1
|
Lozzi I, Arnold A, Barone M, Johnson JC, Sinn BV, Eschrich J, Gebert P, Wang R, Hu M, Feldbrügge L, Schirmeier A, Reutzel-Selke A, Malinka T, Krenzien F, Schöning W, Modest DP, Pratschke J, Sauer IM, Felsenstein M. Clinical prognosticators and targets in the immune microenvironment of intrahepatic cholangiocarcinoma. Oncoimmunology 2024; 13:2406052. [PMID: 39359389 PMCID: PMC11445892 DOI: 10.1080/2162402x.2024.2406052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 08/06/2024] [Accepted: 09/14/2024] [Indexed: 10/04/2024] Open
Abstract
Background Intrahepatic cholangiocarcinoma (ICC) is a disease with poor prognosis and limited therapeutic options. We investigated the tumor immune microenvironment (TIME) to identify predictors of disease outcome and to explore targets for therapeutic modulation. Methods Liver tissue samples were collected during 2008-2019 from patients (n = 139) diagnosed with ICC who underwent curative intent surgery without neoadjuvant chemotherapy. Samples from the discovery cohort (n = 86) were immunohistochemically analyzed on tissue microarrays (TMAs) for the expression of CD68, CD3, CD4, CD8, Foxp3, PD-L1, STAT1, and p-STAT1 in tumor core and stroma areas. Results were digitally analyzed using QuPath software and correlated with clinicopathological characteristics. For validation of TIME-related biomarkers, we performed multiplex imaging mass cytometry (IMC) in a validation cohort (n = 53). Results CD68+ cells were the predominant immune cell type in the TIME of ICC. CD4+high T cell density correlated with better overall survival (OS). Prediction modeling together with validation cohort confirmed relevance of CD4+ cells, PD-L1 expression by immune cells in the stroma and N-stage on overall disease outcome. In turn, IMC analyses revealed that silent CD3+CD4+ clusters inversely impacted survival. Among annotated immune cell clusters, PD-L1 was most relevantly expressed by CD4+FoxP3+ cells. A subset of tumors with high density of immune cells ("hot" cluster) correlated with PD-L1 expression and could identify a group of candidates for immune checkpoint inhibition (ICI). Ultimately, higher levels of STAT1 expression were associated with higher lymphocyte infiltration and PD-L1 expression. Conclusions These results highlight the importance of CD4+ T cells in immune response against ICC. Secondly, a subset of tumors with "hot" TIME represents potential candidates for ICI, while stimulation of STAT1 pathway could be a potential target to turn "cold" into "hot" TIME in ICC.
Collapse
Affiliation(s)
- Isis Lozzi
- Department of Surgery, Experimental Surgery, CCM, CVK, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alexander Arnold
- Department of Pathology, CCM, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Matthias Barone
- Translational Immunology, Berlin Institute of Health & Charité University Medicine, Berlin, Germany
| | - Juliette Claire Johnson
- Translational Immunology, Berlin Institute of Health & Charité University Medicine, Berlin, Germany
| | - Bruno V Sinn
- Department of Pathology, CCM, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Johannes Eschrich
- Department of Hepatology and Gastroenterology, CCM, CVK, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- BIH Charité Clinician Scientist Program Charité - Universitätsmedizin Berlin and The Berlin Institute of Health at Charité (BIH), Berlin, Germany
| | - Pimrapat Gebert
- Institute of Biometry and Clinical Epidemiology, CCM, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ruonan Wang
- Department of Surgery, Experimental Surgery, CCM, CVK, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mengwen Hu
- Department of Surgery, Experimental Surgery, CCM, CVK, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Linda Feldbrügge
- Department of Surgery, Experimental Surgery, CCM, CVK, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- BIH Charité Clinician Scientist Program Charité - Universitätsmedizin Berlin and The Berlin Institute of Health at Charité (BIH), Berlin, Germany
| | - Anja Schirmeier
- Department of Surgery, Experimental Surgery, CCM, CVK, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anja Reutzel-Selke
- Department of Surgery, Experimental Surgery, CCM, CVK, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thomas Malinka
- Department of Surgery, Experimental Surgery, CCM, CVK, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Felix Krenzien
- Department of Surgery, Experimental Surgery, CCM, CVK, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- BIH Charité Clinician Scientist Program Charité - Universitätsmedizin Berlin and The Berlin Institute of Health at Charité (BIH), Berlin, Germany
| | - Wenzel Schöning
- Department of Surgery, Experimental Surgery, CCM, CVK, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dominik P Modest
- Department of Hematology, Oncology, and Cancer Immunology, CCM, CVK, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- DKFZ, German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Johann Pratschke
- Department of Surgery, Experimental Surgery, CCM, CVK, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Igor M Sauer
- Department of Surgery, Experimental Surgery, CCM, CVK, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Matthäus Felsenstein
- Department of Surgery, Experimental Surgery, CCM, CVK, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- BIH Charité Clinician Scientist Program Charité - Universitätsmedizin Berlin and The Berlin Institute of Health at Charité (BIH), Berlin, Germany
| |
Collapse
|
2
|
Wang D, Kaniowski D, Jacek K, Su YL, Yu C, Hall J, Li H, Feng M, Hui S, Kaminska B, DeFranciscis V, Esposito CL, DiRuscio A, Zhang B, Marcucci G, Kuo YH, Kortylewski M. Bi-functional CpG-STAT3 decoy oligonucleotide triggers multilineage differentiation of acute myeloid leukemia in mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102268. [PMID: 39171140 PMCID: PMC11338104 DOI: 10.1016/j.omtn.2024.102268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 07/12/2024] [Indexed: 08/23/2024]
Abstract
Acute myeloid leukemia (AML) cells resist differentiation stimuli despite high expression of innate immune receptors, such as Toll-like receptor 9 (TLR9). We previously demonstrated that targeting Signal Transducer and Activator of Transcription 3 (STAT3) using TLR9-targeted decoy oligodeoxynucleotide (CpG-STAT3d) increases immunogenicity of human and mouse AML cells. Here, we elucidated molecular mechanisms of inv(16) AML reprogramming driven by STAT3-inhibition/TLR9-activation in vivo. At the transcriptional levels, AML cells isolated from mice after intravenous administration of CpG-STAT3d or leukemia-targeted Stat3 silencing and TLR9 co-stimulation, displayed similar upregulation of myeloid cell differentiation (Irf8, Cebpa, Itgam) and antigen-presentation (Ciita, Il12a, B2m)-related genes with concomitant reduction of leukemia-promoting Runx1. Single-cell transcriptomics revealed that CpG-STAT3d induced multilineage differentiation of AML cells into monocytes/macrophages, erythroblastic and B cell subsets. As shown by an inducible Irf8 silencing in vivo, IRF8 upregulation was critical for monocyte-macrophage differentiation of leukemic cells. TLR9-driven AML cell reprogramming was likely enabled by downregulation of STAT3-controlled methylation regulators, such as DNMT1 and DNMT3. In fact, the combination of DNA methyl transferase (DNMT) inhibition using azacitidine with CpG oligonucleotides alone mimicked CpG-STAT3d effects, resulting in AML cell differentiation, T cell activation, and systemic leukemia regression. These findings highlight immunotherapeutic potential of bi-functional oligonucleotides to unleash TLR9-driven differentiation of leukemic cells by concurrent STAT3 and/or DNMT inhibition.
