Affiliation(s)
- Hui-Ju Tsai
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
| | - Ya-Fang Chang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
| | - Ya-Ju Hsieh
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
| | - Jiaan-Der Wang
- Children's Medical Center, Taichung Veterans General Hospital, Taichung, 407, Taiwan, Republic of China
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, 402, Taiwan, Republic of China
| | - Chih-Ching Wu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan, Republic of China
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
| | - Meng-Ying Ho
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
| | - Ju-Chien Cheng
- Department of Medical Laboratory Science and Biotechnology, China Medical University, North District, Taichung, 404, Taiwan, Republic of China
| | - Ding-Ping Chen
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan, Republic of China
| | - Hsiang-Rui Liao
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan, Republic of China
| | - Ching-Ping Tseng
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China.
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China.
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan, Republic of China.
| |
Collapse
|
6
|
Eshaq AM, Flanagan TW, Hassan SY, Al Asheikh SA, Al-Amoudi WA, Santourlidis S, Hassan SL, Alamodi MO, Bendhack ML, Alamodi MO, Haikel Y, Megahed M, Hassan M. Non-Receptor Tyrosine Kinases: Their Structure and Mechanistic Role in Tumor Progression and Resistance. Cancers (Basel) 2024; 16:2754. [PMID: 39123481 PMCID: PMC11311543 DOI: 10.3390/cancers16152754] [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: 05/29/2024] [Revised: 06/30/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Protein tyrosine kinases (PTKs) function as key molecules in the signaling pathways in addition to their impact as a therapeutic target for the treatment of many human diseases, including cancer. PTKs are characterized by their ability to phosphorylate serine, threonine, or tyrosine residues and can thereby rapidly and reversibly alter the function of their protein substrates in the form of significant changes in protein confirmation and affinity for their interaction with protein partners to drive cellular functions under normal and pathological conditions. PTKs are classified into two groups: one of which represents tyrosine kinases, while the other one includes the members of the serine/threonine kinases. The group of tyrosine kinases is subdivided into subgroups: one of them includes the member of receptor tyrosine kinases (RTKs), while the other subgroup includes the member of non-receptor tyrosine kinases (NRTKs). Both these kinase groups function as an "on" or "off" switch in many cellular functions. NRTKs are enzymes which are overexpressed and activated in many cancer types and regulate variable cellular functions in response to extracellular signaling-dependent mechanisms. NRTK-mediated different cellular functions are regulated by kinase-dependent and kinase-independent mechanisms either in the cytoplasm or in the nucleus. Thus, targeting NRTKs is of great interest to improve the treatment strategy of different tumor types. This review deals with the structure and mechanistic role of NRTKs in tumor progression and resistance and their importance as therapeutic targets in tumor therapy.
Collapse
Affiliation(s)
- Abdulaziz M. Eshaq
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, George Washington University, Washington, DC 20052, USA;
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Thomas W. Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA;
| | - Sofie-Yasmin Hassan
- Department of Pharmacy, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sara A. Al Asheikh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Waleed A. Al-Amoudi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Simeon Santourlidis
- Institute of Cell Therapeutics and Diagnostics, University Medical Center of Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sarah-Lilly Hassan
- Department of Chemistry, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Maryam O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Marcelo L. Bendhack
- Department of Urology, Red Cross University Hospital, Positivo University, Rua Mauá 1111, Curitiba 80030-200, Brazil;
| | - Mohammed O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Youssef Haikel
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Mossad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany;
| | - Mohamed Hassan
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
7
|
Chouhan S, Sridaran D, Weimholt C, Luo J, Li T, Hodgson MC, Santos LN, Le Sommer S, Fang B, Koomen JM, Seeliger M, Qu CK, Yart A, Kontaridis MI, Mahajan K, Mahajan NP. SHP2 as a primordial epigenetic enzyme expunges histone H3 pTyr-54 to amend androgen receptor homeostasis. Nat Commun 2024; 15:5629. [PMID: 38965223 PMCID: PMC11224269 DOI: 10.1038/s41467-024-49978-4] [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: 04/05/2023] [Accepted: 06/27/2024] [Indexed: 07/06/2024] Open
Abstract
Mutations that decrease or increase the activity of the tyrosine phosphatase, SHP2 (encoded by PTPN11), promotes developmental disorders and several malignancies by varying phosphatase activity. We uncovered that SHP2 is a distinct class of an epigenetic enzyme; upon phosphorylation by the kinase ACK1/TNK2, pSHP2 was escorted by androgen receptor (AR) to chromatin, erasing hitherto unidentified pY54-H3 (phosphorylation of histones H3 at Tyr54) epigenetic marks to trigger a transcriptional program of AR. Noonan Syndrome with Multiple Lentigines (NSML) patients, SHP2 knock-in mice, and ACK1 knockout mice presented dramatic increase in pY54-H3, leading to loss of AR transcriptome. In contrast, prostate tumors with high pSHP2 and pACK1 activity exhibited progressive downregulation of pY54-H3 levels and higher AR expression that correlated with disease severity. Overall, pSHP2/pY54-H3 signaling acts as a sentinel of AR homeostasis, explaining not only growth retardation, genital abnormalities and infertility among NSML patients, but also significant AR upregulation in prostate cancer patients.
Collapse
Affiliation(s)
- Surbhi Chouhan
- Department of Surgery, Washington University in St Louis, St Louis, MO, 63110, USA
- 6601, Cancer Research Building, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Dhivya Sridaran
- Department of Surgery, Washington University in St Louis, St Louis, MO, 63110, USA
- 6601, Cancer Research Building, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Cody Weimholt
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Jingqin Luo
- Division of Public Health Sciences, Washington University in St Louis, St Louis, MO, 63110, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Tiandao Li
- Bioinformatics Research Core, Center of Regenerative Medicine, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Myles C Hodgson
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY, 13501, USA
| | - Luana N Santos
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY, 13501, USA
| | - Samantha Le Sommer
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY, 13501, USA
| | - Bin Fang
- Moffitt Cancer Center, SRB3, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - John M Koomen
- Moffitt Cancer Center, SRB3, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Markus Seeliger
- Department of Pharmacological Sciences, Stony Brook University Medical School, BST 7-120, Stony Brook, NY, 11794-8651, USA
| | - Cheng-Kui Qu
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Armelle Yart
- UMR 1301-Inserm 5070-CNRS EFS Univ. P. Sabatier, 4bis Ave Hubert Curien, 31100, Toulouse, France
| | - Maria I Kontaridis
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY, 13501, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kiran Mahajan
- Department of Surgery, Washington University in St Louis, St Louis, MO, 63110, USA
- 6601, Cancer Research Building, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Nupam P Mahajan
- Department of Surgery, Washington University in St Louis, St Louis, MO, 63110, USA.
- 6601, Cancer Research Building, Washington University in St Louis, St Louis, MO, 63110, USA.
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, 63110, USA.
| |
Collapse
|
8
|
Guillet S, Lazarov T, Jordan N, Boisson B, Tello M, Craddock B, Zhou T, Nishi C, Bareja R, Yang H, Rieux-Laucat F, Lorenzo RIF, Dyall SD, Isenberg D, D’Cruz D, Lachmann N, Elemento O, Viale A, Socci ND, Abel L, Nagata S, Huse M, Miller WT, Casanova JL, Geissmann F. ACK1 and BRK non-receptor tyrosine kinase deficiencies are associated with familial systemic lupus and involved in efferocytosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.15.24302255. [PMID: 38883731 PMCID: PMC11177913 DOI: 10.1101/2024.02.15.24302255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Systemic Lupus Erythematosus (SLE) is an autoimmune disease, the pathophysiology and genetic basis of which are incompletely understood. Using a forward genetic screen in multiplex families with systemic lupus erythematosus (SLE) we identified an association between SLE and compound heterozygous deleterious variants in the non-receptor tyrosine kinases (NRTKs) ACK1 and BRK. Experimental blockade of ACK1 or BRK increased circulating autoantibodies in vivo in mice and exacerbated glomerular IgG deposits in an SLE mouse model. Mechanistically, non-receptor tyrosine kinases (NRTKs) regulate activation, migration, and proliferation of immune cells. We found that the patients' ACK1 and BRK variants impair efferocytosis, the MERTK-mediated anti-inflammatory response to apoptotic cells, in human induced Pluripotent Stem Cell (hiPSC)-derived macrophages, which may contribute to SLE pathogenesis. Overall, our data suggest that ACK1 and BRK deficiencies are associated with human SLE and impair efferocytosis in macrophages.
Collapse
Affiliation(s)
- Stephanie Guillet
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Ecole doctorale Bio Sorbonne Paris Cité, Université Paris Descartes-Sorbonne Paris Cité.Paris, France
| | - Tomi Lazarov
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of MedicalSciences, New York, New York 10065, USA
| | - Natasha Jordan
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King’s College London and Louise Coote Lupus Unit, Guy’s and Thomas’ Hospitals, London SE1 1UL, UK
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
- University of Paris Cité, Imagine Institute, Paris, France
| | - Maria Tello
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Barbara Craddock
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794-8661
| | - Ting Zhou
- SKI Stem Cell Research Core, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Chihiro Nishi
- Laboratory of Biochemistry & Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871
| | - Rohan Bareja
- Cary and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Meyer Cancer Center Weill Cornell Medical College, New York, New York 10065, USA
| | - Hairu Yang
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | | | | | - Sabrina D. Dyall
- Department of Biosciences and Ocean Studies, Faculty of Science, University of Mauritius, Reduit, Mauritius
| | - David Isenberg
- Centre for Rheumatology, Division of Medicine, University College London, The Rayne Building, University College London
| | - David D’Cruz
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King’s College London and Louise Coote Lupus Unit, Guy’s and Thomas’ Hospitals, London SE1 1UL, UK
| | - Nico Lachmann
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover 30625, Germany
| | - Olivier Elemento
- Cary and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Meyer Cancer Center Weill Cornell Medical College, New York, New York 10065, USA
| | - Agnes Viale
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Nicholas D. Socci
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Bioinformatics Core, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
- University of Paris Cité, Imagine Institute, Paris, France
| | - Shigekazu Nagata
- Laboratory of Biochemistry & Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871
| | - Morgan Huse
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - W. Todd Miller
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794-8661
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
- University of Paris Cité, Imagine Institute, Paris, France
- Howard Hughes Medical Institute, New York, 10065 NY, USA
- Lab of Human Genetics of Infectious Diseases, INSERM, Necker Hospital for Sick Children, Paris, France, EU
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France, EU
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of MedicalSciences, New York, New York 10065, USA
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King’s College London and Louise Coote Lupus Unit, Guy’s and Thomas’ Hospitals, London SE1 1UL, UK
| |
Collapse
|
9
|
Hobsteter AW, Irazoqui AP, Gonzalez A, Picco AS, Rubert AA, Buitrago CG, Lo Fiego MJ, Silbestri GF. Acetylated galactopyranosyl N-heterocyclic monocarbene complexes of Silver(I) as novel anti-proliferative agents in a rhabdomyosarcoma cell line. Bioorg Med Chem 2024; 107:117756. [PMID: 38759255 DOI: 10.1016/j.bmc.2024.117756] [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: 04/15/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
Herein, four silver(I) complexes bearing acetylated d-galactopyranoside-based N-heterocyclic carbene ligands were synthesized and fully characterized by elemental analysis, NMR, and X-ray photoelectron spectroscopy. All complexes were obtained with an anomeric β-configuration and as monocarbene species. In this study, we investigated the biological effects of the silver(I) complexes 2a-d on the human rhabdomyosarcoma cell line, RD. Our results show concentration-dependent effects on cell density, growth inhibition, and activation of key signaling pathways such as Akt 1/2, ERK 1/2, and p38-MAPK, indicating their potential as anticancer agents. Notably, at 35.5 µM, the complexes induced mitochondrial network disruption, as observed with 2b and 2c, whereas with 2a, this disruption was accompanied by nuclear content release. These results provide insight into the utility of carbohydrate incorporated NHC complexes of silver(I) as new agents in cancer therapy.
Collapse
Affiliation(s)
- Ariana W Hobsteter
- INQUISUR, Departamento de Química, Universidad Nacional del Sur (UNS-CONICET), 8000 Bahía Blanca, Argentina
| | - Ana P Irazoqui
- INBIOSUR (UNS-CONICET), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, 8000 Bahía Blanca, Argentina; Comisión de Investigaciones Científicas de la provincia de Buenos Aires (CIC PBA), Argentina.
| | - Agustina Gonzalez
- INBIOSUR (UNS-CONICET), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, 8000 Bahía Blanca, Argentina
| | - Agustín S Picco
- INIFTA, Fac. de Cs. Exactas, Universidad Nacional de La Plata-CONICET, 1900 La Plata, Argentina
| | - Aldo A Rubert
- INIFTA, Fac. de Cs. Exactas, Universidad Nacional de La Plata-CONICET, 1900 La Plata, Argentina
| | - Claudia G Buitrago
- INBIOSUR (UNS-CONICET), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, 8000 Bahía Blanca, Argentina
| | - Marcos J Lo Fiego
- INQUISUR, Departamento de Química, Universidad Nacional del Sur (UNS-CONICET), 8000 Bahía Blanca, Argentina.
| | - Gustavo F Silbestri
- INQUISUR, Departamento de Química, Universidad Nacional del Sur (UNS-CONICET), 8000 Bahía Blanca, Argentina
| |
Collapse
|
10
|
Wang A, Shuai W, Wu C, Pei J, Yang P, Wang X, Li S, Liu J, Wang Y, Wang G, Ouyang L. Design, Synthesis, and Biological Evaluation of Dual Inhibitors of EGFR L858R/T790M/ACK1 to Overcome Osimertinib Resistance in Nonsmall Cell Lung Cancers. J Med Chem 2024; 67:2777-2801. [PMID: 38323982 DOI: 10.1021/acs.jmedchem.3c01934] [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: 02/08/2024]
Abstract
Activation of the alternative pathways and abnormal signaling transduction are frequently observed in third-generation EGFR-TKIs (epidermal growth factor receptor tyrosine kinase inhibitors)-resistant patients. Wherein, hyperphosphorylation of ACK1 contributes to EGFR-TKIs acquired resistance. Dual inhibition of EGFRL858R/T790M and ACK1 might improve therapeutic efficacy and overcome resistance in lung cancers treatment. Here, we identified a EGFRL858R/T790M/ACK1 dual-targeting compound 21a with aminoquinazoline scaffold, which showed excellent inhibitory activities against EGFRL858R/T790M (IC50 = 23 nM) and ACK1 (IC50 = 263 nM). The cocrystal and docking analysis showed that 21a occupied the ATP binding pockets of EGFRL858R/T790M and ACK1. Moreover, 21a showed potent antiproliferative activities against the H1975 cells, MCF-7 cells and osimertinib-resistant cells AZDR. Further, 21a showed significant antitumor effects and good safety in ADZR xenograft-bearing mice. Taken together, 21a was a potent dual inhibitor of EGFRL858R/T790M/ACK1, which is deserved as a potential lead for overcoming acquired resistance to osimertinib during the EGFR-targeted therapy.