Collapse
Affiliation(s)
- Dongfang Wang
- Department of Immuno-Oncology, Beckman Research Institute at City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Damian Kaniowski
- Department of Immuno-Oncology, Beckman Research Institute at City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Karol Jacek
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Yu-Lin Su
- Department of Immuno-Oncology, Beckman Research Institute at City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Chunsong Yu
- Department of Immuno-Oncology, Beckman Research Institute at City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Jeremy Hall
- Department of Immuno-Oncology, Beckman Research Institute at City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Haiqing Li
- Integrative Genomics Core, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute at City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Susanta Hui
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Bożena Kaminska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | | | - Carla Lucia Esposito
- Institute for Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), CNR, 80100 Naples, Italy
| | - Annalisa DiRuscio
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Bin Zhang
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute at City of Hope Comprehensive Cancer Center, Duarte, CA, USA
- Gehr Family Center for Leukemia Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Guido Marcucci
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute at City of Hope Comprehensive Cancer Center, Duarte, CA, USA
- Gehr Family Center for Leukemia Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Ya-Huei Kuo
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute at City of Hope Comprehensive Cancer Center, Duarte, CA, USA
- Gehr Family Center for Leukemia Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Marcin Kortylewski
- Department of Immuno-Oncology, Beckman Research Institute at City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| |
Collapse
|
3
|
Li Y, Jiang F, Zhu S, Jia H, Li C. STAT3 drives the malignant progression of low-grade gliomas through modulating the expression of STAT1, FOXO1, and MYC. Front Mol Biosci 2024; 11:1419072. [PMID: 38948079 PMCID: PMC11211654 DOI: 10.3389/fmolb.2024.1419072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/06/2024] [Indexed: 07/02/2024] Open
Abstract
Low-grade glioma (LGG) is a prevalent and lethal primary brain malignancy, with most patients succumbing to recurrence and progression. The signal transducer and activator of transcription (STAT) family has long been implicated in tumor initiation and progression. However, a comprehensive evaluation of the expression status and overall function of STAT genes in LGG remains largely unreported. In this study, we investigated the association between the expression of STAT family genes and the progression of LGG. Through a comprehensive analysis that combined bioinformatics screening and validation assays, we determined that STAT1, STAT3, and STAT5A were upregulated and contributed to the malignant progression of LGG. Notably, our findings suggest that STAT3 is a critical prognostic marker that regulates the progression of LGG. STAT3 emerged as the most significant prognostic indicator governing the advancement of LGG. Additionally, our inquiry into the STAT3-binding proteins and differentially expressed-correlated genes (DEGs) revealed that STAT3 played a pivotal role in the progression of LGG by stimulating the expression of STAT1, FOXO1, and MYC. In summary, our recent study conducted a thorough analysis of the STAT family genes and revealed that directing therapeutic interventions towards STAT3 holds potential as a viable strategy for treating patients with LGG.
Collapse
Affiliation(s)
| | | | | | - Hongwei Jia
- Department of Pharmacy, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou First People’s Hospital, Xuzhou, China
| | - Changwei Li
- Department of Pharmacy, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou First People’s Hospital, Xuzhou, China
| |
Collapse
|
4
|
Perzolli A, Koedijk JB, Zwaan CM, Heidenreich O. Targeting the innate immune system in pediatric and adult AML. Leukemia 2024; 38:1191-1201. [PMID: 38459166 PMCID: PMC11147779 DOI: 10.1038/s41375-024-02217-7] [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: 01/29/2024] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/10/2024]
Abstract
While the introduction of T cell-based immunotherapies has improved outcomes in many cancer types, the development of immunotherapies for both adult and pediatric AML has been relatively slow and limited. In addition to the need to identify suitable target antigens, a better understanding of the immunosuppressive tumor microenvironment is necessary for the design of novel immunotherapy approaches. To date, most immune characterization studies in AML have focused on T cells, while innate immune lineages such as monocytes, granulocytes and natural killer (NK) cells, received less attention. In solid cancers, studies have shown that innate immune cells, such as macrophages, myeloid-derived suppressor cells and neutrophils are highly plastic and may differentiate into immunosuppressive cells depending on signals received in their microenvironment, while NK cells appear to be functionally impaired. Hence, an in-depth characterization of the innate immune compartment in the TME is urgently needed to guide the development of immunotherapeutic interventions for AML. In this review, we summarize the current knowledge on the innate immune compartment in AML, and we discuss how targeting its components may enhance T cell-based- and other immunotherapeutic approaches.
Collapse
Affiliation(s)
- Alicia Perzolli
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - Joost B Koedijk
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - C Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - Olaf Heidenreich
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands.
- Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK.
| |
Collapse
|
5
|
Alshaalan KS, Albawardi TK, Zhra M, Bin Sulaiman N, Jnied OY, Saleem RA, Aljada A. Differential Expression of LMNA/C and Insulin Receptor Transcript Variants in Peripheral Blood Mononuclear Cells of Leukemia Patients. J Clin Med 2024; 13:2568. [PMID: 38731097 PMCID: PMC11084221 DOI: 10.3390/jcm13092568] [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: 03/18/2024] [Revised: 04/16/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
Background: Recent research has identified alternative transcript variants of LMNA/C (LMNA, LMNC, LMNAΔ10, and LMNAΔ50) and insulin receptors (INSRs) as potential biomarkers for various types of cancer. The objective of this study was to assess the expression of LMNA/C and INSR transcript variants in peripheral blood mononuclear cells (PBMCs) of leukemia patients to investigate their potential as diagnostic biomarkers. Methods: Quantitative TaqMan reverse transcriptase polymerase chain reaction (RT-qPCR) was utilized to quantify the mRNA levels of LMNA/C (LMNA, LMNC, LMNAΔ10, and LMNAΔ50) as well as INSR (IR-A and IR-B) variants in PBMCs obtained from healthy individuals (n = 32) and patients diagnosed with primary leukemias (acute myeloid leukemia (AML): n = 17; acute lymphoblastic leukemia (ALL): n = 8; chronic myeloid leukemia (CML): n = 5; and chronic lymphocytic leukemia (CLL): n = 15). Results: Only LMNA and LMNC transcripts were notably present in PBMCs. Both exhibited significantly decreased expression levels in leukemia patients compared to the healthy control group. Particularly, the LMNC:LMNA ratio was notably higher in AML patients. Interestingly, IR-B expression was not detectable in any of the PBMC samples, precluding the calculation of the IR-A:IR-B ratio as a diagnostic marker. Despite reduced expression across all types of leukemia, IR-A levels remained detectable, indicating its potential involvement in disease progression. Conclusions: This study highlights the distinct expression patterns of LMNA/C and INSR transcript variants in PBMCs of leukemia patients. The LMNC:LMNA ratio shows promise as a potential diagnostic indicator for AML, while further research is necessary to understand the role of IR-A in leukemia pathogenesis and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Khalid Saud Alshaalan
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Turki Khalid Albawardi
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Mahmoud Zhra
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.)
| | - Norah Bin Sulaiman
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.)
| | - Osama Yaheia Jnied
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.)
| | - Rimah Abdullah Saleem
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.)
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.)
| |
Collapse
|
6
|
Kaur G, Roy B. Decoding Tumor Angiogenesis for Therapeutic Advancements: Mechanistic Insights. Biomedicines 2024; 12:827. [PMID: 38672182 PMCID: PMC11048662 DOI: 10.3390/biomedicines12040827] [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: 03/15/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Tumor angiogenesis, the formation of new blood vessels within the tumor microenvironment, is considered a hallmark of cancer progression and represents a crucial target for therapeutic intervention. The tumor microenvironment is characterized by a complex interplay between proangiogenic and antiangiogenic factors, regulating the vascularization necessary for tumor growth and metastasis. The study of angiogenesis involves a spectrum of techniques, spanning from biomarker assessment to advanced imaging modalities. This comprehensive review aims to provide insights into the molecular intricacies, regulatory dynamics, and clinical implications of tumor angiogenesis. By delving into these aspects, we gain a deeper understanding of the processes driving vascularization in tumors, paving the way for the development of novel and effective antiangiogenic therapies in the fight against cancer.
Collapse
Affiliation(s)
- Geetika Kaur
- Integrative Biosciences Center, Wayne State University, Detroit, MI 48202, USA;
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Bipradas Roy
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
7
|
Chen H, Bian A, Zhou W, Miao Y, Ye J, Li J, He P, Zhang Q, Sun Y, Sun Z, Ti C, Chen Y, Yi Z, Liu M. Discovery of the Highly Selective and Potent STAT3 Inhibitor for Pancreatic Cancer Treatment. ACS CENTRAL SCIENCE 2024; 10:579-594. [PMID: 38559310 PMCID: PMC10979493 DOI: 10.1021/acscentsci.3c01440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/11/2024] [Accepted: 01/19/2024] [Indexed: 04/04/2024]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is an attractive cancer therapeutic target. Unfortunately, targeting STAT3 with small molecules has proven to be very challenging, and for full activation of STAT3, the cooperative phosphorylation of both tyrosine 705 (Tyr705) and serine 727 (Ser727) is needed. Further, a selective inhibitor of STAT3 dual phosphorylation has not been developed. Here, we identified a low nanomolar potency and highly selective small-molecule STAT3 inhibitor that simultaneously inhibits both STAT3 Tyr705 and Ser727 phosphorylation. YY002 potently inhibited STAT3-dependent tumor cell growth in vitro and achieved potent suppression of tumor growth and metastasis in vivo. More importantly, YY002 exhibited favorable pharmacokinetics, an acceptable safety profile, and superior antitumor efficacy compared to BBI608 (STAT3 inhibitor that has advanced into phase III trials). For the mechanism, YY002 is selectively bound to the STAT3 Src Homology 2 (SH2) domain over other STAT members, which strongly suppressed STAT3 nuclear and mitochondrial functions in STAT3-dependent cells. Collectively, this study suggests the potential of small-molecule STAT3 inhibitors as possible anticancer therapeutic agents.
Collapse
Affiliation(s)
- Huang Chen
- Shanghai
Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences
and School of Life Sciences, East China
Normal University, Shanghai 200241, P.R. China
- Shanghai
Yuyao Biotech Co., LTD. Shanghai 200241, China
| | - Aiwu Bian
- Shanghai
Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences
and School of Life Sciences, East China
Normal University, Shanghai 200241, P.R. China
- Shanghai
Yuyao Biotech Co., LTD. Shanghai 200241, China
| | - Wenbo Zhou
- Shanghai
Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences
and School of Life Sciences, East China
Normal University, Shanghai 200241, P.R. China
- Shanghai
Yuyao Biotech Co., LTD. Shanghai 200241, China
| | - Ying Miao
- Shanghai
Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences
and School of Life Sciences, East China
Normal University, Shanghai 200241, P.R. China
| | - Jiangnan Ye
- Shanghai
Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences
and School of Life Sciences, East China
Normal University, Shanghai 200241, P.R. China
| | - Jiahui Li
- Southern
Medical University Affiliated Fengxian Hospital, Shanghai 201499, China
| | - Peng He
- Shanghai
Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences
and School of Life Sciences, East China
Normal University, Shanghai 200241, P.R. China
| | - Qiansen Zhang
- Shanghai
Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences
and School of Life Sciences, East China
Normal University, Shanghai 200241, P.R. China
| | - Yue Sun
- Shanghai
Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences
and School of Life Sciences, East China
Normal University, Shanghai 200241, P.R. China
| | - Zhenliang Sun
- Southern
Medical University Affiliated Fengxian Hospital, Shanghai 201499, China
| | - Chaowen Ti
- Shanghai
Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences
and School of Life Sciences, East China
Normal University, Shanghai 200241, P.R. China
| | - Yihua Chen
- Shanghai
Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences
and School of Life Sciences, East China
Normal University, Shanghai 200241, P.R. China
| | - Zhengfang Yi
- Shanghai
Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences
and School of Life Sciences, East China
Normal University, Shanghai 200241, P.R. China
| | - Mingyao Liu
- Shanghai
Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences
and School of Life Sciences, East China
Normal University, Shanghai 200241, P.R. China
- Shanghai
Yuyao Biotech Co., LTD. Shanghai 200241, China
| |
Collapse
|
8
|
Adesoye T, Tripathy D, Hunt KK, Keyomarsi K. Exploring Novel Frontiers: Leveraging STAT3 Signaling for Advanced Cancer Therapeutics. Cancers (Basel) 2024; 16:492. [PMID: 38339245 PMCID: PMC10854592 DOI: 10.3390/cancers16030492] [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: 10/18/2023] [Revised: 12/21/2023] [Accepted: 12/25/2023] [Indexed: 02/12/2024] Open
Abstract
Signal Transducer and Activator of Transcription 3 (STAT3) plays a significant role in diverse physiologic processes, including cell proliferation, differentiation, angiogenesis, and survival. STAT3 activation via phosphorylation of tyrosine and serine residues is a complex and tightly regulated process initiated by upstream signaling pathways with ligand binding to receptor and non-receptor-linked kinases. Through downstream deregulation of target genes, aberrations in STAT3 activation are implicated in tumorigenesis, metastasis, and recurrence in multiple cancers. While there have been extensive efforts to develop direct and indirect STAT3 inhibitors using novel drugs as a therapeutic strategy, direct clinical application remains in evolution. In this review, we outline the mechanisms of STAT3 activation, the resulting downstream effects in physiologic and malignant settings, and therapeutic strategies for targeting STAT3. We also summarize the pre-clinical and clinical evidence of novel drug therapies targeting STAT3 and discuss the challenges of establishing their therapeutic efficacy in the current clinical landscape.
Collapse
Affiliation(s)
- Taiwo Adesoye
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Debasish Tripathy
- Department of Breast Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Kelly K. Hunt
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Khandan Keyomarsi
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
9
|
Ke J, Zhang DG, Liu SZ, Luo Z. Functional analysis of selenok, selenot and selenop promoters and their regulation by selenium in yellow catfish Pelteobagrus fulvidraco. Gene 2023; 873:147461. [PMID: 37149273 DOI: 10.1016/j.gene.2023.147461] [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: 02/03/2023] [Revised: 04/25/2023] [Accepted: 05/01/2023] [Indexed: 05/08/2023]
Abstract
The selenok, selenot and selenop are three key selenoproteins involved in stress response. Our study, using the yellow catfish Pelteobagrus fulvidraco as the experimental animal, obtained the 1993-bp, 2000-bp and 1959-bp sequences of selenok, selenot and selenop promoters, respectively, and predicted the binding sites of several transcriptional factors on their promoters, such as Forkhead box O 4 (FoxO4), activating transcription factor 4 (ATF4), Kruppel-like factor 4 (KLF4) and nuclear factor erythroid 2-related factor 2 (NRF2). Selenium (Se) increased the activities of the selenok, selenot and selenop promoters. FoxO4 and Nrf2 can directly bind with selenok promoter and controlled selenok promoter activities positively; KLF4 and Nrf2 can directly bind with selenot promoter and controlled selenot promoter activities positively; FoxO4 and ATF4 can directly bind to selenop promoter and regulated selenop promoter activities positively. Se promoted FoxO4 and Nrf2 binding to selenok promoter, KLF4 and Nrf2 binding to selenot promoter, and FoxO4 and ATF4 binding to selenop promoter. Thus, we provide the first evidence for FoxO4 and Nrf2 bindnig elements in selenok promoter, KLF4 and Nrf2 binding elements in selenot promoter, and FoxO4 and ATF4 binding elements in selenop promoter, and offer novel insight into regulatory mechanism of these selenoproteins induced by Se.