Collapse
Affiliation(s)
- Aoxue Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Wen Shuai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Chengyong Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Junping Pei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Panpan Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Xin Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Shutong Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Jiaxi Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Yuxi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| |
Collapse
|
11
|
Balasooriya ER, Madhusanka D, López-Palacios TP, Eastmond RJ, Jayatunge D, Owen JJ, Gashler JS, Egbert CM, Bulathsinghalage C, Liu L, Piccolo SR, Andersen JL. Integrating Clinical Cancer and PTM Proteomics Data Identifies a Mechanism of ACK1 Kinase Activation. Mol Cancer Res 2024; 22:137-151. [PMID: 37847650 PMCID: PMC10831333 DOI: 10.1158/1541-7786.mcr-23-0153] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 08/17/2023] [Accepted: 10/13/2023] [Indexed: 10/19/2023]
Abstract
Beyond the most common oncogenes activated by mutation (mut-drivers), there likely exists a variety of low-frequency mut-drivers, each of which is a possible frontier for targeted therapy. To identify new and understudied mut-drivers, we developed a machine learning (ML) model that integrates curated clinical cancer data and posttranslational modification (PTM) proteomics databases. We applied the approach to 62,746 patient cancers spanning 84 cancer types and predicted 3,964 oncogenic mutations across 1,148 genes, many of which disrupt PTMs of known and unknown function. The list of putative mut-drivers includes established drivers and others with poorly understood roles in cancer. This ML model is available as a web application. As a case study, we focused the approach on nonreceptor tyrosine kinases (NRTK) and found a recurrent mutation in activated CDC42 kinase-1 (ACK1) that disrupts the Mig6 homology region (MHR) and ubiquitin-association (UBA) domains on the ACK1 C-terminus. By studying these domains in cultured cells, we found that disruption of the MHR domain helps activate the kinase while disruption of the UBA increases kinase stability by blocking its lysosomal degradation. This ACK1 mutation is analogous to lymphoma-associated mutations in its sister kinase, TNK1, which also disrupt a C-terminal inhibitory motif and UBA domain. This study establishes a mut-driver discovery tool for the research community and identifies a mechanism of ACK1 hyperactivation shared among ACK family kinases. IMPLICATIONS This research identifies a potentially targetable activating mutation in ACK1 and other possible oncogenic mutations, including PTM-disrupting mutations, for further study.
Collapse
Affiliation(s)
- Eranga R. Balasooriya
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Dept. of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Deshan Madhusanka
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah
| | - Tania P. López-Palacios
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah
| | - Riley J. Eastmond
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
| | - Dasun Jayatunge
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah
| | - Jake J. Owen
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
| | - Jack S. Gashler
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
| | - Christina M. Egbert
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
| | | | - Lu Liu
- Department of Computer Science, North Dakota State University, Fargo, North Dakota
| | | | - Joshua L. Andersen
- The Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
12
|
Sridaran D, Bradshaw E, DeSelm C, Pachynski R, Mahajan K, Mahajan NP. Prostate cancer immunotherapy: Improving clinical outcomes with a multi-pronged approach. Cell Rep Med 2023; 4:101199. [PMID: 37738978 PMCID: PMC10591038 DOI: 10.1016/j.xcrm.2023.101199] [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: 04/26/2023] [Revised: 08/07/2023] [Accepted: 08/25/2023] [Indexed: 09/24/2023]
Abstract
Cancer immunotherapy has gained traction in recent years owing to remarkable tumor clearance in some patients. Despite the notable success of immune checkpoint blockade (ICB) in multiple malignancies, engagement of the immune system for targeted prostate cancer (PCa) therapy is still in its infancy. Multiple factors contribute to limited response, including the heterogeneity of PCa, the cold tumor microenvironment, and a low number of neoantigens. Significant effort is being invested in improving immune-based PCa therapies. This review is a summary of the status of immunotherapy in treating PCa, with a discussion of multiple immune modalities, including vaccines, adoptively transferred T cells, and bispecific T cell engagers, some of which are undergoing clinical trials. In addition, this review also focuses on emerging mechanism-based small-molecule tyrosine kinase inhibitors with immune modulatory properties that, either as single agents or in combination with other immunotherapies, have the potential to improve clinical outcomes.
Collapse
Affiliation(s)
- Dhivya Sridaran
- Division of Urologic Surgery, Department of Surgery, Washington University in St Louis, Cancer Research Building, 660 S. Euclid Avenue, St Louis, MO 63110, USA
| | - Elliot Bradshaw
- Division of Urologic Surgery, Department of Surgery, Washington University in St Louis, Cancer Research Building, 660 S. Euclid Avenue, St Louis, MO 63110, USA
| | - Carl DeSelm
- Bursky Center for Human Immunology and Immunotherapy Programs (CHiiPs), Washington University in St Louis, Cancer Research Building, 660 S. Euclid Avenue, St Louis, MO 63110, USA; Department of Radiation Oncology, Washington University in St Louis, Cancer Research Building, 660 S. Euclid Avenue, St Louis, MO 63110, USA
| | - Russell Pachynski
- Bursky Center for Human Immunology and Immunotherapy Programs (CHiiPs), Washington University in St Louis, Cancer Research Building, 660 S. Euclid Avenue, St Louis, MO 63110, USA; Division of Oncology, Department of Medicine, Washington University in St Louis, Cancer Research Building, 660 S. Euclid Avenue, St Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St Louis, Cancer Research Building, 660 S. Euclid Avenue, St Louis, MO 63110, USA
| | - Kiran Mahajan
- Division of Urologic Surgery, Department of Surgery, Washington University in St Louis, Cancer Research Building, 660 S. Euclid Avenue, St Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St Louis, Cancer Research Building, 660 S. Euclid Avenue, St Louis, MO 63110, USA
| | - Nupam P Mahajan
- Division of Urologic Surgery, Department of Surgery, Washington University in St Louis, Cancer Research Building, 660 S. Euclid Avenue, St Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St Louis, Cancer Research Building, 660 S. Euclid Avenue, St Louis, MO 63110, USA.
| |
Collapse
|
13
|
Burguete MC, Jover-Mengual T, Castelló-Ruiz M, López-Morales MA, Centeno JM, Aliena-Valero A, Alborch E, Torregrosa G, Salom JB. Cerebroprotective Effect of 17β-Estradiol Replacement Therapy in Ovariectomy-Induced Post-Menopausal Rats Subjected to Ischemic Stroke: Role of MAPK/ERK1/2 Pathway and PI3K-Independent Akt Activation. Int J Mol Sci 2023; 24:14303. [PMID: 37762606 PMCID: PMC10531725 DOI: 10.3390/ijms241814303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/11/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Despite the overwhelming advances in the understanding of the pathogenesis of stroke, a devastating disease affecting millions of people worldwide, currently there are only a limited number of effective treatments available. Preclinical and clinical studies show that stroke is a sexually dimorphic disorder, affecting males and females differently. Strong experimental evidence indicates that estrogen may play a role in this difference and that exogenous 17β-estradiol (E2) is neuroprotective against stroke in both male and female rodents. However, the molecular mechanisms by which E2 intervenes in ischemia-induced cell death, revealing these sex differences, remain unclear. The present study was aimed to determine, in female rats, the molecular mechanisms of two well-known pro-survival signaling pathways, MAPK/ERK1/2 and PI3K/Akt, that mediate E2 neuroprotection in response to acute ischemic stroke. E2 pretreatment reduced brain damage and attenuated apoptotic cell death in ovariectomized female rats after an ischemic insult. Moreover, E2 decreased phosphorylation of ERK1/2 and prevented ischemia/reperfusion-induced dephosphorylation of both Akt and the pro-apoptotic protein, BAD. However, MAPK/ERK1/2 inhibitor PD98059, but not the PI3K inhibitor LY294002, attenuated E2 neuroprotection. Thus, these results suggested that E2 pretreatment in ovariectomized female rats modulates MAPK/ERK1/2 and activates Akt independently of PI3K to promote cerebroprotection in ischemic stroke. A better understanding of the mechanisms and the influence of E2 in the female sex paves the way for the design of future successful hormone replacement therapies.
Collapse
Affiliation(s)
- María C. Burguete
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Universitat de València, 46100 Burjassot, Spain; (M.C.B.); (M.C.-R.); (M.A.L.-M.); (J.M.C.); (A.A.-V.); (E.A.); (G.T.); (J.B.S.)
- Departamento de Fisiología, Universitat de València, 46100 Burjassot, Spain
| | - Teresa Jover-Mengual
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Universitat de València, 46100 Burjassot, Spain; (M.C.B.); (M.C.-R.); (M.A.L.-M.); (J.M.C.); (A.A.-V.); (E.A.); (G.T.); (J.B.S.)
- Departamento de Fisiología, Universitat de València, 46100 Burjassot, Spain
| | - María Castelló-Ruiz
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Universitat de València, 46100 Burjassot, Spain; (M.C.B.); (M.C.-R.); (M.A.L.-M.); (J.M.C.); (A.A.-V.); (E.A.); (G.T.); (J.B.S.)
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universitat de València, 46100 Burjassot, Spain
| | - Mikahela A. López-Morales
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Universitat de València, 46100 Burjassot, Spain; (M.C.B.); (M.C.-R.); (M.A.L.-M.); (J.M.C.); (A.A.-V.); (E.A.); (G.T.); (J.B.S.)
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - José M. Centeno
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Universitat de València, 46100 Burjassot, Spain; (M.C.B.); (M.C.-R.); (M.A.L.-M.); (J.M.C.); (A.A.-V.); (E.A.); (G.T.); (J.B.S.)
- Departamento de Fisiología, Universitat de València, 46100 Burjassot, Spain
| | - Alicia Aliena-Valero
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Universitat de València, 46100 Burjassot, Spain; (M.C.B.); (M.C.-R.); (M.A.L.-M.); (J.M.C.); (A.A.-V.); (E.A.); (G.T.); (J.B.S.)
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Enrique Alborch
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Universitat de València, 46100 Burjassot, Spain; (M.C.B.); (M.C.-R.); (M.A.L.-M.); (J.M.C.); (A.A.-V.); (E.A.); (G.T.); (J.B.S.)
- Departamento de Fisiología, Universitat de València, 46100 Burjassot, Spain
| | - Germán Torregrosa
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Universitat de València, 46100 Burjassot, Spain; (M.C.B.); (M.C.-R.); (M.A.L.-M.); (J.M.C.); (A.A.-V.); (E.A.); (G.T.); (J.B.S.)
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Juan B. Salom
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Universitat de València, 46100 Burjassot, Spain; (M.C.B.); (M.C.-R.); (M.A.L.-M.); (J.M.C.); (A.A.-V.); (E.A.); (G.T.); (J.B.S.)
- Departamento de Fisiología, Universitat de València, 46100 Burjassot, Spain
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| |
Collapse
|
14
|
Saygin C, Giordano G, Shimamoto K, Eisfelder B, Thomas-Toth A, Venkataraman G, Ananthanarayanan V, Vincent TL, DuVall A, Patel AA, Chen Y, Tan F, Anthony SP, Chen Y, Shen Y, Odenike O, Teachey DT, Kee BL, LaBelle J, Stock W. Dual Targeting of Apoptotic and Signaling Pathways in T-Lineage Acute Lymphoblastic Leukemia. Clin Cancer Res 2023; 29:3151-3161. [PMID: 37363966 PMCID: PMC10425730 DOI: 10.1158/1078-0432.ccr-23-0415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/08/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023]
Abstract
PURPOSE Relapsed T-acute lymphoblastic leukemia (T-ALL) has limited treatment options. We investigated mechanisms of resistance to BH3 mimetics in T-ALL to develop rational combination strategies. We also looked at the preclinical efficacy of NWP-0476, a novel BCL-2/BCL-xL inhibitor, as single agent and combination therapy in T-ALL. EXPERIMENTAL DESIGN We used BH3 profiling as a predictive tool for BH3 mimetic response in T-ALL. Using isogenic control, venetoclax-resistant (ven-R) and NWP-0476-resistant (NWP-R) cells, phosphokinase array was performed to identify differentially regulated signaling pathways. RESULTS Typical T-ALL cells had increased dependence on BCL-xL, whereas early T-precursor (ETP)-ALL cells had higher BCL-2 dependence for survival. BCL-2/BCL-xL dual inhibitors were effective against both subtypes of T-lineage ALL. A 71-protein human phosphokinase array showed increased LCK activity in ven-R cells, and increased ACK1 activity in ven-R and NWP-R cells. We hypothesized that pre-TCR and ACK1 signaling pathways are drivers of resistance to BCL-2 and BCL-xL inhibition, respectively. First, we silenced LCK gene in T-ALL cell lines, which resulted in increased sensitivity to BCL-2 inhibition. Mechanistically, LCK activated NF-κB pathway and the expression of BCL-xL. Silencing ACK1 gene resulted in increased sensitivity to both BCL-2 and BCL-xL inhibitors. ACK1 signaling upregulated AKT pathway, which inhibited the pro-apoptotic function of BAD. In a T-ALL patient-derived xenograft model, combination of NWP-0476 and dasatinib demonstrated synergy without major organ toxicity. CONCLUSIONS LCK and ACK1 signaling pathways are critical regulators of BH3 mimetic resistance in T-ALL. Combination of BH3 mimetics with tyrosine kinase inhibitors might be effective against relapsed T-ALL.