Collapse
Affiliation(s)
- Jiang Ke
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan 430070, China
| | - Dian-Guang Zhang
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan 430070, China
| | - Sheng-Zan Liu
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan 430070, China
| | - Zhi Luo
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan 430070, China; Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| |
Collapse
|
10
|
He Z, Zhang X, Wang S, Dai X, Wang Q, Lu Q, Lu H, Wu Y, Wang H, Wang X, Wang H, Liu Y. The predictive value of prognosis and therapeutic response for STAT family in pancreatic cancer. Heliyon 2023; 9:e16150. [PMID: 37215832 PMCID: PMC10199255 DOI: 10.1016/j.heliyon.2023.e16150] [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: 12/09/2022] [Revised: 05/02/2023] [Accepted: 05/07/2023] [Indexed: 05/24/2023] Open
Abstract
Background Signal transducers and activators of transcription (STAT) proteins, well-known cytoplasmic transcription factors, were found to be abnormally expressed in various cancers and play essential parts in the initiation, progression and therapy resistance of cancer. Nevertheless, the functions of different STATs in pancreatic cancer (PC) and their relationship to the prognosis and immune infiltration as well as drug efficacy in PC patients have not been systematically elucidated. Methods Expression, prognosis, genetic alterations and pathway enrichment analyses of the STAT family were investigated via Oncomine, GEPIA, Kaplan Meier-plotter, cBioPortal, Metascape and GSEA. Analysis of tumor immune microenvironment was conducted by ESTIMATE and TIMER. "pRRophetic" packages were used for analysis of chemotherapeutic response. Finally, the diagnostic and prognostic value of key STATs were further validated through public datasets and immunohistochemistry. Results In this study, only STAT1 mRNA level was significantly increased in tumor tissues and highly expressed in PC cell lines via multiple datasets. PC patients with higher STAT1/4/6 expression had a worse overall survival (OS) and progression-free survival (PFS), while higher STAT5B expression was correlated with better prognosis in the TCGA cohort. The STATs-associated genes were enriched in pathways about the remodeling of tumor immune microenvironment. The STATs levels were significantly correlated with immune infiltration, except STAT6. The STAT1 was identified as a potential biomarker and its diagnostic and prognostic value were further validated at mRNA and protein levels. GSEA showed that STAT1 may be involved in the progression and immune regulations of PC. Moreover, STAT1 expression was significantly related to the level of immune checkpoint, and predicted immunotherapy and chemotherapy responses. Conclusion STAT family members were comprehensively analyzed and STAT1 was identified as an effective biomarker for predicting the survival and therapeutic response, which could be beneficial to develop better treatment strategies.
Collapse
Affiliation(s)
- Zhengfei He
- Yangzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Yangzhou 225002, China
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou 225002, China
| | - Xiaochun Zhang
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou 225002, China
| | - Shanshan Wang
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou 225002, China
| | - Xiaojun Dai
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou 225002, China
| | - Qingying Wang
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou 225002, China
| | - Qingyun Lu
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou 225002, China
| | - Haiyan Lu
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou 225002, China
| | - Yongjian Wu
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou 225002, China
| | - Hui Wang
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou 225002, China
| | - Xuemei Wang
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou 225002, China
| | - Haibo Wang
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China
- Medical College of Yangzhou University, Yangzhou, 225002, China
| | - Yanqing Liu
- Yangzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Yangzhou 225002, China
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou 225002, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China
- Medical College of Yangzhou University, Yangzhou, 225002, China
| |
Collapse
|
11
|
Steinskog ESS, Finne K, Enger M, Helgeland L, Iversen PO, McCormack E, Wiig H, Tenstad O. Isolation of lymph shows dysregulation of STAT3 and CREB pathways in the spleen and liver during leukemia development in a rat model. Microcirculation 2023; 30:e12800. [PMID: 36702790 DOI: 10.1111/micc.12800] [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: 11/17/2021] [Revised: 01/16/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND AIMS Acute myeloid leukemia (AML) is a heterogeneous malignant condition characterized by massive infiltration of poorly differentiated white blood cells in the blood stream, bone marrow, and extramedullary sites. During leukemic development, hepatosplenomegaly is expected to occur because large blood volumes are continuously filtered through these organs. We asked whether infiltration of leukemic blasts initiated a response that could be detected in the interstitial fluid phase of the spleen and liver. MATERIAL AND METHODS We used a rat model known to mimic human AML in growth characteristics and behavior. By cannulating efferent lymphatic vessels from the spleen and liver, we were able to monitor the response of the microenvironment during AML development. RESULTS AND DISCUSSION Flow cytometric analysis of lymphocytes showed increased STAT3 and CREB signaling in spleen and depressed signaling in liver, and proteins related to these pathways were identified with a different profile in lymph and plasma in AML compared with control. Additionally, several proteins were differently regulated in the microenvironment of spleen and liver in AML when compared with control. CONCLUSION Interstitial fluid, and its surrogate efferent lymph, can be used to provide unique information about responses in AML-infiltered organs and substances released to the general circulation during leukemia development.
Collapse
Affiliation(s)
| | - Kenneth Finne
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Marianne Enger
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Lars Helgeland
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Per Ole Iversen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Emmet McCormack
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Olav Tenstad
- Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
12
|
Qiao C, Liu Z, Qie S. The Implications of Microglial Regulation in Neuroplasticity-Dependent Stroke Recovery. Biomolecules 2023; 13:biom13030571. [PMID: 36979506 PMCID: PMC10046452 DOI: 10.3390/biom13030571] [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: 01/17/2023] [Revised: 02/23/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Stroke causes varying degrees of neurological deficits, leading to corresponding dysfunctions. There are different therapeutic principles for each stage of pathological development. Neuroprotection is the main treatment in the acute phase, and functional recovery becomes primary in the subacute and chronic phases. Neuroplasticity is considered the basis of functional restoration and neurological rehabilitation after stroke, including the remodeling of dendrites and dendritic spines, axonal sprouting, myelin regeneration, synapse shaping, and neurogenesis. Spatiotemporal development affects the spontaneous rewiring of neural circuits and brain networks. Microglia are resident immune cells in the brain that contribute to homeostasis under physiological conditions. Microglia are activated immediately after stroke, and phenotypic polarization changes and phagocytic function are crucial for regulating focal and global brain inflammation and neurological recovery. We have previously shown that the development of neuroplasticity is spatiotemporally consistent with microglial activation, suggesting that microglia may have a profound impact on neuroplasticity after stroke and may be a key therapeutic target for post-stroke rehabilitation. In this review, we explore the impact of neuroplasticity on post-stroke restoration as well as the functions and mechanisms of microglial activation, polarization, and phagocytosis. This is followed by a summary of microglia-targeted rehabilitative interventions that influence neuroplasticity and promote stroke recovery.