Collapse
Affiliation(s)
- Caner Saygin
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Giorgia Giordano
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Kathryn Shimamoto
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Bart Eisfelder
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | | | | | | | - Tiffaney L. Vincent
- The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Adam DuVall
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Anand A. Patel
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Yi Chen
- Newave Pharmaceutical Inc., Pleasanton, California
| | - Fenlai Tan
- Newave Pharmaceutical Inc., Pleasanton, California
| | | | - Yu Chen
- Newave Pharmaceutical Inc., Pleasanton, California
| | - Yue Shen
- Newave Pharmaceutical Inc., Pleasanton, California
| | - Olatoyosi Odenike
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - David T. Teachey
- The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Barbara L. Kee
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - James LaBelle
- Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - Wendy Stock
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| |
Collapse
|
15
|
Sawant M, Wilson A, Sridaran D, Mahajan K, O'Conor CJ, Hagemann IS, Luo J, Weimholt C, Li T, Roa JC, Pandey A, Wu X, Mahajan NP. Epigenetic reprogramming of cell cycle genes by ACK1 promotes breast cancer resistance to CDK4/6 inhibitor. Oncogene 2023; 42:2263-2277. [PMID: 37330596 PMCID: PMC10348910 DOI: 10.1038/s41388-023-02747-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 05/16/2023] [Accepted: 06/08/2023] [Indexed: 06/19/2023]
Abstract
Hormone receptor-positive, HER2-negative advanced breast cancers exhibit high sensitivity to CDK4/6 inhibitors such as palbociclib. However, most patients inevitably develop resistance, thus identification of new actionable therapeutic targets to overcome the recurrent disease is an urgent need. Immunohistochemical studies of tissue microarray revealed increased activation of non-receptor tyrosine kinase, ACK1 (also known as TNK2) in most of the breast cancer subtypes, independent of their hormone receptor status. Chromatin immunoprecipitation studies demonstrated that the nuclear target of activated ACK1, pY88-H4 epigenetic marks, were deposited at cell cycle genes, CCNB1, CCNB2 and CDC20, which in turn initiated their efficient transcription. Pharmacological inhibition of ACK1 using its inhibitor, (R)-9b dampened CCNB1, CCNB2 and CDC20 expression, caused G2/M arrest, culminating in regression of palbociclib-resistant breast tumor growth. Further, (R)-9b suppressed expression of CXCR4 receptor, which resulted in significant impairment of metastasis of breast cancer cells to lung. Overall, our pre-clinical data identifies activated ACK1 as an oncogene that epigenetically controls the cell cycle genes governing the G2/M transition in breast cancer cells. ACK1 inhibitor, (R)-9b could be a novel therapeutic option for the breast cancer patients that have developed resistance to CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Mithila Sawant
- Department of Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
- Division of Urologic Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Audrey Wilson
- Department of Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
- Division of Urologic Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Dhivya Sridaran
- Department of Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
- Division of Urologic Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Kiran Mahajan
- Department of Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
- Division of Urologic Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Christopher J O'Conor
- Department of Pathology and Immunology, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Ian S Hagemann
- Department of Pathology and Immunology, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Jingqin Luo
- Siteman Cancer Center, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Cody Weimholt
- Department of Pathology and Immunology, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
- Siteman Cancer Center, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Tiandao Li
- Bioinformatics Research Core, Center of Regenerative Medicine, Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Juan Carlos Roa
- Department of Pathology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xinyan Wu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Nupam P Mahajan
- Department of Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA.
- Division of Urologic Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA.
- Siteman Cancer Center, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA.
| |
Collapse
|
16
|
Li Q, Zhang T, Song P, Tong L, Feng F, Guo J, Zhou Y, Xie H, Lu X. Design, Synthesis, and Evaluation of ( R)-8-((Tetrahydrofuran-2-yl)methyl)pyrido[2,3- d]pyrimidin-7-ones as Novel Selective ACK1 Inhibitors to Combat Acquired Resistance to the Third-Generation EGFR Inhibitor. J Med Chem 2023; 66:6905-6921. [PMID: 37134203 DOI: 10.1021/acs.jmedchem.3c00319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Activated Cdc42-associated kinase 1 (ACK1) alterations have been considered to mediate bypass acquired resistance to the third-generation EGFR inhibitors (ASK120067 and osimertinib) in NSCLC. Despite many efforts to develop ACK1 small molecule inhibitors, no selective inhibitors have entered clinical trials. We used structure-based drug design to obtain a series of (R)-8-((tetrahydrofuran-2-yl)methyl)pyrido [2,3-d]pyrimidin-7-ones as novel selective ACK1 inhibitors. One of the representative compounds, 10zi, potently inhibited ACK1 kinase with an IC50 of 2.1 nM, while sparing SRC kinase (IC50 = 218.7 nM). Further, 10zi displayed good kinome selectivity in a profiling of 468 kinases. In the ASK120067-resistant lung cancer cell line (67R), 10zi dose-dependently inhibited the phosphorylation of ACK1 and downstream AKT pathway and showed a strong synergistic anti-tumor effect in combination with ASK120067 in vitro. Additionally, 10zi also exhibited reasonable PK profiles with an oral bioavailability of 19.8% at the dose of 10 mg/kg, which provided a promising lead for further development of new anticancer drugs.
Collapse
Affiliation(s)
- Qian Li
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China
| | - Tao Zhang
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, #555 Zuchongzhi Road, Shanghai 201203, China
| | - Peiran Song
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Cuiheng New District, Zhongshan 528400, China
| | - Linjiang Tong
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, #555 Zuchongzhi Road, Shanghai 201203, China
| | - Fang Feng
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, #555 Zuchongzhi Road, Shanghai 201203, China
| | - Jing Guo
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China
| | - Yang Zhou
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China
| | - Hua Xie
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, #555 Zuchongzhi Road, Shanghai 201203, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Cuiheng New District, Zhongshan 528400, China
| | - Xiaoyun Lu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Guangzhou 510632, China
| |
Collapse
|
17
|
Kan Y, Paung Y, Kim Y, Seeliger MA, Miller WT. Biochemical Studies of Systemic Lupus Erythematosus-Associated Mutations in Nonreceptor Tyrosine Kinases Ack1 and Brk. Biochemistry 2023; 62:1124-1137. [PMID: 36854171 PMCID: PMC10052838 DOI: 10.1021/acs.biochem.2c00685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Tyrosine kinases (TKs) play essential roles in signaling processes that regulate cell survival, migration, and proliferation. Dysregulation of tyrosine kinases underlies many disorders, including cancer, cardiovascular and developmental diseases, as well as pathologies of the immune system. Ack1 and Brk are nonreceptor tyrosine kinases (NRTKs) best known for their roles in cancer. Here, we have biochemically characterized novel Ack1 and Brk mutations identified in patients with systemic lupus erythematosus (SLE). These mutations are the first SLE-linked polymorphisms found among NRTKs. We show that two of the mutants are catalytically inactive, while the other three have reduced activity. To understand the structural changes associated with the loss-of-function phenotype, we solved the crystal structure of one of the Ack1 kinase mutants, K161Q. Furthermore, two of the mutated residues (Ack1 A156 and K161) critical for catalytic activity are highly conserved among other TKs, and their substitution in other members of the kinase family could have implications in cancer. In contrast to canonical gain-of-function mutations in TKs observed in many cancers, we report loss-of-function mutations in Ack1 and Brk, highlighting the complexity of TK involvement in human diseases.
Collapse
Affiliation(s)
- Yagmur Kan
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, New York 11794-8661, United States
| | - YiTing Paung
- Department of Pharmacology, School of Medicine, Stony Brook University, Stony Brook, New York 11794-8661, United States
| | - Yunyoung Kim
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, New York 11794-8661, United States
| | - Markus A Seeliger
- Department of Pharmacology, School of Medicine, Stony Brook University, Stony Brook, New York 11794-8661, United States
| | - W Todd Miller
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, New York 11794-8661, United States
- Department of Veterans Affairs Medical Center, Northport, New York 11768, United States
| |
Collapse
|
18
|
Kan Y, Paung Y, Seeliger MA, Miller WT. Domain Architecture of the Nonreceptor Tyrosine Kinase Ack1. Cells 2023; 12:900. [PMID: 36980241 PMCID: PMC10047419 DOI: 10.3390/cells12060900] [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: 01/30/2023] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
The nonreceptor tyrosine kinase (NRTK) Ack1 comprises a distinct arrangement of non-catalytic modules. Its SH3 domain has a C-terminal to the kinase domain (SH1), in contrast to the typical SH3-SH2-SH1 layout in NRTKs. The Ack1 is the only protein that shares a region of high homology to the tumor suppressor protein Mig6, a modulator of EGFR. The vertebrate Acks make up the only tyrosine kinase (TK) family known to carry a UBA domain. The GTPase binding and SAM domains are also uncommon in the NRTKs. In addition to being a downstream effector of receptor tyrosine kinases (RTKs) and integrins, Ack1 can act as an epigenetic regulator, modulate the degradation of the epidermal growth factor receptor (EGFR), confer drug resistance, and mediate the progression of hormone-sensitive tumors. In this review, we discuss the domain architecture of Ack1 in relation to other protein kinases that possess such defined regulatory domains.
Collapse
Affiliation(s)
- Yagmur Kan
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - YiTing Paung
- Department of Pharmacology, School of Medicine, Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Markus A. Seeliger
- Department of Pharmacology, School of Medicine, Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - W. Todd Miller
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, NY 11794-8661, USA
- Department of Veterans Affairs Medical Center, Northport, NY 11768-2200, USA
| |
Collapse
|
19
|
Chouhan S, Sawant M, Weimholt C, Luo J, Sprung RW, Terrado M, Mueller DM, Earp HS, Mahajan NP. TNK2/ACK1-mediated phosphorylation of ATP5F1A (ATP synthase F1 subunit alpha) selectively augments survival of prostate cancer while engendering mitochondrial vulnerability. Autophagy 2023; 19:1000-1025. [PMID: 35895804 PMCID: PMC9980697 DOI: 10.1080/15548627.2022.2103961] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 11/02/2022] Open
Abstract
The challenge of rapid macromolecular synthesis enforces the energy-hungry cancer cell mitochondria to switch their metabolic phenotypes, accomplished by activation of oncogenic tyrosine kinases. Precisely how kinase activity is directly exploited by cancer cell mitochondria to meet high-energy demand, remains to be deciphered. Here we show that a non-receptor tyrosine kinase, TNK2/ACK1 (tyrosine kinase non receptor 2), phosphorylated ATP5F1A (ATP synthase F1 subunit alpha) at Tyr243 and Tyr246 (Tyr200 and 203 in the mature protein, respectively) that not only increased the stability of complex V, but also increased mitochondrial energy output in cancer cells. Further, phospho-ATP5F1A (p-Y-ATP5F1A) prevented its binding to its physiological inhibitor, ATP5IF1 (ATP synthase inhibitory factor subunit 1), causing sustained mitochondrial activity to promote cancer cell growth. TNK2 inhibitor, (R)-9b reversed this process and induced mitophagy-based autophagy to mitigate prostate tumor growth while sparing normal prostate cells. Further, depletion of p-Y-ATP5F1A was needed for (R)-9b-mediated mitophagic response and tumor growth. Moreover, Tnk2 transgenic mice displayed increased p-Y-ATP5F1A and loss of mitophagy and exhibited formation of prostatic intraepithelial neoplasia (PINs). Consistent with these data, a marked increase in p-Y-ATP5F1A was seen as prostate cancer progressed to the malignant stage. Overall, this study uncovered the molecular intricacy of tyrosine kinase-mediated mitochondrial energy regulation as a distinct cancer cell mitochondrial vulnerability and provided evidence that TNK2 inhibitors can act as "mitocans" to induce cancer-specific mitophagy.Abbreviations: ATP5F1A: ATP synthase F1 subunit alpha; ATP5IF1: ATP synthase inhibitory factor subunit 1; CRPC: castration-resistant prostate cancer; DNM1L: dynamin 1 like; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; Mdivi-1: mitochondrial division inhibitor 1; Mut-ATP5F1A: Y243,246A mutant of ATP5F1A; OXPHOS: oxidative phosphorylation; PC: prostate cancer; PINK1: PTEN induced kinase 1; p-Y-ATP5F1A: phosphorylated tyrosine 243 and 246 on ATP5F1A; TNK2/ACK1: tyrosine kinase non receptor 2; Ub: ubiquitin; WT: wild type.