Collapse
Affiliation(s)
- Chenye Qiao
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Zongjian Liu
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Shuyan Qie
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| |
Collapse
|
13
|
Hou H, Xu Y, Xie M, Chen R. Exploring the potential molecular mechanism of trastuzumab-induced cardiotoxicity based on RNA sequencing and bioinformatics analysis. Biochem Pharmacol 2023; 208:115388. [PMID: 36563885 DOI: 10.1016/j.bcp.2022.115388] [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: 09/14/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
The cardiotoxicity of trastuzumab (TRZ) seriously affects the prognosis of breast cancer patients, but the underlying mechanisms remains to be elucidated. This study aimed to investigate the potential molecular mechanisms of TRZ-induced cardiotoxicity based on RNA sequencing (RNA-Seq) and bioinformatics analysis. Kunming mice were exposed to 10 mg/kg TRZ for 6 and 10 days, followed by echocardiography, histopathology and serum biochemical analysis to evaluate the cardiotoxicity model. The results showed no significant changes after 6 days administration of TRZ. After 10 days administration of TRZ, the mice showed cardiac dysfunction, myocardial injury and fibrosis, and the serum levels of LDH, CK, CK-MB and cTnI were increased compared to the control [CON (Day 10)] group, indicating the cardiotoxicity model was successfully established. We compared gene expression levels in mice cardiac tissues by RNA-Seq and screened out 593 differentially expressed genes (DEGs). Results based on Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, protein-protein interaction (PPI) network analysis and RT-PCR revealed that the CD74/STAT1 signaling pathway might play an important role in TRZ-induced cardiotoxicity. In the TRZ group, the protein expressions of CD74, p-STAT1 (Tyr) and p-STAT1 (Ser) were increased. The TUNEL staining showed increased apoptosis of cardiomyocytes. In addition, an increased expressions of Bax, Caspase-3, IFN-γ and TNF-α and a decreased expression of Bcl-2 were observed in Western blot results, indicating the apoptosis and inflammation levels were increased. These findings suggested that TRZ may induce cardiotoxicity in mice by activating the CD74/STAT1 signaling pathway, which might be related to the induction of apoptosis and inflammation.
Collapse
Affiliation(s)
- Huan Hou
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Ying Xu
- Department of Pharmacy, Yancheng Third People's Hospital, Yancheng, Jiangsu 224008, China
| | - Meilin Xie
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Rong Chen
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China.
| |
Collapse
|
14
|
Li D, Jiao Y, Gao W, Hu S, Li D, Zhao W, Chen P, Jin L, Zhao Y, Ma Z, Wu X, Yan Y, Sun W, Du X, Dong G. Comprehensive analysis of the prognostic and immunotherapeutic implications of STAT family members in human colorectal cancer. Front Genet 2022; 13:951252. [PMID: 36061181 PMCID: PMC9437353 DOI: 10.3389/fgene.2022.951252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Colorectal cancer (CRC) is the third most prevalent cancer worldwide and the second leading cause of cancer mortality. Signal transducer and activator of transcription (STAT) proteins are a group of transcription factors implicated in cell signal transduction and gene transcription in several cancer types. However, the level of expression, genetic alterations, and biological function of different STATs, as well as their prognostic and immunotherapeutic value in CRC remain unclear.Methods: The mRNA and protein expression levels, genetic alterations, prognostic value, gene–gene and protein–protein interaction networks, and biological function of STATs in CRC were studied using the GEPIA, HPA, cBioPortal, PrognoScan, Kaplan–Meier plotter, GeneMANIA, STRING, and Metascape databases. The expression of STATs in CRC was confirmed using immunohistochemistry (IHC). Finally, the relationship between STAT expression and immune infiltration as well as immunotherapy-associated indicators was also investigated.Results: The expression levels of STAT2/5A/5B are downregulated in CRC, and the STAT1/3/4/5B expressions were significantly associated with the tumor stage of patients with CRC. The abnormal expression of STAT2/4/5B in patients with CRC is related to the prognosis of patients with CRC. The STATs and their neighboring proteins are primarily associated with lymphocyte activation, cytokine-mediated signaling pathways, positive regulation of immune response, regulation of cytokine production, and growth hormone receptor signaling pathways in cancer. The expression of STATs was significantly associated with immune infiltration and immunotherapy response-associated indicators.Conclusion: This study may help further understand the molecular mechanism of CRC and provide new prognostic biomarkers and immunotherapy targets in patients with CRC.
Collapse
Affiliation(s)
- Dingchang Li
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yanan Jiao
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Wenxing Gao
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Shidong Hu
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Dingling Li
- Medical College of Qinghai University, Xining, China
| | - Wen Zhao
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Peng Chen
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Lujia Jin
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yingjie Zhao
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Zhaofu Ma
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xiansheng Wu
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yang Yan
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Wen Sun
- Department of Anesthesiology, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaohui Du
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Xiaohui Du, ; Guanglong Dong,
| | - Guanglong Dong
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Xiaohui Du, ; Guanglong Dong,
| |
Collapse
|
15
|
Tezcanli Kaymaz B, Selvi Gunel N, Sogutlu F, Ozates Ay NP, Baran Y, Gunduz C, Biray Avci C. Investigating the potential therapeutic role of targeting STAT3 for overcoming drug resistance by regulating energy metabolism in chronic myeloid leukemia cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:904-912. [PMID: 36033954 PMCID: PMC9392574 DOI: 10.22038/ijbms.2022.64138.14121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/26/2022] [Indexed: 11/16/2022]
Abstract
Objectives STATs are one of the initial targets of emerging anti-cancer agents due to their regulatory roles in survival, apoptosis, drug response, and cellular metabolism in CML. Aberrant STAT3 activity promotes malignancy, and acts as a metabolic switcher in cancer cell metabolism, contributing to resistance to TKI nilotinib. To investigate the possible therapeutic effects of targeting STAT3 to overcome nilotinib resistance by evaluating various cellular responses in both sensitive and nilotinib resistant CML cells and to test the hypothesis that energy metabolism modulation could be a mechanism for re-sensitization to nilotinib in resistant cells. Materials and Methods By using RNAi-mediated STAT3 gene silencing, cell viability and proliferation assays, apoptotic analysis, expressional regulations of STAT mRNA transcripts, STAT3 total, pTyr705, pSer727 protein expression levels, and metabolic activity as energy metabolism was determined in CML model K562 cells, in vitro. Results Targeting STAT3 sensitized both parental and especially nilotinib resistant cells by decreasing leukemic cell survival; inducing leukemic cell apoptosis, and decreasing STAT3 mRNA and protein expression levels. Besides, cell energy phenotype was modulated by switching energy metabolism from aerobic glycolysis to mitochondrial respiration in resistant cells. RNAi-mediated STAT3 silencing accelerated the sensitization of leukemia cells to nilotinib treatment, and STAT3-dependent energy metabolism regulation could be another underlying mechanism for regaining nilotinib response. Conclusion Targeting STAT3 is an efficient strategy for improving the development of novel CML therapeutics for regaining nilotinib response, and re-sensitization of resistant cells could be mediated by induced apoptosis and regulation in energy metabolism.
Collapse
Affiliation(s)
- Burcin Tezcanli Kaymaz
- Department of Medical Biology, Ege University Medicine Faculty, 35100, Izmir, Turkey ,Corresponding author: Burçin Tezcanlı Kaymaz. Ege University Medical School, Medical Biology Department-İzmir, Turkey Tel: 0232 390 22 98; ;
| | - Nur Selvi Gunel
- Department of Medical Biology, Ege University Medicine Faculty, 35100, Izmir, Turkey
| | - Fatma Sogutlu
- Department of Medical Biology, Ege University Medicine Faculty, 35100, Izmir, Turkey
| | | | - Yusuf Baran
- Department of Molecular Biology and Genetics, Faculty of Science, Izmir Institute of Technology, 35433, Izmir, Turkey
| | - Cumhur Gunduz
- Department of Medical Biology, Ege University Medicine Faculty, 35100, Izmir, Turkey
| | - Cigir Biray Avci
- Department of Medical Biology, Ege University Medicine Faculty, 35100, Izmir, Turkey
| |
Collapse
|
16
|
Hamed G, Omar HM, Sarhan AM, Salah HE. Proviral Integration of Moloney Virus-2 (PIM-2) Expression Level as a Prognostic Marker in Patients with Acute Myeloid Leukemia. Int J Gen Med 2022; 15:4247-4258. [PMID: 35480994 PMCID: PMC9035444 DOI: 10.2147/ijgm.s354092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/22/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose This study aimed to assess PIM-2 gene expression level as a prognostic marker in AML patients and to correlate the results with their clinical outcome. Patients and Methods This study was conducted on 50 de novo younger AML patients (median age 44). Quantitative real-time polymerase chain reaction (QRT-PCR) was used to assess the expression level of the PIM-2 gene. The transcription level of the target gene (PIM-2) was normalized to that of the reference gene (GAPDH). Twenty control samples were withdrawn from 20 age- and sex-matched individuals for the analysis of the results using the 2−ΔΔCT method. On day 28 following induction chemotherapy, patients’ bone marrow (BM) was examined for evaluation of their remission status. Results PIM-2 gene expression was higher among AML patients who did not achieve complete remission (CR); also, it was higher in patients in the intermediate and poor cytogenetic risk groups. A significant positive correlation was found between PIM-2 level and BM blasts on day 28. In AML patients, PIM-2 has been discovered to be an independent predictive factor for achieving CR following standard induction treatment. Receiver operating characteristic curve (ROC) and area under the curve (AUC) were performed for PIM-2 level at diagnosis to evaluate its role in achieving remission after induction. It was found that PIM-2 at cutoff ≤1.6 had an AUC (0.903) with a sensitivity (90.48%) and specificity (86.21%), P <0.001. Conclusion Overexpression of the PIM-2 gene is associated with induction failure and low CR.
Collapse
Affiliation(s)
- Gehad Hamed
- Department of Clinical Pathology, Faculty of Medicine, Zagazig University, Zagazig, Al-Sharkia, Egypt
- Correspondence: Gehad Hamed, Department of Clinical Pathology, Faculty of Medicine, Zagazig University, Zagazig, Al-Sharkia, 44519, Egypt, Tel +201092034529, Email
| | - Hisham M Omar
- Department of Clinical Pathology, Faculty of Medicine, Zagazig University, Zagazig, Al-Sharkia, Egypt
| | - Abbas M Sarhan
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Zagazig University, Zagazig, Al-Sharkia, Egypt
| | - Hossam E Salah
- Department of Clinical Pathology, Faculty of Medicine, Zagazig University, Zagazig, Al-Sharkia, Egypt
| |
Collapse
|
17
|
Li WZ, Xi HZ, Wang YJ, Ma HB, Cheng ZQ, Yang Y, Wu ML, Liu TM, Yang W, Wang Q, Liao MY, Xia Y, Zhang YW. Design, synthesis, and biological evaluation of benzo[b]thiophene 1,1-dioxide derivatives as potent STAT3 inhibitors. Chem Biol Drug Des 2021; 98:835-849. [PMID: 34416096 DOI: 10.1111/cbdd.13939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/06/2021] [Accepted: 07/23/2021] [Indexed: 02/05/2023]
Abstract
As a member of the signal transducer and activator of transcription (STAT) family, STAT3 plays a critical role in several biological pathways such as cell proliferation, migration, survival, and differentiation. Due to abnormal continuous activation in tumors, inhibition of STAT3 has emerged as an attractive approach for the treatment of various cancer cells. Herein, we report a series of novel STAT3 inhibitors based on benzo[b]thiophene 1,1-dioxide scaffold and evaluated their anticancer potency. Among them, compound 8b exhibited the best activity against cancer cells. Compound 8b induced apoptosis and blocked the cell cycle. Meanwhile, 8b reduced intracellular ROS content and caused the loss of mitochondrial membrane potential. Further research revealed that 8b significantly blocked STAT3 phosphorylation and STAT3-dependent dual-luciferase reporter gene experiments showed that compound 8b has a marked inhibition of STAT3-mediated Firefly luciferase activity. Molecular modeling studies revealed compound 8b occupied the pocket well with the SH2 domain in a favorable conformation.
Collapse
Affiliation(s)
- Wen-Zhen Li
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hui-Zhi Xi
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yi-Jie Wang
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong-Bo Ma
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhi-Qiang Cheng
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yu Yang
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Meng-Ling Wu
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ting-Mei Liu
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wen Yang
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qin Wang
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Meng-Ya Liao
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Xia
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yi-Wen Zhang
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Rasic P, Jovanovic-Tucovic M, Jeremic M, Djuricic SM, Vasiljevic ZV, Milickovic M, Savic D. B7 homologue 3 as a prognostic biomarker and potential therapeutic target in gastrointestinal tumors. World J Gastrointest Oncol 2021; 13:799-821. [PMID: 34457187 PMCID: PMC8371522 DOI: 10.4251/wjgo.v13.i8.799] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/19/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
The most common digestive system (DS) cancers, including tumors of the gastrointestinal tract (GIT) such as colorectal cancer (CRC), gastric cancer (GC) and esophageal cancer (EC) as well as tumors of DS accessory organs such as pancreatic and liver cancer, are responsible for more than one-third of all cancer-related deaths worldwide, despite the progress that has been achieved in anticancer therapy. Due to these limitations in treatment strategies, oncological research has taken outstanding steps towards a better understanding of cancer cell biological complexity and heterogeneity. These studies led to new molecular target-driven therapeutic approaches. Different in vivo and in vitro studies have revealed significant expression of B7 homologue 3 (B7-H3) among the most common cancers of the GIT, including CRC, GC, and EC, whereas B7-H3 expression in normal healthy tissue of these organs was shown to be absent or minimal. This molecule is able to influence the biological behavior of GIT tumors through the various immunological and nonimmunological molecular mechanisms, and some of them are shown to be the result of B7-H3-related induction of signal transduction pathways, such as Janus kinase 2/signal transducer and activator of transcription 3, phosphatidylinositol 3-kinase/protein kinase B, extracellular signal-regulated kinase, and nuclear factor-κB. B7-H3 exerts an important role in progression, metastasis and resistance to anticancer therapy in these tumors. In addition, the results of many studies suggest that B7-H3 stimulates immune evasion in GIT tumors by suppressing antitumor immune response. Accordingly, it was observed that experimental depletion or inhibition of B7-H3 in gastrointestinal cancers improved antitumor immune response, impaired tumor progression, invasion, angiogenesis, and metastasis and decreased resistance to anticancer therapy. Finally, the high expression of B7-H3 in most common cancers of the GIT was shown to be associated with poor prognosis. In this review, we summarize the established data from different GIT cancer-related studies and suggest that the B7-H3 molecule could be a promising prognostic biomarker and therapeutic target for anticancer immunotherapy in these tumors.
Collapse
Affiliation(s)
- Petar Rasic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia “Dr. Vukan Cupic“, Belgrade 11 000, Serbia
| | - Maja Jovanovic-Tucovic
- Institute of Medical and Clinical Biochemistry, School of Medicine, University of Belgrade, Belgrade 11 000, Serbia
| | - Marija Jeremic
- Institute of Medical and Clinical Biochemistry, School of Medicine, University of Belgrade, Belgrade 11 000, Serbia
| | - Slavisa M Djuricic
- Department of Clinical Pathology, Mother and Child Health Care Institute of Serbia “Dr. Vukan Cupic“, Belgrade 11 000, Serbia
- Faculty of Medicine, University of Banja Luka, Banja Luka 78 000, Bosnia and Herzegovina
| | - Zorica V Vasiljevic
- Department of Clinical Microbiology, Mother and Child Health Care Institute of Serbia “Dr. Vukan Cupic“, Belgrade 11 000, Serbia
| | - Maja Milickovic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia “Dr. Vukan Cupic“, Belgrade 11 000, Serbia
- School of Medicine, University of Belgrade, Belgrade 11 000, Serbia
| | - Djordje Savic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia “Dr. Vukan Cupic“, Belgrade 11 000, Serbia
- School of Medicine, University of Belgrade, Belgrade 11 000, Serbia
| |
Collapse
|
19
|
Abstract
The carbazole class is made up of heterocyclically structured compounds first isolated from coal tar. Their structural motif is preponderant in different synthetic materials and naturally occurring alkaloids extracted from the taxonomically related higher plants of the genus Murraya, Glycosmis, and Clausena from the Rutaceae family. Concerning the biological activity of these compounds, many research groups have assessed their antiproliferative action of carbazoles on different types of tumoral cells, such as breast, cervical, ovarian, hepatic, oral cavity, and small-cell lung cancer, and underlined their potential effects against psoriasis. One of the principal mechanisms likely involved in these effects is the ability of carbazoles to target the JAK/STATs pathway, considered essential for cell differentiation, proliferation, development, apoptosis, and inflammation. In this review, we report the studies carried out, over the years, useful to synthesize compounds with carbazole moiety designed to target these kinds of kinases.