Collapse
Affiliation(s)
- Surbhi Chouhan
- Department of Surgery, Cancer Research Building, St. Louis, MO, USA
- Division of Urologic Surgery Washington University, St. Louis, MO, USA
| | - Mithila Sawant
- Department of Surgery, Cancer Research Building, St. Louis, MO, USA
- Division of Urologic Surgery Washington University, St. Louis, MO, USA
| | - Cody Weimholt
- Department of Pathology & Immunology Washington University, St. Louis, MO, USA
| | - Jingqin Luo
- Division of Public Health Sciences, Washington University, St. Louis, MO, USA
| | - Robert W. Sprung
- Department of Surgery, Cancer Research Building, St. Louis, MO, USA
| | - Mailyn Terrado
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University, North Chicago, IL, USA
| | - David M. Mueller
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University, North Chicago, IL, USA
| | - H. Shelton Earp
- Lineberger Comprehensive Cancer Center, Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Nupam P. Mahajan
- Department of Surgery, Cancer Research Building, St. Louis, MO, USA
- Division of Urologic Surgery Washington University, St. Louis, MO, USA
- Siteman Cancer Center Washington University, St. Louis, MO, USA
| |
Collapse
|
20
|
Liao Y, Chan SC, Welsh EA, Fang B, Sun L, Schönbrunn E, Koomen JM, Duckett DR, Haura EB, Monastyrskyi A, Rix U. Chemical Proteomics with Novel Fully Functionalized Fragments and Stringent Target Prioritization Identifies the Glutathione-Dependent Isomerase GSTZ1 as a Lung Cancer Target. ACS Chem Biol 2023; 18:251-264. [PMID: 36630201 PMCID: PMC12097648 DOI: 10.1021/acschembio.2c00587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Photoreactive fragment-like probes have been applied to discover target proteins that constitute novel cellular vulnerabilities and to identify viable chemical hits for drug discovery. Through forming covalent bonds, functionalized probes can achieve stronger target engagement and require less effort for on-target mechanism validation. However, the design of probe libraries, which directly affects the biological target space that is interrogated, and effective target prioritization remain critical challenges of such a chemical proteomic platform. In this study, we designed and synthesized a diverse panel of 20 fragment-based probes containing natural product-based privileged structural motifs for small-molecule lead discovery. These probes were fully functionalized with orthogonal diazirine and alkyne moieties and used for protein crosslinking in live lung cancer cells, target enrichment via "click chemistry," and subsequent target identification through label-free quantitative liquid chromatography-tandem mass spectrometry analysis. Pair-wise comparison with a blunted negative control probe and stringent prioritization via individual cross-comparisons against the entire panel identified glutathione S-transferase zeta 1 (GSTZ1) as a specific and unique target candidate. DepMap database query, RNA interference-based gene silencing, and proteome-wide tyrosine reactivity profiling suggested that GSTZ1 cooperated with different oncogenic alterations by supporting survival signaling in refractory non-small cell lung cancer cells. This finding may form the basis for developing novel GSTZ1 inhibitors to improve the therapeutic efficacy of oncogene-directed targeted drugs. In summary, we designed a novel fragment-based probe panel and developed a target prioritization scheme with improved stringency, which allows for the identification of unique target candidates, such as GSTZ1 in refractory lung cancer.
Collapse
Affiliation(s)
- Yi Liao
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612, United States
| | - Sean Chin Chan
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612, United States
- Cancer Chemical Biology Ph.D. Program, University of South Florida, Tampa, Florida 33620, United States
| | - Eric A. Welsh
- Biostatistics and Bioinformatics Shared Resource, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612, United States
| | - Bin Fang
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612, United States
| | - Luxin Sun
- Chemical Biology Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612, United States
| | - Ernst Schönbrunn
- Department of Drug Discovery and Chemical Biology Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612, United States
| | - John M. Koomen
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612, United States
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida 33620, United States
| | - Derek R. Duckett
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612, United States
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida 33620, United States
| | - Eric B. Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612, United States
| | - Andrii Monastyrskyi
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612, United States
- Department of Oncologic Sciences and Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612, United States
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida 33620, United States
| |
Collapse
|
21
|
Nourse JB, Russell SN, Moniz NA, Peter K, Seyfarth LM, Scott M, Park HA, Caldwell KA, Caldwell GA. Integrated regulation of dopaminergic and epigenetic effectors of neuroprotection in Parkinson's disease models. Proc Natl Acad Sci U S A 2023; 120:e2210712120. [PMID: 36745808 PMCID: PMC9963946 DOI: 10.1073/pnas.2210712120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 01/05/2023] [Indexed: 02/08/2023] Open
Abstract
Whole-exome sequencing of Parkinson's disease (PD) patient DNA identified single-nucleotide polymorphisms (SNPs) in the tyrosine nonreceptor kinase-2 (TNK2) gene. Although this kinase had a previously demonstrated activity in preventing the endocytosis of the dopamine reuptake transporter (DAT), a causal role for TNK2-associated dysfunction in PD remains unresolved. We postulated the dopaminergic neurodegeneration resulting from patient-associated variants in TNK2 were a consequence of aberrant or prolonged TNK2 overactivity, the latter being a failure in TNK2 degradation by an E3 ubiquitin ligase, neuronal precursor cell-expressed developmentally down-regulated-4 (NEDD4). Interestingly, systemic RNA interference protein-3 (SID-3) is the sole TNK2 ortholog in the nematode Caenorhabditis elegans, where it is an established effector of epigenetic gene silencing mediated through the dsRNA-transporter, SID-1. We hypothesized that TNK2/SID-3 represents a node of integrated dopaminergic and epigenetic signaling essential to neuronal homeostasis. Use of a TNK2 inhibitor (AIM-100) or a NEDD4 activator [N-aryl benzimidazole 2 (NAB2)] in bioassays for either dopamine- or dsRNA-uptake into worm dopaminergic neurons revealed that sid-3 mutants displayed robust neuroprotection from 6-hydroxydopamine (6-OHDA) exposures, as did AIM-100 or NAB2-treated wild-type animals. Furthermore, NEDD4 activation by NAB2 in rat primary neurons correlated to a reduction in TNK2 levels and the attenuation of 6-OHDA neurotoxicity. CRISPR-edited nematodes engineered to endogenously express SID-3 variants analogous to TNK2 PD-associated SNPs exhibited enhanced susceptibility to dopaminergic neurodegeneration and circumvented the RNAi resistance characteristic of SID-3 dysfunction. This research exemplifies a molecular etiology for PD whereby dopaminergic and epigenetic signaling are coordinately regulated to confer susceptibility or resilience to neurodegeneration.
Collapse
Affiliation(s)
- J. Brucker Nourse
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL35487
| | - Shannon N. Russell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL35487
| | - Nathan A. Moniz
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL35487
| | - Kylie Peter
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL35487
| | - Lena M. Seyfarth
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL35487
| | - Madison Scott
- Department of Human Nutrition and Hospitality Management, The University of Alabama, Tuscaloosa, AL35487
| | - Han-A Park
- Department of Human Nutrition and Hospitality Management, The University of Alabama, Tuscaloosa, AL35487
- Alabama Research Institute on Aging, The University of Alabama, Tuscaloosa, AL35487
- Center for Convergent Bioscience and Medicine, The University of Alabama, Tuscaloosa, AL35487
| | - Kim A. Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL35487
- Alabama Research Institute on Aging, The University of Alabama, Tuscaloosa, AL35487
- Center for Convergent Bioscience and Medicine, The University of Alabama, Tuscaloosa, AL35487
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center of Excellence for Research in the Basic Biology of Aging, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL35294
| | - Guy A. Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL35487
- Center for Convergent Bioscience and Medicine, The University of Alabama, Tuscaloosa, AL35487
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center of Excellence for Research in the Basic Biology of Aging, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL35294
| |
Collapse
|
22
|
Yachida N, Hoshino F, Murakami C, Ebina M, Miura Y, Sakane F. Saturated fatty acid- and/or monounsaturated fatty acid-containing phosphatidic acids selectively interact with heat shock protein 27. J Biol Chem 2023; 299:103019. [PMID: 36791913 PMCID: PMC10023972 DOI: 10.1016/j.jbc.2023.103019] [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: 08/11/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Diacylglycerol kinase (DGK) α, which is a key enzyme in the progression of cancer and, in contrast, in T-cell activity attenuation, preferentially produces saturated fatty acid (SFA)- and/or monounsaturated fatty acid (MUFA)-containing phosphatidic acids (PAs), such as 16:0/16:0-, 16:0/18:0-, and 16:1/16:1-PA, in melanoma cells. In the present study, we searched for the target proteins of 16:0/16:0-PA in melanoma cells and identified heat shock protein (HSP) 27, which acts as a molecular chaperone and contributes to cancer progression. HSP27 more strongly interacted with PA than other phospholipids, including phosphatidylcholine, phosphatidylserine, phosphatidylglycerol, cardiolipin, phosphatidylinositol, phosphatidylinositol 4-monophosphate, and phosphatidylinositol 4,5-bisphosphate. Moreover, HSP27 is more preferentially bound to SFA- and/or MUFA-containing PAs, including 16:0/16:0- and 16:0/18:1-PAs, than PUFA-containing PAs, including 18:0/20:4- and 18:0/22:6-PA. Furthermore, HSP27 and constitutively active DGKα expressed in COS-7 cells colocalized in a DGK activity-dependent manner. Notably, 16:0/16:0-PA, but not phosphatidylcholine or 16:0/16:0-phosphatidylserine, induced oligomer dissociation of HSP27, which enhances its chaperone activity. Intriguingly, HSP27 protein was barely detectable in Jurkat T cells, while the protein band was intensely detected in AKI melanoma cells. Taken together, these results strongly suggest that SFA- and/or MUFA-containing PAs produced by DGKα selectively target HSP27 and regulate its cancer-progressive function in melanoma cells but not in T cells.
Collapse
Affiliation(s)
- Naoto Yachida
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, Japan
| | - Fumi Hoshino
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, Japan
| | - Chiaki Murakami
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, Japan; Institute for Advanced Academic Research, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, Japan
| | - Masayuki Ebina
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, Japan
| | - Yuri Miura
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, Japan.
| |
Collapse
|
23
|
Identification of Activated Cdc42-Associated Kinase Inhibitors as Potential Anticancer Agents Using Pharmacoinformatic Approaches. Biomolecules 2023; 13:biom13020217. [PMID: 36830587 PMCID: PMC9953130 DOI: 10.3390/biom13020217] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/08/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Activated Cdc42-associated kinase (ACK1) is essential for numerous cellular functions, such as growth, proliferation, and migration. ACK1 signaling occurs through multiple receptor tyrosine kinases; therefore, its inhibition can provide effective antiproliferative effects against multiple human cancers. A number of ACK1-specific inhibitors were designed and discovered in the previous decade, but none have reached the clinic. Potent and selective ACK1 inhibitors are urgently needed. METHODS In the present investigation, the pharmacophore model (PM) was rationally built utilizing two distinct inhibitors coupled with ACK1 crystal structures. The generated PM was utilized to screen the drug-like database generated from the four chemical databases. The binding mode of pharmacophore-mapped compounds was predicted using a molecular docking (MD) study. The selected hit-protein complexes from MD were studied under all-atom molecular dynamics simulations (MDS) for 500 ns. The obtained trajectories were ranked using binding free energy calculations (ΔG kJ/mol) and Gibb's free energy landscape. RESULTS Our results indicate that the three hit compounds displayed higher binding affinity toward ACK1 when compared with the known multi-kinase inhibitor dasatinib. The inter-molecular interactions of Hit1 and Hit3 reveal that compounds form desirable hydrogen bond interactions with gatekeeper T205, hinge region A208, and DFG motif D270. As a result, we anticipate that the proposed scaffolds might help in the design of promising selective ACK1 inhibitors.
Collapse
|
24
|
Zhu J, Cao K, Zhao M, Ma K, Jiang X, Bai Y, Ling X, Ma J. Improvement of ACK1-targeted therapy efficacy in lung adenocarcinoma using chloroquine or bafilomycin A1. Mol Med 2023; 29:6. [PMID: 36647009 PMCID: PMC9843944 DOI: 10.1186/s10020-023-00602-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/08/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Activated Cdc42-associated kinase 1 (ACK1) is a promising druggable target for cancer, but its inhibitors only showed moderate effects in clinical trials. The study aimed to investigate the underlying mechanisms and improve the antitumor efficacy of ACK1 inhibitors. METHODS RNA-seq was performed to determine the downstream pathways of ACK. Using Lasso Cox regression analysis, we built a risk signature with ACK1-related autophagy genes in the lung adenocarcinoma (LUAD) patients from The Cancer Genome Atlas (TCGA) project. The performance of the signature in predicting the tumor immune environment and response to immunotherapy and chemotherapy were assessed in LUAD. CCK8, mRFP-GFP-LC3 assay, western blot, colony formation, wound healing, and transwell migration assays were conducted to evaluate the effects of the ACK1 inhibitor on lung cancer cells. A subcutaneous NSCLC xenograft model was used for in vivo study. RESULTS RNA-seq revealed the regulatory role of ACK1 in autophagy. Furthermore, the risk signature separated LUAD patients into low- and high-risk groups with significantly different prognoses. The two groups displayed different tumor immune environments regarding 28 immune cell subsets. The low-risk groups showed high immune scores, high CTLA4 expression levels, high immunophenoscore, and low DNA mismatch repair capacity, suggesting a better response to immunotherapy. This signature also predicted sensitivity to commonly used chemotherapy and targeted drugs. In vitro, the ACK1 inhibitors (AIM-100 and Dasatinib) appeared to trigger adaptive autophagy-like response to protect lung cancer cells from apoptosis and activated the AMPK/mTOR signaling pathway, partially explaining its moderate antitumor efficacy. However, blocking lysosomal degradation with chloroquine/Bafilamycine A1 or inhibiting AMPK signaling with compound C/shPRKAA1 enhanced the ACK1 inhibitor's cytotoxic effects on lung cancer cells. The efficacy of the combined therapy was also verified using a mouse xenograft model. CONCLUSIONS The resulting signature from ACK1-related autophagy genes robustly predicted survival and drug sensitivity in LUAD. The lysosomal degradation inhibition improved the therapeutic effects of the ACK1 inhibitor, suggesting a potential role for autophagy in therapy evasion.