Collapse
|
20
|
Beshay BY, Abdellatef AA, Loksha YM, Fahmy SM, Habib NS, Bekhit AEDA, Georghiou PE, Hayakawa Y, Bekhit AA. Design and synthesis of 2-Substituted-4-benzyl-5-methylimidazoles as new potential Anti-breast cancer agents to inhibit oncogenic STAT3 functions. Bioorg Chem 2021; 113:105033. [PMID: 34089945 DOI: 10.1016/j.bioorg.2021.105033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/17/2021] [Accepted: 05/24/2021] [Indexed: 12/31/2022]
Abstract
STAT3 signaling is known to be associated with tumorigenesis and further cancer cell-intrinsic activation of STAT3 leads to altered regulation of several oncogenic processes. Given the importance of STAT3 in cancer development and progression particularly breast cancer, it is crucial to discover new chemical entities of STAT3 inhibitor to develop anti-breast cancer drug candidates. Herein, 4-benzyl-2-benzylthio-5-methyl-1H-imidazole (2a) and 4-benzyl-5-methyl-2-[(2,6-difluorobenzyl)thio]-1H-imidazole (2d) from a group of thirty imidazole-bearing compounds showed greater STAT3 inhibition than their lead compounds VS1 and the oxadiazole derivative MD77. Within all tested compounds, ten derivatives effectively inhibited the growth of the two tested breast cancer cells with IC50 values ranging from 6.66 to 26.02 µM. In addition, the most potent derivatives 2a and 2d inhibited the oncogenic function of STAT3 as seen in the inhibition of colony formation and IL-6 production of breast cancer cell lines. Modeling studies provided evidence for the possible interactions of the synthesized compounds with the key residues of the STAT3-SH2 domain. Collectively, our present study suggests 2-substituted-4-benzyl-5-methylimidazoles are a new class of anti-cancer drug candidates to inhibit oncogenic STAT3 function.
Collapse
Affiliation(s)
- Botros Y Beshay
- Department of Pharmaceutical Chemistry, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport, Alexandria, Egypt
| | - Amira A Abdellatef
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Yasser M Loksha
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sinai University, Al-Arish, North Sinai, Egypt
| | - Salwa M Fahmy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, 2152 Alexandria, Egypt
| | - Nargues S Habib
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, 2152 Alexandria, Egypt
| | | | - Paris E Georghiou
- Department of Chemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Yoshihiro Hayakawa
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan.
| | - Adnan A Bekhit
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, 2152 Alexandria, Egypt; Pharmacy Program, Allied Health Department, College of Health and Sport Sciences, University of Bahrain, Bahrain; Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| |
Collapse
|
21
|
Liu H, Du T, Li C, Yang G. STAT3 phosphorylation in central leptin resistance. Nutr Metab (Lond) 2021; 18:39. [PMID: 33849593 PMCID: PMC8045279 DOI: 10.1186/s12986-021-00569-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/03/2021] [Indexed: 12/20/2022] Open
Abstract
Mechanism exploitation of energy homeostasis is urgently required because of the worldwide prevailing of obesity-related metabolic disorders in human being. Although it is well known that leptin plays a central role in regulating energy balance by suppressing food intake and promoting energy expenditure, the existence of leptin resistance in majority of obese individuals hampers the utilization of leptin therapy against these disorders. However, the mechanism of leptin resistance is largely unknown in spite of the globally enormous endeavors. Current theories to interpret leptin resistance include the impairment of leptin transport, attenuation of leptin signaling, chronic inflammation, ER tress, deficiency of autophagy, as well as leptin itself. Leptin-activated leptin receptor (LepRb) signals in hypothalamus via several pathways, in which JAK2-STAT3 pathway, the most extensively investigated one, is considered to mediate the major action of leptin in energy regulation. Upon leptin stimulation the phosphorylation of STAT3 is one of the key events in JAK2-STAT3 pathway, followed by the dimerization and nuclear translocation of this molecule. Phosphorylated STAT3 (p-STAT3), as a transcription factor, binds to and regulates its target gene such as POMC gene, playing the physiological function of leptin. Regarding POMC gene in hypothalamus however little is known about the detail of its interaction with STAT3. Moreover the status of p-STAT3 and its significance in hypothalamus of DIO mice needs to be well elucidated. This review comprehends literatures on leptin and leptin resistance and especially discusses what STAT3 phosphorylation would contribute to central leptin resistance.
Collapse
Affiliation(s)
- Huimin Liu
- College of Life Science, Henan Agricultural University, 95 Wen Hua Road, Zhengzhou, 450002, China
| | - Tianxin Du
- College of Life Science, Henan Agricultural University, 95 Wen Hua Road, Zhengzhou, 450002, China
| | - Chen Li
- College of Life Science, Henan Agricultural University, 95 Wen Hua Road, Zhengzhou, 450002, China
| | - Guoqing Yang
- College of Life Science, Henan Agricultural University, 95 Wen Hua Road, Zhengzhou, 450002, China.
| |
Collapse
|
22
|
Yu S, Chen J, Quan M, Li L, Li Y, Gao Y. CD63 negatively regulates hepatocellular carcinoma development through suppression of inflammatory cytokine-induced STAT3 activation. J Cell Mol Med 2021; 25:1024-1034. [PMID: 33277798 PMCID: PMC7812266 DOI: 10.1111/jcmm.16167] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 11/12/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022] Open
Abstract
Tetraspanin CD63 has been widely implicated in tumour progression of human malignancies. However, its role in the tumorigenesis and metastasis of hepatocellular carcinoma (HCC) remains unclear yet. In the present study, we aimed to investigate the specific function and underlying mechanisms of CD63 in HCC progression. CD63 expression in HCC tissues was detected using immunohistochemistry and quantitative real-time PCR analyses; effects of CD63 on HCC cell proliferation and migration were investigated by CCK-8 assay, colony formation assay, transwell assay and a xenograft model of nude mice. RNA-sequencing, bioinformatics analysis, dual-luciferase reporter assay and Western blot analysis were performed to explore the underlying molecular mechanisms. Results of our experiments showed that CD63 expression was frequently reduced in HCC tissues compared with adjacent normal tissues, and decreased CD63 expression was significantly associated with larger tumour size, distant site metastasis and higher tumour stages of HCC. Overexpression of CD63 inhibited HCC cell proliferation and migration, whereas knockdown of CD63 promoted these phenotypes. IL-6, IL-27 and STAT3 activity was regulated by CD63, and blockade of STAT3 activation impaired the promotive effects of CD63 knockdown on HCC cell growth and migration. Our findings identified a novel CD63-IL-6/IL-27-STAT3 axis in the development of HCC and provided a potential target for the diagnosis and treatment of this disease.