Collapse
Affiliation(s)
- Jinhong Zhu
- grid.412651.50000 0004 1808 3502Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040 Heilongjiang China
| | - Kui Cao
- grid.412651.50000 0004 1808 3502Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040 Heilongjiang China
| | - Meng Zhao
- grid.412651.50000 0004 1808 3502Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040 Heilongjiang China
| | - Keru Ma
- grid.412651.50000 0004 1808 3502Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040 Heilongjiang China
| | - Xiangyu Jiang
- grid.412651.50000 0004 1808 3502Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040 Heilongjiang China
| | - Yuwen Bai
- grid.412651.50000 0004 1808 3502Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040 Heilongjiang China
| | - Xiaodong Ling
- grid.412651.50000 0004 1808 3502Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040 Heilongjiang China
| | - Jianqun Ma
- grid.412651.50000 0004 1808 3502Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040 Heilongjiang China
| |
Collapse
|
25
|
Peng HH, Yang HC, Rupa D, Yen CH, Chiu YW, Yang WJ, Luo FJ, Yuan TC. ACK1 upregulated the proliferation of head and neck squamous cell carcinoma cells by promoting p27 phosphorylation and degradation. J Cell Commun Signal 2022; 16:567-578. [PMID: 35247157 PMCID: PMC9733751 DOI: 10.1007/s12079-022-00670-6] [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: 05/31/2021] [Accepted: 02/02/2022] [Indexed: 12/24/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a malignancy with a worldwide distribution. Although intensive studies have been made, the underlying oncogenic mechanism of HNSCC requires further investigation. In this study, we examined the oncogenic role of activated Cdc42-associated kinase 1 (ACK1), an oncogenic tyrosine kinase, in regulating the proliferation of HNSCC cells and its underlying molecular mechanism. Results from immunohistochemical studies revealed that ACK1 was highly expressed in HNSCC tumors, with 77% (77/100) of tumors showing a high ACK1 immunoreactivity compared to 40% (8/20) of normal mucosa. Knockdown of ACK1 expression in HNSCC cells resulted in elevated p27 expression, reduced cell proliferation, and G1-phase cell cycle arrest. Rescue of ACK1 expression in the ACK1-knockdown cells suppressed p27 expression and restored cell proliferation. Compared to ACK1-knockdown cells, ACK1-rescued cells exhibited a restored p27 expression after MG132 treatment and showed an elevated level of ubiquitinated p27. Our data further showed that knockdown of ubiquitin ligase Skp2 resulted in elevated p27 expression. Importantly, the expression of p27(WT), p27(Y74F), or p27(Y89F) in ACK1-overexpressed 293T cells or ACK1-rescued SAS cells showed higher levels of tyrosyl-phosphorylated p27 and interaction with ACK1 or Skp2. However, the expression of p27(Y88F) mutant exhibited a relatively low phosphorylation level and barely bound with ACK1 or Skp2, showing a basal interaction as the control cells. These results suggested that ACK1 is highly expressed in HNSCC tumors and functions to promote cell proliferation by the phosphorylation and degradation of p27 in the Skp2-mediated mechanism.
Collapse
Affiliation(s)
- Hsuan-Hsiang Peng
- grid.260567.00000 0000 8964 3950Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien, 974301 Taiwan, Republic of China
| | - Hao-Chin Yang
- grid.260567.00000 0000 8964 3950Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien, 974301 Taiwan, Republic of China
| | - Darius Rupa
- grid.260567.00000 0000 8964 3950Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien, 974301 Taiwan, Republic of China
| | - Chun-Han Yen
- grid.260567.00000 0000 8964 3950Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien, 974301 Taiwan, Republic of China
| | - Ya-Wen Chiu
- grid.260567.00000 0000 8964 3950Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien, 974301 Taiwan, Republic of China
| | - Wei-Jia Yang
- grid.415323.20000 0004 0639 3300Department of Pathology, Mennonite Christian Hospital, Hualien, 970 Taiwan, Republic of China
| | - Fuh-Jinn Luo
- grid.415323.20000 0004 0639 3300Department of Pathology, Mennonite Christian Hospital, Hualien, 970 Taiwan, Republic of China
| | - Ta-Chun Yuan
- grid.260567.00000 0000 8964 3950Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien, 974301 Taiwan, Republic of China
| |
Collapse
|
26
|
Sridaran D, Chouhan S, Mahajan K, Renganathan A, Weimholt C, Bhagwat S, Reimers M, Kim EH, Thakur MK, Saeed MA, Pachynski RK, Seeliger MA, Miller WT, Feng FY, Mahajan NP. Inhibiting ACK1-mediated phosphorylation of C-terminal Src kinase counteracts prostate cancer immune checkpoint blockade resistance. Nat Commun 2022; 13:6929. [PMID: 36376335 PMCID: PMC9663509 DOI: 10.1038/s41467-022-34724-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/04/2022] [Indexed: 11/16/2022] Open
Abstract
Solid tumours are highly refractory to immune checkpoint blockade (ICB) therapies due to the functional impairment of effector T cells and their inefficient trafficking to tumours. T-cell activation is negatively regulated by C-terminal Src kinase (CSK); however, the exact mechanism remains unknown. Here we show that the conserved oncogenic tyrosine kinase Activated CDC42 kinase 1 (ACK1) is able to phosphorylate CSK at Tyrosine 18 (pY18), which enhances CSK function, constraining T-cell activation. Mice deficient in the Tnk2 gene encoding Ack1, are characterized by diminished CSK Y18-phosphorylation and spontaneous activation of CD8+ and CD4+ T cells, resulting in inhibited growth of transplanted ICB-resistant tumours. Furthermore, ICB treatment of castration-resistant prostate cancer (CRPC) patients results in re-activation of ACK1/pY18-CSK signalling, confirming the involvement of this pathway in ICB insensitivity. An ACK1 small-molecule inhibitor, (R)-9b, recapitulates inhibition of ICB-resistant tumours, which provides evidence for ACK1 enzymatic activity playing a pivotal role in generating ICB resistance. Overall, our study identifies an important mechanism of ICB resistance and holds potential for expanding the scope of ICB therapy to tumours that are currently unresponsive.
Collapse
Affiliation(s)
- Dhivya Sridaran
- Department of Surgery, Washington University at St Louis, St Louis, MO, 63110, USA
- Division of Urologic Surgery, Washington University at St Louis, St Louis, MO, 63110, USA
| | - Surbhi Chouhan
- Department of Surgery, Washington University at St Louis, St Louis, MO, 63110, USA
- Division of Urologic Surgery, Washington University at St Louis, St Louis, MO, 63110, USA
| | - Kiran Mahajan
- Department of Surgery, Washington University at St Louis, St Louis, MO, 63110, USA
- Division of Urologic Surgery, Washington University at St Louis, St Louis, MO, 63110, USA
- Siteman Cancer Center, Washington University at St Louis, St Louis, MO, 63110, USA
| | - Arun Renganathan
- Department of Surgery, Washington University at St Louis, St Louis, MO, 63110, USA
- Division of Urologic Surgery, Washington University at St Louis, St Louis, MO, 63110, USA
| | - Cody Weimholt
- Department of Surgery, Washington University at St Louis, St Louis, MO, 63110, USA
- Division of Oncology, Department of Medicine, Washington University at St Louis, St Louis, MO, 63110, USA
- Anatomic and Clinical Pathology, Washington University at St Louis, St Louis, MO, 63110, USA
| | - Shambhavi Bhagwat
- Department of Surgery, Washington University at St Louis, St Louis, MO, 63110, USA
- Division of Urologic Surgery, Washington University at St Louis, St Louis, MO, 63110, USA
| | - Melissa Reimers
- Siteman Cancer Center, Washington University at St Louis, St Louis, MO, 63110, USA
- Division of Oncology, Department of Medicine, Washington University at St Louis, St Louis, MO, 63110, USA
| | - Eric H Kim
- Department of Surgery, Washington University at St Louis, St Louis, MO, 63110, USA
- Division of Urologic Surgery, Washington University at St Louis, St Louis, MO, 63110, USA
| | - Manish K Thakur
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Muhammad A Saeed
- Siteman Cancer Center, Washington University at St Louis, St Louis, MO, 63110, USA
| | - Russell K Pachynski
- Siteman Cancer Center, Washington University at St Louis, St Louis, MO, 63110, USA
- Division of Oncology, Department of Medicine, Washington University at St Louis, St Louis, MO, 63110, USA
| | - Markus A Seeliger
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - W Todd Miller
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
- Department of Veterans Affairs Medical Center, Northport, NY, 11768, USA
| | - Felix Y Feng
- Department of Radiation Oncology, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| | - Nupam P Mahajan
- Department of Surgery, Washington University at St Louis, St Louis, MO, 63110, USA.
- Division of Urologic Surgery, Washington University at St Louis, St Louis, MO, 63110, USA.
- Siteman Cancer Center, Washington University at St Louis, St Louis, MO, 63110, USA.
| |
Collapse
|
27
|
Kan Y, Miller WT. Activity of the nonreceptor tyrosine kinase Ack1 is regulated by tyrosine phosphorylation of its Mig6 homology region. FEBS Lett 2022; 596:2808-2820. [PMID: 36178070 PMCID: PMC9879303 DOI: 10.1002/1873-3468.14505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/19/2022] [Indexed: 01/28/2023]
Abstract
Ack1 is a proto-oncogenic tyrosine kinase with homology to the tumour suppressor Mig6, an inhibitor of the epidermal growth factor receptor (EGFR). The residues critical for binding of Mig6 to EGFR are conserved within the Mig6 homology region (MHR) of Ack1. We tested whether intramolecular interactions between the Ack1 MHR and kinase domain (KD) are regulated by phosphorylation. We identified two Src phosphorylation sites within the MHR (Y859, Y860). Addition of Src-phosphorylated MHR to the Ack1 KD enhanced enzymatic activity. Co-expression of Src in cells led to increased Ack1 activity; mutation of Y859/Y860 blocked this increase. Collectively, the data suggest that phosphorylation of the Ack1 MHR regulates its kinase activity. Phosphorylation of Y859/Y860 occurs in cancers of the brain, breast, colon, and prostate, where genomic amplification or somatic mutations of Ack1 play a role in disease progression. Our findings suggest that MHR phosphorylation could contribute to Ack1 dysregulation in tumours.
Collapse
Affiliation(s)
- Yağmur Kan
- Department of Physiology and Biophysics, School of Medicine Stony Brook University NY USA
| | - W. Todd Miller
- Department of Physiology and Biophysics, School of Medicine Stony Brook University NY USA
| |
Collapse
|
28
|
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the pathogen responsible for the coronavirus disease 2019 (COVID-19) pandemic. Of particular interest for this topic are the signaling cascades that regulate cell survival and death, two opposite cell programs whose control is hijacked by viral infections. The AKT and the Unfolded Protein Response (UPR) pathways, which maintain cell homeostasis by regulating these two programs, have been shown to be deregulated during SARS-CoVs infection as well as in the development of cancer, one of the most important comorbidities in relation to COVID-19. Recent evidence revealed two way crosstalk mechanisms between the AKT and the UPR pathways, suggesting that they might constitute a unified homeostatic control system. Here, we review the role of the AKT and UPR pathways and their interaction in relation to SARS-CoV-2 infection as well as in tumor onset and progression. Feedback regulation between AKT and UPR pathways emerges as a master control mechanism of cell decision making in terms of survival or death and therefore represents a key potential target for developing treatments for both viral infection and cancer. In particular, drug repositioning, the investigation of existing drugs for new therapeutic purposes, could significantly reduce time and costs compared to de novo drug discovery.
Collapse
|
29
|
Shen Y, Jing D, Zhao Z. The effect of AKT in extracellular matrix stiffness induced osteogenic differentiation of hBMSCs. Cell Signal 2022; 99:110404. [PMID: 35835331 DOI: 10.1016/j.cellsig.2022.110404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/02/2022] [Accepted: 07/08/2022] [Indexed: 02/05/2023]
Abstract
Extracellular matrix (ECM) stiffness is an important biophysical factor in human bone marrow mesenchymal stem cells (hBMSCs) differentiation. Although there is evidence that Yes-associated protein (YAP) plays an important role in ECM elasticity induced osteogenesis, but the regulatory mechanism and signaling pathways have not been distinctly uncovered. In this study, hBMSCs were cultured on collagen-coated polydimethylsiloxane hydrogels with stiffness corresponding to Young's moduli of 0.5 kPa and 32 kPa, and gene chip analyses revealed the phosphoinositide 3-kinase (PI3K)-AKT pathway was highly correlated with ECM stiffness. Following western blots indicated that AKT phosphorylation was evidently affected in 5th-7th days after ECM stiffness stimulation, while PI3K showed little difference. The AKT activator SC79 and inhibitor MK2206 were utilized to modulate AKT phosphorylation. SC79 and MK2206 caused alteration in the mRNA expression and protein level of alkaline phosphatase (ALP), collagen type I alpha 1 (COL1A1) and runt related transcription factor 2 (RUNX2). On 32 kPa substrates, YAP enrichment in nucleus were significantly promoted by SC79 and remarkably decreased by MK2206. Besides, the ratio of YAP/p-YAP is upregulated by SC79 on both 32 kPa and 0.5 kPa substrates. In conclusion, these findings suggest that AKT is involved in the modulation of ECM stiffness induced osteogenesis, and AKT phosphorylation also influences the subcellular localization and activation of YAP.
Collapse
Affiliation(s)
- Yu Shen
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China
| | - Dian Jing
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, China.
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
30
|
Sawant M, Mahajan K, Renganathan A, Weimholt C, Luo J, Kukshal V, Jez JM, Jeon MS, Zhang B, Li T, Fang B, Luo Y, Lawrence NJ, Lawrence HR, Feng FY, Mahajan NP. Chronologically modified androgen receptor in recurrent castration-resistant prostate cancer and its therapeutic targeting. Sci Transl Med 2022; 14:eabg4132. [PMID: 35704598 PMCID: PMC10259236 DOI: 10.1126/scitranslmed.abg4132] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Resistance to second-generation androgen receptor (AR) antagonists such as enzalutamide is an inevitable consequence in patients with castration-resistant prostate cancer (CRPC). There are no effective therapeutic options for this recurrent disease. The expression of truncated AR variant 7 (AR-V7) has been suggested to be one mechanism of resistance; however, its low frequency in patients with CRPC does not explain the almost universal acquisition of resistance. We noted that the ability of AR to translocate to nucleus in an enzalutamide-rich environment opens up the possibility of a posttranslational modification in AR that is refractory to enzalutamide binding. Chemical proteomics in enzalutamide-resistant CRPC cells revealed acetylation at Lys609 in the zinc finger DNA binding domain of AR (acK609-AR) that not only allowed AR translocation but also galvanized a distinct global transcription program, conferring enzalutamide insensitivity. Mechanistically, acK609-AR was recruited to the AR and ACK1/TNK2 enhancers, up-regulating their transcription. ACK1 kinase-mediated AR Y267 phosphorylation was a prerequisite for AR K609 acetylation, which spawned positive feedback loops at both the transcriptional and posttranslational level that regenerated and sustained high AR and ACK1 expression. Consistent with these findings, oral and subcutaneous treatment with ACK1 small-molecule inhibitor, (R)-9b, not only curbed AR Y267 phosphorylation and subsequent K609 acetylation but also compromised enzalutamide-resistant CRPC xenograft tumor growth in mice. Overall, these data uncover chronological modification events in AR that allows prostate cancer to evolve through progressive stages to reach the resilient recurrent CRPC stage, opening up a therapeutic vulnerability.
Collapse
Affiliation(s)
- Mithila Sawant
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Division of Urologic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kiran Mahajan
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Division of Urologic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO 63110, USA
| | - Arun Renganathan
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Division of Urologic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Cody Weimholt
- Department of Anatomic and Clinical Pathology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Jingqin Luo
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO 63110, USA
| | - Vandna Kukshal
- Department of Biology, Washington University in St. Louis, One Brookings Drive, St. Louis, MO 63110, USA
| | - Joseph M. Jez
- Department of Biology, Washington University in St. Louis, One Brookings Drive, St. Louis, MO 63110, USA
| | - Myung Sik Jeon
- Siteman Cancer Center, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO 63110, USA
| | - Bo Zhang
- Bioinformatics Research Core, Center of Regenerative Medicine, Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Tiandao Li
- Bioinformatics Research Core, Center of Regenerative Medicine, Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Bin Fang
- Drug Discovery Department, Moffitt Cancer Center, Department of Oncologic Sciences, University of South Florida, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Yunting Luo
- Drug Discovery Department, Moffitt Cancer Center, Department of Oncologic Sciences, University of South Florida, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Nicholas J. Lawrence
- Drug Discovery Department, Moffitt Cancer Center, Department of Oncologic Sciences, University of South Florida, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Harshani R. Lawrence
- Drug Discovery Department, Moffitt Cancer Center, Department of Oncologic Sciences, University of South Florida, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Felix Y. Feng
- Helen Diller Family Cancer Research Building, 1450 Third Street, Room 383, University of California, San Francisco, CA 94158, USA
| | - Nupam P. Mahajan
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Division of Urologic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO 63110, USA
| |
Collapse
|
31
|
Clayton NS, Fox M, Vicenté-Garcia JJ, Schroeder CM, Littlewood TD, Wilde JI, Krishnan K, Brown MJB, Crafter C, Mott HR, Owen D. Assembly of nuclear dimers of PI3K regulatory subunits is regulated by the Cdc42-activated tyrosine kinase ACK. J Biol Chem 2022; 298:101916. [PMID: 35429500 PMCID: PMC9127371 DOI: 10.1016/j.jbc.2022.101916] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 11/25/2022] Open
Abstract
Activated Cdc42-associated kinase (ACK) is an oncogenic nonreceptor tyrosine kinase associated with poor prognosis in several human cancers. ACK promotes proliferation, in part by contributing to the activation of Akt, the major effector of class 1A phosphoinositide 3-kinases (PI3Ks), which transduce signals via membrane phosphoinositol lipids. We now show that ACK also interacts with other key components of class 1A PI3K signaling, the PI3K regulatory subunits. We demonstrate ACK binds to all five PI3K regulatory subunit isoforms and directly phosphorylates p85α, p85β, p50α, and p55α on Tyr607 (or analogous residues). We found that phosphorylation of p85β promotes cell proliferation in HEK293T cells. We demonstrate that ACK interacts with p85α exclusively in nuclear-enriched cell fractions, where p85α phosphorylated at Tyr607 (pTyr607) also resides, and identify an interaction between pTyr607 and the N-terminal SH2 domain that supports dimerization of the regulatory subunits. We infer from this that ACK targets p110-independent p85 and further postulate that these regulatory subunit dimers undertake novel nuclear functions underpinning ACK activity. We conclude that these dimers represent a previously undescribed mode of regulation for the class1A PI3K regulatory subunits and potentially reveal additional avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Natasha S Clayton
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Millie Fox
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | | | | | - Trevor D Littlewood
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Jonathon I Wilde
- GlaxoSmithKline Medicines Research Centre, Screening and Compound Profiling, Stevenage, Herts, United Kingdom
| | - Kadalmani Krishnan
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Murray J B Brown
- GlaxoSmithKline Medicines Research Centre, Screening and Compound Profiling, Stevenage, Herts, United Kingdom
| | - Claire Crafter
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Helen R Mott
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
| | - Darerca Owen
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
32
|
Rangwala AM, Mingione VR, Georghiou G, Seeliger MA. Kinases on Double Duty: A Review of UniProtKB Annotated Bifunctionality within the Kinome. Biomolecules 2022; 12:biom12050685. [PMID: 35625613 PMCID: PMC9138534 DOI: 10.3390/biom12050685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 01/27/2023] Open
Abstract
Phosphorylation facilitates the regulation of all fundamental biological processes, which has triggered extensive research of protein kinases and their roles in human health and disease. In addition to their phosphotransferase activity, certain kinases have evolved to adopt additional catalytic functions, while others have completely lost all catalytic activity. We searched the Universal Protein Resource Knowledgebase (UniProtKB) database for bifunctional protein kinases and focused on kinases that are critical for bacterial and human cellular homeostasis. These kinases engage in diverse functional roles, ranging from environmental sensing and metabolic regulation to immune-host defense and cell cycle control. Herein, we describe their dual catalytic activities and how they contribute to disease pathogenesis.
Collapse
|
33
|
Srivastava R. Chemical Reactivity and Optical and Pharmacokinetics Studies of 14 Multikinase Inhibitors and Their Docking Interactions Toward ACK1 for Precision Oncology. Front Chem 2022; 10:843642. [PMID: 35494626 PMCID: PMC9050413 DOI: 10.3389/fchem.2022.843642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 03/16/2022] [Indexed: 11/23/2022] Open
Abstract
Activated Cdc42-associated kinase 1 (ACK1/TNK2) has a significant role in cell endocytosis, survival, proliferation, and migration. Mutations in ACK1 are closely associated with the occurrence and development of cancers. In this work, a conceptual density functional theory (CDFT)-based computational peptidology (CDFT-CP) method is used to study the chemical reactivity of 14 multikinase inhibitors. Optical properties of these inhibitors are studied by time-dependent density functional theory (TDDFT). Various biological and pharmacokinetic parameters are studied by Osiris, Molinspiration, and BOILED-Egg in SwissADME software tools. Physicochemical and biopharmaceutical (PCB), Salmonella typhimurium reverse mutation assay (AMES) mutagenicity, toxicity, and risk prediction are estimated by Simulations plus ADMET Predictor 10.2 software. MD simulations for an active model of ACK1 is carried out by the CABS-flex 2.0 web server, and potential binding pockets for ACK1 are searched using the PrankWeb server. SwissTargetPrediction is used to predict the potential targets for the multikinase inhibitors. Docking studies are carried out for ACK1–multikinase inhibitors using Autodock 4.2 software. Noncovalent interactions for ACK1–multikinase inhibitor complexes are studied using the Protein–Ligand Interaction Profiler (PLIP) server. Results indicated higher binding affinities and strong noncovalent interactions in ACK1–multikinase inhibitor complexes.
Collapse
|
34
|
Miller KJ, Asim M. Unravelling the Role of Kinases That Underpin Androgen Signalling in Prostate Cancer. Cells 2022; 11:cells11060952. [PMID: 35326402 PMCID: PMC8946764 DOI: 10.3390/cells11060952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023] Open
Abstract
The androgen receptor (AR) signalling pathway is the key driver in most prostate cancers (PCa), and is underpinned by several kinases both upstream and downstream of the AR. Many popular therapies for PCa that target the AR directly, however, have been circumvented by AR mutation, such as androgen receptor variants. Some upstream kinases promote AR signalling, including those which phosphorylate the AR and others that are AR-regulated, and androgen regulated kinase that can also form feed-forward activation circuits to promotes AR function. All of these kinases represent potentially druggable targets for PCa. There has generally been a divide in reviews reporting on pathways upstream of the AR and those reporting on AR-regulated genes despite the overlap that constitutes the promotion of AR signalling and PCa progression. In this review, we aim to elucidate which kinases—both upstream and AR-regulated—may be therapeutic targets and require future investigation and ongoing trials in developing kinase inhibitors for PCa.
Collapse
|
35
|
Anwar MK, Ahmed U, Rehman Z, Fahim A, Jamal SB, Faheem M, Hanif R. Structural and functional characterization of disease-associated NOTCH4: a potential modulator of PI3K/AKT-mediated insulin signaling pathway. APPLIED NANOSCIENCE 2022. [DOI: 10.1007/s13204-021-02281-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
36
|
Kong D, Li G, Yang Z, Cheng S, Zhang W, Feng L, Zhang K. Identification of an ACK1/TNK2-based prognostic signature for colon cancer to predict survival and inflammatory landscapes. BMC Cancer 2022; 22:84. [PMID: 35057760 PMCID: PMC8772074 DOI: 10.1186/s12885-021-09165-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022] Open
Abstract
Activated Cdc42-associated kinase 1 (ACK1), a kind of tyrosine kinase, is considered to be an oncogene in many cancers, and it is likely to become a potential target for cancer treatment. We found that the expression of the ACK1 gene in colon cancer was higher than that in normal tissues adjacent to cancer, and high expression of the ACK1 gene was associated with poor prognosis of patients. We assessed the prognosis of colon cancer based on ACK1-related genes and constructed a model that can predict the prognosis of colon cancer patients in colon cancer data from The Cancer Genome Atlas (TCGA) database. We then explored the relationship between ACK1 and the immune microenvironment of colon cancer. The overexpression of ACK1 might hinder the function of antigen-presenting cells. The colon cancer prognosis prediction model we constructed has certain significance for clinicians to judge the prognosis of patients with colon cancer. The expression of the ACK1 gene might affect the infiltration level of a variety of immune cells and immunomodulators in the immune microenvironment.
Collapse
Affiliation(s)
- Defeng Kong
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, PR China
| | - Guoliang Li
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, PR China
| | - Zhenrong Yang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, PR China
| | - Shujun Cheng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, PR China
| | - Wen Zhang
- Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, PR China.
| | - Lin Feng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, PR China.
| | - Kaitai Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, PR China.
| |
Collapse
|
37
|
Wang A, Pei J, Shuai W, Lin C, Feng L, Wang Y, Lin F, Ouyang L, Wang G. Small Molecules Targeting Activated Cdc42-Associated Kinase 1 (ACK1/TNK2) for the Treatment of Cancers. J Med Chem 2021; 64:16328-16348. [PMID: 34735773 DOI: 10.1021/acs.jmedchem.1c01030] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Activated Cdc42-associated kinase 1 (ACK1/TNK2) is a nonreceptor tyrosine kinase with a unique structure. It not only can act as an activated transmembrane effector of receptor tyrosine kinases (RTKs) to transmit various RTK signals but also can play a corresponding role in epigenetic regulation. A number of studies have shown that ACK1 is a carcinogenic factor. Blockage of ACK1 has been proven to be able to inhibit cancer cell survival, proliferation, migration, and radiation resistance. Thus, ACK1 is a promising potential antitumor target. To date, despite many efforts to develop ACK1 inhibitors, no specific small molecule inhibitors have entered clinical trials. This Perspective provides an overview of the structural features, biological functions, and association with diseases of ACK1 and in vitro and in vivo activities, selectivity, and therapeutic potential of small molecule ACK1 inhibitors with different chemotypes.
Collapse
Affiliation(s)
- Aoxue Wang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Junping Pei
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Wen Shuai
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Congcong Lin
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Lu Feng
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Yuxi Wang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Feng Lin
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China.,Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
38
|
Ghildiyal R, Sawant M, Renganathan A, Mahajan K, Kim EH, Luo J, Dang HX, Maher CA, Feng FY, Mahajan NP. Loss of long non-coding RNA NXTAR in prostate cancer augments androgen receptor expression and enzalutamide resistance. Cancer Res 2021; 82:155-168. [PMID: 34740892 DOI: 10.1158/0008-5472.can-20-3845] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/19/2021] [Accepted: 10/19/2021] [Indexed: 11/16/2022]
Abstract
Androgen receptor (AR) signaling continues to play a dominant role in all stages of prostate cancer (PC), including castration-resistant prostate cancers (CRPC) that have developed resistance to second-generation AR antagonists such as enzalutamide. In this study, we identified a long non-coding RNA (lncRNA), NXTAR (LOC105373241), that is located convergent with the AR gene and is repressed in human prostate tumors and cell lines. NXTAR bound upstream of the AR promoter and promoted EZH2 recruitment, causing significant loss of AR (and AR-V7) expression. Paradoxically, AR bound the NXTAR promoter, and inhibition of AR by the ACK1/TNK2 small molecule inhibitor (R)-9b excluded AR from the NXTAR promoter. The histone acetyltransferase GCN5 bound and deposited H3K14 acetylation marks, enhancing NXTAR expression. Application of an oligonucleotide derived from NXTAR exon 5 (NXTAR-N5) suppressed AR/AR-V7 expression and prostate cancer cell proliferation, indicating the translational relevance of the negative regulation of AR. In addition, pharmacological restoration of NXTAR using (R)-9b abrogated enzalutamide-resistant prostate xenograft tumor growth. Overall, this study uncovers a positive feedback loop, wherein NXTAR acts as a novel prostate tumor-suppressing lncRNA by inhibiting AR/AR-V7 expression, which in turn upregulates NXTAR levels, compromising enzalutamide-resistant prostate cancer. The restoration of NXTAR could serve as a new therapeutic modality for patients who have acquired resistance to second-generation AR antagonists.
Collapse
Affiliation(s)
| | | | | | | | - Eric H Kim
- Siteman Cancer Center, Moffitt Cancer Center
| | - Jingqin Luo
- Division of Public Health Sciences, Department of Surgery, Washington University in St. Louis School of Medicine
| | - Ha X Dang
- Internal Medicine, Washington University in St. Louis
| | | | - Felix Y Feng
- Radiation Oncology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco
| | | |
Collapse
|
39
|
Tahir R, Madugundu AK, Udainiya S, Cutler JA, Renuse S, Wang L, Pearson NA, Mitchell CJ, Mahajan N, Pandey A, Wu X. Proximity-Dependent Biotinylation to Elucidate the Interactome of TNK2 Nonreceptor Tyrosine Kinase. J Proteome Res 2021; 20:4566-4577. [PMID: 34428048 DOI: 10.1021/acs.jproteome.1c00551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Nonreceptor tyrosine kinases (NRTKs) represent an important class of signaling molecules driving diverse cellular pathways. Aberrant expression and hyperphosphorylation of TNK2, an NRTK, have been implicated in multiple cancers. However, the exact proteins and cellular events that mediate phenotypic changes downstream of TNK2 are unclear. Biological systems that employ proximity-dependent biotinylation methods, such as BioID, are being increasingly used to map protein-protein interactions, as they provide increased sensitivity in discovering interaction partners. In this study, we employed stable isotope labeling with amino acids in cell culture and BioID coupled to the biotinylation site identification technology (BioSITe) method that we recently developed to quantitatively explore the interactome of TNK2. By performing a controlled comparative analysis between full-length TNK2 and its truncated counterpart, we were able to not only identify site-level biotinylation of previously well-established TNK2 binders and substrates including NCK1, NCK2, CTTN, and STAT3, but also discover several novel TNK2 interacting partners. We also performed co-immunoprecipitation and immunofluorescence analysis to validate the interaction between TNK2 and CLINT1, a novel TNK2 interacting protein. Overall, this work reveals the power of the BioSITe method coupled to BioID and highlights several molecules that warrant further exploration to assess their functional significance in TNK2-mediated signaling.
Collapse
Affiliation(s)
- Raiha Tahir
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Ginkgo Bioworks, Boston, Massachusetts 02210, United States
| | - Anil K Madugundu
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Institute of Bioinformatics, International Technology Park, Bangalore 560066, Karnataka, India.,Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India.,Center for Molecular Medicine, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bangalore 560029, Karnataka, India.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Savita Udainiya
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Center for Molecular Medicine, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bangalore 560029, Karnataka, India.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Jevon A Cutler
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Pre-Doctoral Training Program in Human Genetics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Santosh Renuse
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, United States.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Li Wang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Nicole A Pearson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | | | - Nupam Mahajan
- Siteman Cancer Center, Washington University, St. Louis, Missouri 63110, United States
| | - Akhilesh Pandey
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Center for Molecular Medicine, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bangalore 560029, Karnataka, India.,Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, United States.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Xinyan Wu
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, United States.,Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55905, United States
| |
Collapse
|
40
|
Callahan V, Hawks S, Crawford MA, Lehman CW, Morrison HA, Ivester HM, Akhrymuk I, Boghdeh N, Flor R, Finkielstein CV, Allen IC, Weger-Lucarelli J, Duggal N, Hughes MA, Kehn-Hall K. The Pro-Inflammatory Chemokines CXCL9, CXCL10 and CXCL11 Are Upregulated Following SARS-CoV-2 Infection in an AKT-Dependent Manner. Viruses 2021; 13:1062. [PMID: 34205098 PMCID: PMC8226769 DOI: 10.3390/v13061062] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/22/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a highly transmissible RNA virus that is the causative agent of the Coronavirus disease 2019 (COVID-19) pandemic. Patients with severe COVID-19 may develop acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) and require mechanical ventilation. Key features of SARS-CoV-2 induced pulmonary complications include an overexpression of pro-inflammatory chemokines and cytokines that contribute to a 'cytokine storm.' In the current study an inflammatory state in Calu-3 human lung epithelial cells was characterized in which significantly elevated transcripts of the immunostimulatory chemokines CXCL9, CXCL10, and CXCL11 were present. Additionally, an increase in gene expression of the cytokines IL-6, TNFα, and IFN-γ was observed. The transcription of CXCL9, CXCL10, IL-6, and IFN-γ was also induced in the lungs of human transgenic angiotensin converting enzyme 2 (ACE2) mice infected with SARS-CoV-2. To elucidate cell signaling pathways responsible for chemokine upregulation in SARS-CoV-2 infected cells, small molecule inhibitors targeting key signaling kinases were used. The induction of CXCL9, CXCL10, and CXCL11 gene expression in response to SARS-CoV-2 infection was markedly reduced by treatment with the AKT inhibitor GSK690693. Samples from COVID-19 positive individuals also displayed marked increases in CXCL9, CXCL10, and CXCL11 transcripts as well as transcripts in the AKT pathway. The current study elucidates potential pathway specific targets for reducing the induction of chemokines that may be contributing to SARS-CoV-2 pathogenesis via hyperinflammation.
Collapse
Affiliation(s)
- Victoria Callahan
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (V.C.); (N.B.); (R.F.)
| | - Seth Hawks
- Department of Biomedical Science and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA; (S.H.); (C.W.L.); (H.A.M.); (I.A.); (I.C.A.); (J.W.-L.); (N.D.)
| | - Matthew A. Crawford
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA; (M.A.C.); (M.A.H.)
| | - Caitlin W. Lehman
- Department of Biomedical Science and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA; (S.H.); (C.W.L.); (H.A.M.); (I.A.); (I.C.A.); (J.W.-L.); (N.D.)
| | - Holly A. Morrison
- Department of Biomedical Science and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA; (S.H.); (C.W.L.); (H.A.M.); (I.A.); (I.C.A.); (J.W.-L.); (N.D.)
| | - Hannah M. Ivester
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA 24061, USA;
| | - Ivan Akhrymuk
- Department of Biomedical Science and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA; (S.H.); (C.W.L.); (H.A.M.); (I.A.); (I.C.A.); (J.W.-L.); (N.D.)
| | - Niloufar Boghdeh
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (V.C.); (N.B.); (R.F.)
| | - Rafaela Flor
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (V.C.); (N.B.); (R.F.)
| | - Carla V. Finkielstein
- Integrated Cellular Responses Laboratory, Department of Biological Sciences and Center for Drug Discovery, Fralin Biomedical Research Institute, Virginia Polytechnic Institute and State University, Roanoke, VA 24016, USA;
| | - Irving Coy Allen
- Department of Biomedical Science and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA; (S.H.); (C.W.L.); (H.A.M.); (I.A.); (I.C.A.); (J.W.-L.); (N.D.)
- Virginia Tech Carilion School of Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24016, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - James Weger-Lucarelli
- Department of Biomedical Science and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA; (S.H.); (C.W.L.); (H.A.M.); (I.A.); (I.C.A.); (J.W.-L.); (N.D.)
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Nisha Duggal
- Department of Biomedical Science and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA; (S.H.); (C.W.L.); (H.A.M.); (I.A.); (I.C.A.); (J.W.-L.); (N.D.)
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Molly A. Hughes
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA; (M.A.C.); (M.A.H.)
| | - Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (V.C.); (N.B.); (R.F.)
- Department of Biomedical Science and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA; (S.H.); (C.W.L.); (H.A.M.); (I.A.); (I.C.A.); (J.W.-L.); (N.D.)
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| |
Collapse
|
41
|
Brandao R, Kwa MQ, Yarden Y, Brakebusch C. ACK1 is dispensable for development, skin tumor formation, and breast cancer cell proliferation. FEBS Open Bio 2021; 11:1579-1592. [PMID: 33730447 PMCID: PMC8167857 DOI: 10.1002/2211-5463.13149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/16/2021] [Indexed: 11/29/2022] Open
Abstract
Activated Cdc42‐associated kinase 1 (ACK1), a widely expressed nonreceptor tyrosine kinase, is often amplified in cancer and has been shown to interact with Cell division cycle 42 (Cdc42), Epidermal growth factor receptor (EGFR), and several other cancer‐relevant molecules, suggesting a possible role for ACK1 in development and tumor formation. To directly address this scenario, we generated mice lacking a functional ACK1 gene (ACK1 ko) using CRISPR genome editing. ACK1 ko mice developed normally, displayed no obvious defect in tissue maintenance, and were fertile. Primary ACK1‐null keratinocytes showed normal phosphorylation of EGFR, but a tendency toward reduced activation of AKT serine/threonine kinase 1 (Akt) and Mitogen‐activated protein kinase 1 (Erk). DMBA/TPA‐induced skin tumor formation did not reveal significant differences between ACK1 ko and control mice. Deletion of the ACK1 gene in the breast cancer cell lines MDA‐MB‐231, 67NR, MCF7, 4T1, and T47D caused no differences in growth. Furthermore, EGF‐induced phosphorylation kinetics of Erk, Akt, and p130Cas were not detectably altered in T47D cells by the loss of ACK1. Finally, loss of ACK1 in MDA‐MB‐231 and T47D breast cancer cells had a very limited or no effect on directed cell migration. These data do not support a major role for ACK1 in Cdc42 and EGFR signaling, development, or tumor formation.
Collapse
Affiliation(s)
- Rafael Brandao
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Denmark
| | - Mei Qi Kwa
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Denmark
| | | | - Cord Brakebusch
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Denmark
| |
Collapse
|
42
|
Yu X, Liu J, Qiu H, Hao H, Zhu J, Peng S. Combined inhibition of ACK1 and AKT shows potential toward targeted therapy against KRAS-mutant non-small-cell lung cancer. Bosn J Basic Med Sci 2021; 21:198-207. [PMID: 32530390 PMCID: PMC7982072 DOI: 10.17305/bjbms.2020.4746] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) with Kirsten RAt Sarcoma 2 viral oncogene homolog (KRAS) mutation has become a clinical challenge in cancer treatment as KRAS-mutant tumors are often resistant to conventional anti-tumor therapies. Activated CDC42-associated kinase 1 (ACK1), an activator of protein kinase B (AKT), is a promising target for KRAS-mutant tumor therapy, but the downstream ACK1 signaling remains poorly understood. The aim of this study was to evaluate the effectiveness of combined ACK1/AKT inhibition on the proliferation, migration, invasion, and apoptosis of KRAS-mutant NSCLC cell lines (NCI-H23, NCI-H358, and A549). The cells were treated with an inhibitor of either ACK1 (dasatinib or sunitinib) or AKT (MK-2206 or GDC-0068), and the optimal concentrations of the two yielding synergistic tumor-killing effects were determined by applying the Chou-Talalay equation for drug combinations. We showed that combined administration of ACK1 and AKT inhibitors at the optimal concentrations effectively suppressed NSCLC cell viability and promoted apoptosis while inducing cell cycle arrest at the G2 phase. Moreover, NSCLC cell migration and invasion were inhibited by combined ACK1/AKT inhibition. These phenomena were associated with the reduced phosphorylation levels of ACK1 and AKT (at Ser473 and Thr308), as well as alterations in caspase-dependent apoptotic signaling. Collectively, our results demonstrate the promising therapeutic potential of combined ACK1/AKT inhibition as a strategy against KRAS-mutant NSCLC. Our findings provide the basis for the clinical translation of biological targeted drugs (ACK1 and AKT inhibitors) and their rational combination in cancer treatment.
Collapse
Affiliation(s)
- Xiangjing Yu
- Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jie Liu
- Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Huawei Qiu
- Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Huiting Hao
- Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jinhong Zhu
- Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shiyun Peng
- Precision Medicine Center, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
43
|
Zhang A, Zhang R, Yang Z, Tian R. TNK2 promoted esophageal cancer progression via activating egfr-akt signaling. J Clin Lab Anal 2021; 35:e23700. [PMID: 33484472 PMCID: PMC7958005 DOI: 10.1002/jcla.23700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/15/2020] [Accepted: 12/30/2020] [Indexed: 11/16/2022] Open
Abstract
Background This study investigated the clinical implication of TNK2 expression in esophageal cancer patients’ cancer tissue samples. Methods The expression of TNK2 in esophageal cancer tissues and para‐carcinoma tissue was assessed with immunohistochemistry and Western blot analysis; besides, the proteins of CDC42, EGFR, and Akt were also analyzed. Then, Kaplan‐Meier survival curves of TNK2 protein expression level were assayed with 184 esophageal cancer patients from TCGA database. Moreover, with multiple linear regression analysis, we detected the correlations of TNK2 expression associated with tumor differentiation degree and metastasis status. Results It revealed that TNK2 was highly expressed in the cytoplasm of esophageal cancer tissues compared with para‐carcinoma tissue; besides, the proteins of CDC42, EGFR, and Akt were also up‐regulated in different levels of esophageal cancer tissues. However, there was no significant difference of the overall survival time of TNK2 protein expression in 184 esophageal cancer patients from TCGA database (p = 0.37). But, in the included study samples of our study, there was positive coefficience between TNK2 protein expression and differentiation degree in esophageal cancer with multiple linear regression analysis [R = 0.928, 95% confidence interval (0.085‐0.12)]. Conclusion Our results indicated that TNK2 was a potential diagnostic marker and promoted esophageal cancer progression through activating EGFR‐AKT signaling.
Collapse
Affiliation(s)
- Anqing Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
| | - Rongxin Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
| | - Zhiming Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
| | - Rui Tian
- Department of Thoracic Surgery, The First Affiliated Hospital of USTC, Division of life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
| |
Collapse
|
44
|
Zhu J, Liu Y, Zhao M, Cao K, Ma J, Peng S. Identification of downstream signaling cascades of ACK1 and prognostic classifiers in non-small cell lung cancer. Aging (Albany NY) 2021; 13:4482-4502. [PMID: 33495411 PMCID: PMC7906148 DOI: 10.18632/aging.202408] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/27/2020] [Indexed: 12/29/2022]
Abstract
Activated Cdc42-associated kinase 1 (ACK1) is an oncogene in multiple cancers, but the underlying mechanisms of its oncogenic role remain unclear in non-small cell lung cancer (NSCLC). Herein, we comprehensively investigated the ACK1-regulated cell processes and downstream signaling pathways, as well as its prognostic value in NSCLC. We found that ACK1 gene amplification was associated with mRNA levels in The Cancer Genome Atlas (TCGA) lung cancer cohort. The Oncomine databases showed significantly elevated ACK1 levels in lung cancer. In vitro, an ACK1 inhibitor (dasatinib) increased the sensitivity of NSCLC cell lines to AKT or MEK inhibitors. RNA-sequencing results demonstrated that an ACK1 deficiency in A549 cells affected the MAPK, PI3K/AKT, and Wnt pathways. These results were validated by gene set enrichment analysis (GSEA) of data from 188 lung cancer cell lines. Using Cytoscape, we dissected 14 critical ACK1-regulated genes. The signature with the 14 genes and ACK1 could significantly dichotomize the TCGA lung cohort regarding overall survival. The prognostic accuracy of this signature was confirmed in five independent lung cancer cohorts and was further validated by a prognostic nomogram. Our study unveiled several downstream signaling pathways for ACK1, and the proposed signature may be a promising prognostic predictor for NSCLC.
Collapse
Affiliation(s)
- Jinhong Zhu
- Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Yang Liu
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Meng Zhao
- Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Kui Cao
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Jianqun Ma
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Shiyun Peng
- Department of Precision Medicine, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| |
Collapse
|
45
|
Gu J, Qian L, Zhang G, Mahajan NP, Owonikoko TK, Ramalingam SS, Sun SY. Inhibition of ACK1 delays and overcomes acquired resistance of EGFR mutant NSCLC cells to the third generation EGFR inhibitor, osimertinib. Lung Cancer 2020; 150:26-35. [PMID: 33049499 PMCID: PMC7722235 DOI: 10.1016/j.lungcan.2020.09.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 01/10/2023]
Abstract
OBJECTIVES The emergence of acquired resistance to the third generation EGFR inhibitor, osimertinib (AZD9291 or TAGRISSO™), is an unavoidable huge clinical challenge. The involvement of ACK1, a non-receptor tyrosine kinase with an oncogenic function, in regulating cell response to osimertinib has not been investigated and thus is the focus of this study. MATERIAL AND METHODS Drug effects on cell growth were evaluated by measuring cell numbers and colony formation. Apoptosis was monitored with flow cytometry for annexin V-positive cells and Western blotting for protein cleavage. Intracellular protein and mRNA alterations were detected with Western blotting and qRT-PCR, respectively. Drug effects on delaying osimertinib acquired resistance were determined using colony formation in vitro and xenografts in nude mice in vivo, respectively. Cell senescence was assayed by β-galactosidase staining. RESULTS Inhibition of ACK1 with the novel ACK1 inhibitor, (R)-9b synergized with osimertinib in inhibiting the growth of EGFR mutant NSCLC cell lines. Similar results were also generated with ACK1 gene knockdown. The combination of osimertinib and (R)-9b enhanced induction of apoptosis. In both in vitro and in vivo long-term resistance delay assays, the combination of (R)-9b and osimertinib clearly delayed the emergence of osimertinib-resistance. Further, the (R)-9b and osimertinib combination was also effective in inhibiting the growth of EGFR mutant NSCLC cell lines with acquired resistance to osimertinib, which possess elevated levels of ACK1, and the growth of osimertinib-resistant tumors in vivo. In some resistant cell lines, the combinations induced senescence in addition to induction of apoptosis. CONCLUSIONS These novel findings suggest that ACK1 inhibition might be a potential and innovative strategy for delaying and overcoming osimertinb acquired resistance.
Collapse
Affiliation(s)
- Jiajia Gu
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Nanjing, Jiangsu, PR China; Departments of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA, USA
| | - Luxi Qian
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Nanjing, Jiangsu, PR China; Departments of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA, USA
| | - Guojing Zhang
- Departments of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA, USA
| | - Nupam P Mahajan
- Department of Surgery, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Taofeek K Owonikoko
- Departments of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA, USA
| | - Suresh S Ramalingam
- Departments of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA, USA
| | - Shi-Yong Sun
- Departments of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA, USA.
| |
Collapse
|
46
|
Ji Z, Njauw CN, Guhan S, Kumar R, Reddy B, Rajadurai A, Flaherty K, Tsao H. Loss of ACK1 Upregulates EGFR and Mediates Resistance to BRAF Inhibition. J Invest Dermatol 2020; 141:1317-1324.e1. [PMID: 33159968 DOI: 10.1016/j.jid.2020.06.041] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/27/2020] [Accepted: 06/29/2020] [Indexed: 10/23/2022]
Abstract
Targeted BRAF(V600E) suppression by selective BRAF inhibitors (BRAFis; e.g., vemurafenib and dabrafenib) has led to a sea change in the treatment of metastatic melanoma. Despite frequent upfront responses, acquired resistance has compromised long-term applicability. Among the various mechanisms of resistance, activation of multiple receptor tyrosine kinases is a known critical factor that contributes to vemurafenib resistance. EGFR activation has been recurrently identified in a set of vemurafenib-resistant melanomas, but little is known about how EGFR, or possibly other receptor tyrosine kinases, becomes activated. Here, we report that ACK1, a protein kinase that modulates EGFR turnover, is downregulated in vemurafenib-resistant melanoma cells. We also found that ACK1 depletion with short hairpin RNA decreased EGFR degradation when activated by epidermal growth factor, increased EGFR protein expression, and conferred resistance to BRAFis both in vitro and in vivo. Vemurafenib resistance mediated by ACK1 inhibition can be reversed by the EGFR inhibitor gefitinib. Our data indicate that ACK1 loss may be a post-transcriptional mechanism that increases EGFR signaling and contributes to drug resistance.
Collapse
Affiliation(s)
- Zhenyu Ji
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ching-Ni Njauw
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Samantha Guhan
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Raj Kumar
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bobby Reddy
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anpuchelvi Rajadurai
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Keith Flaherty
- Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hensin Tsao
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
47
|
New Era of Diacylglycerol Kinase, Phosphatidic Acid and Phosphatidic Acid-Binding Protein. Int J Mol Sci 2020; 21:ijms21186794. [PMID: 32947951 PMCID: PMC7555651 DOI: 10.3390/ijms21186794] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022] Open
Abstract
Diacylglycerol kinase (DGK) phosphorylates diacylglycerol (DG) to generate phosphatidic acid (PA). Mammalian DGK consists of ten isozymes (α–κ) and governs a wide range of physiological and pathological events, including immune responses, neuronal networking, bipolar disorder, obsessive-compulsive disorder, fragile X syndrome, cancer, and type 2 diabetes. DG and PA comprise diverse molecular species that have different acyl chains at the sn-1 and sn-2 positions. Because the DGK activity is essential for phosphatidylinositol turnover, which exclusively produces 1-stearoyl-2-arachidonoyl-DG, it has been generally thought that all DGK isozymes utilize the DG species derived from the turnover. However, it was recently revealed that DGK isozymes, except for DGKε, phosphorylate diverse DG species, which are not derived from phosphatidylinositol turnover. In addition, various PA-binding proteins (PABPs), which have different selectivities for PA species, were recently found. These results suggest that DGK–PA–PABP axes can potentially construct a large and complex signaling network and play physiologically and pathologically important roles in addition to DGK-dependent attenuation of DG–DG-binding protein axes. For example, 1-stearoyl-2-docosahexaenoyl-PA produced by DGKδ interacts with and activates Praja-1, the E3 ubiquitin ligase acting on the serotonin transporter, which is a target of drugs for obsessive-compulsive and major depressive disorders, in the brain. This article reviews recent research progress on PA species produced by DGK isozymes, the selective binding of PABPs to PA species and a phosphatidylinositol turnover-independent DG supply pathway.
Collapse
|
48
|
Russell SJ, Schneider MF. Alternative signaling pathways from IGF1 or insulin to AKT activation and FOXO1 nuclear efflux in adult skeletal muscle fibers. J Biol Chem 2020; 295:15292-15306. [PMID: 32868454 DOI: 10.1074/jbc.ra120.013634] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 08/20/2020] [Indexed: 12/25/2022] Open
Abstract
Muscle atrophy is regulated by the balance between protein degradation and synthesis. FOXO1, a transcription factor, helps to determine this balance by activating pro-atrophic gene transcription when present in muscle fiber nuclei. Foxo1 nuclear efflux is promoted by AKT-mediated Foxo1 phosphorylation, eliminating FOXO1's atrophy-promoting effect. AKT activation can be promoted by insulin-like growth factor 1 (IGF1) or insulin via a pathway including IGF1 or insulin, phosphatidylinositol 3-kinase, and AKT. We used confocal fluorescence time-lapse imaging of FOXO1-GFP in adult isolated living muscle fibers maintained in culture to explore the effects of IGF1 and insulin on FOXO1-GFP nuclear efflux with and without pharmacological inhibitors. We observed that although AKT inhibitor blocks the IGF1- or insulin-induced effect on FOXO1 nuclear efflux, phosphatidylinositol 3-kinase inhibitors, which we show to be effective in these fibers, do not. We also found that inhibition of the protein kinase ACK1 or ATM contributes to the suppression of FOXO1 nuclear efflux after IGF1. These results indicate a novel pathway that has been unexplored in the IGF1- or insulin-induced regulation of FOXO1 and present information useful both for therapeutic interventions for muscle atrophy and for further investigative areas into insulin insensitivity and type 2 diabetes.
Collapse
Affiliation(s)
- Sarah J Russell
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Martin F Schneider
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
49
|
Kim EH, Cao D, Mahajan NP, Andriole GL, Mahajan K. ACK1-AR and AR-HOXB13 signaling axes: epigenetic regulation of lethal prostate cancers. NAR Cancer 2020; 2:zcaa018. [PMID: 32885168 PMCID: PMC7454006 DOI: 10.1093/narcan/zcaa018] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/22/2020] [Accepted: 08/13/2020] [Indexed: 12/24/2022] Open
Abstract
The androgen receptor (AR) is a critical transcription factor in prostate cancer (PC) pathogenesis. Its activity in malignant cells is dependent on interactions with a diverse set of co-regulators. These interactions fluctuate depending on androgen availability. For example, the androgen depletion increases the dependence of castration-resistant PCs (CRPCs) on the ACK1 and HOXB13 cell survival pathways. Activated ACK1, an oncogenic tyrosine kinase, phosphorylates cytosolic and nuclear proteins, thereby avoiding the inhibitory growth consequences of androgen depletion. Notably, ACK1-mediated phosphorylation of histone H4, which leads to epigenetic upregulation of AR expression, has emerged as a critical mechanism of CRPC resistance to anti-androgens. This resistance can be targeted using the ACK1-selective small-molecule kinase inhibitor (R)- 9b. CRPCs also deploy the bromodomain and extra-terminal domain protein BRD4 to epigenetically increase HOXB13 gene expression, which in turn activates the MYC target genes AURKA/AURKB. HOXB13 also facilitates ligand-independent recruitment of the AR splice variant AR-V7 to chromatin, compensating for the loss of the chromatin remodeling protein, CHD1, and restricting expression of the mitosis control gene HSPB8. These studies highlight the crosstalk between AR-ACK1 and AR-HOXB13 pathways as key mediators of CRPC recurrence.
Collapse
Affiliation(s)
- Eric H Kim
- Division of Urologic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Dengfeng Cao
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Nupam P Mahajan
- Division of Urologic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Gerald L Andriole
- Division of Urologic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kiran Mahajan
- Division of Urologic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
50
|
Zhu J, Liu Y, Ao H, Liu M, Zhao M, Ma J. Comprehensive Analysis of the Immune Implication of ACK1 Gene in Non-small Cell Lung Cancer. Front Oncol 2020; 10:1132. [PMID: 32793482 PMCID: PMC7390926 DOI: 10.3389/fonc.2020.01132] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/05/2020] [Indexed: 01/21/2023] Open
Abstract
Activated Cdc42-associated kinase1 (ACK1), a non-receptor tyrosine kinase, has been considered as an oncogene and therapeutic target in various cancers. However, its contribution to cancer immunity remains uncertain. Here we first compared the profiles of immune cells in cancerous and normal tissues in The Cancer Genome Atlas (TCGA) lung cancer cohorts. Next, we found that the immune cell infiltration levels were associated with the ACK1 gene copy numbers in lung cancer. Consistently, our RNA-seq data unveiled that the silencing of ACK1 upregulated several immune pathways in lung cancer cells, including the T cell receptor signaling pathway. The impacts of ACK1 on immune activity were validated by Gene Set Enrichment Analysis of RNA-seq data of 188 lung cancer cell lines from the public database. A pathway enrichment analysis of 35 ACK1-associated immunomodulators and 50 tightly correlated genes indicated the involvement of the PI3K-Akt and Ras signaling pathways. Based on ACK1-associated immunomodulators, we established multiple-gene risk prediction signatures using the Cox regression model. The resulting risk scores were an independent prognosis predictor in the TCGA lung cohorts. We also accessed the prognostic accuracy of the risk scores with a receiver operating characteristic methodology. Finally, a prognostic nomogram, accompanied by a calibration curve, was constructed to predict individuals' 3- and 5-year survival probabilities. Our findings provided evidence of ACK1's implication in tumor immunity, suggesting that ACK1 may be a potential immunotherapeutic target for non-small cell lung cancer (NSCLC). The nominated immune signature is a promising prognostic biomarker in NSCLC.
Collapse
Affiliation(s)
- Jinhong Zhu
- Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yang Liu
- Department of Clinical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Haijiao Ao
- Department of Clinical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mingdong Liu
- Department of Clinical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Meng Zhao
- Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jianqun Ma
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|