Collapse
Affiliation(s)
- Shijun Yu
- Department of OncologyShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Jingde Chen
- Department of OncologyShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Ming Quan
- Department of OncologyShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Li Li
- Department of OncologyShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Yandong Li
- Department of OncologyShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Yong Gao
- Department of OncologyShanghai East HospitalTongji University School of MedicineShanghaiChina
| |
Collapse
|
23
|
Meng H, Pang Y, Liu G, Luo Z, Tan H, Liu X. Podocarpusflavone A inhibits cell growth of skin cutaneous melanoma by suppressing STAT3 signaling. J Dermatol Sci 2020; 100:201-208. [PMID: 33127205 DOI: 10.1016/j.jdermsci.2020.10.008] [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: 06/08/2020] [Revised: 09/29/2020] [Accepted: 10/11/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND JAK2/STAT3 pathway is involved in the development and progression of melanoma once DNA damage is caused by environment and genetic factors. OBJECTIVE Here, we aimed to identify novel inhibitor of JAK2/STAT3 pathway and reveal the underlying mechanisms. METHODS Eighty MedChemExpress compounds were screened by using STAT3-Luc reporter in A375 cells. Podocarpusflavone A (PCFA) was identified as an inhibitor of STAT3, which was further verified in four melanoma cell lines. The anti-melanoma effects and mechanism of PCFA were examined and explored in melanoma cells and mouse xenograft models by using Western blot and cell-counting kit-8 assay. RESULTS PCFA exhibited potent inhibitory effects on melanoma both in vitro and in vivo. PCFA inhibited the activation of STAT3 through suppressing the phosphorylation of JAK2, and then restrained cell cycle and induced apoptosis of melanoma cells. CONCLUSION PCFA inhibits melanoma growth via the inhibition of JAK2/STAT3 pathway, which provides a promising therapeutic strategies of melanoma treatment.
Collapse
Affiliation(s)
- Huijuan Meng
- Department of Dermatology, the Affiliated Hospital of Weifang Medical University, Shandong, China
| | - Yunyan Pang
- Department of Dermatology, the Affiliated Hospital of Weifang Medical University, Shandong, China
| | - Guoyan Liu
- Department of Dermatology, the Affiliated Hospital of Weifang Medical University, Shandong, China
| | - Zengxiang Luo
- Department of Dermatology, the Affiliated Hospital of Weifang Medical University, Shandong, China
| | - Haiyang Tan
- Department of Pharmacy, the Affiliated Hospital of Weifang Medical University, Shandong, China
| | - Xiangming Liu
- Department of Dermatology, Weifang Medical University, Shandong, China.
| |
Collapse
|
24
|
Bharadwaj U, Kasembeli MM, Robinson P, Tweardy DJ. Targeting Janus Kinases and Signal Transducer and Activator of Transcription 3 to Treat Inflammation, Fibrosis, and Cancer: Rationale, Progress, and Caution. Pharmacol Rev 2020; 72:486-526. [PMID: 32198236 PMCID: PMC7300325 DOI: 10.1124/pr.119.018440] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Before it was molecularly cloned in 1994, acute-phase response factor or signal transducer and activator of transcription (STAT)3 was the focus of intense research into understanding the mammalian response to injury, particularly the acute-phase response. Although known to be essential for liver production of acute-phase reactant proteins, many of which augment innate immune responses, molecular cloning of acute-phase response factor or STAT3 and the research this enabled helped establish the central function of Janus kinase (JAK) family members in cytokine signaling and identified a multitude of cytokines and peptide hormones, beyond interleukin-6 and its family members, that activate JAKs and STAT3, as well as numerous new programs that their activation drives. Many, like the acute-phase response, are adaptive, whereas several are maladaptive and lead to chronic inflammation and adverse consequences, such as cachexia, fibrosis, organ dysfunction, and cancer. Molecular cloning of STAT3 also enabled the identification of other noncanonical roles for STAT3 in normal physiology, including its contribution to the function of the electron transport chain and oxidative phosphorylation, its basal and stress-related adaptive functions in mitochondria, its function as a scaffold in inflammation-enhanced platelet activation, and its contributions to endothelial permeability and calcium efflux from endoplasmic reticulum. In this review, we will summarize the molecular and cellular biology of JAK/STAT3 signaling and its functions under basal and stress conditions, which are adaptive, and then review maladaptive JAK/STAT3 signaling in animals and humans that lead to disease, as well as recent attempts to modulate them to treat these diseases. In addition, we will discuss how consideration of the noncanonical and stress-related functions of STAT3 cannot be ignored in efforts to target the canonical functions of STAT3, if the goal is to develop drugs that are not only effective but safe. SIGNIFICANCE STATEMENT: Key biological functions of Janus kinase (JAK)/signal transducer and activator of transcription (STAT)3 signaling can be delineated into two broad categories: those essential for normal cell and organ development and those activated in response to stress that are adaptive. Persistent or dysregulated JAK/STAT3 signaling, however, is maladaptive and contributes to many diseases, including diseases characterized by chronic inflammation and fibrosis, and cancer. A comprehensive understanding of JAK/STAT3 signaling in normal development, and in adaptive and maladaptive responses to stress, is essential for the continued development of safe and effective therapies that target this signaling pathway.
Collapse
Affiliation(s)
- Uddalak Bharadwaj
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Moses M Kasembeli
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Prema Robinson
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - David J Tweardy
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
25
|
Park M, Vaikari VP, Lam AT, Zhang Y, MacKay JA, Alachkar H. Anti-FLT3 nanoparticles for acute myeloid leukemia: Preclinical pharmacology and pharmacokinetics. J Control Release 2020; 324:317-329. [PMID: 32428520 PMCID: PMC7473778 DOI: 10.1016/j.jconrel.2020.05.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 12/20/2022]
Abstract
FLT3 receptor is an important therapeutic target in acute myeloid leukemia due to high incidence of mutations associated with poor clinical outcome. Targeted therapies against the FLT3 receptor, including small-molecule FLT3 tyrosine kinase inhibitors (TKIs) and anti-FLT3 antibodies, have demonstrated promising preclinical and even clinical efficacy. Yet, even with the current FDA approval for two FLT3 inhibitors, these modalities were unable to cure AML or significantly extend the lives of patients with a common mutation called FLT3-ITD. While FLT3 is a viable target, the approaches to inhibit its activity were inadequate. To develop a new modality for targeting FLT3, our team engineered an α-FLT3-A192 fusion protein composed of a single chain variable fragment antibody conjugated with an elastin-like polypeptide. These fusion proteins assemble into multi-valent nanoparticles with excellent stability and pharmacokinetic properties as well as in vitro and in vivo pharmacological activity in cellular and xenograft murine models of AML. In conclusion, α-FLT3-A192 fusions appear to be a viable new modality for targeting FLT3 in AML and warrant further preclinical development to bring it into the clinic.
Collapse
Affiliation(s)
- Mincheol Park
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States
| | - Vijaya Pooja Vaikari
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States
| | - Albert T Lam
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States
| | - Yong Zhang
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States; USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, United States; Department of Chemistry, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA 90089, United States; Research Center for Liver Diseases, University of Southern California, Los Angeles, CA 90089, United States
| | - John Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States; Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, United States; Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, United States
| | - Houda Alachkar
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, United States; USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, United States.
| |
Collapse
|
26
|
Yu J, Zhou Z, Wei Z, Wu J, OuYang J, Huang W, He Y, Zhang C. FYN promotes gastric cancer metastasis by activating STAT3-mediated epithelial-mesenchymal transition. Transl Oncol 2020; 13:100841. [PMID: 32763503 PMCID: PMC7408597 DOI: 10.1016/j.tranon.2020.100841] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/01/2020] [Accepted: 07/19/2020] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer is one of the most lethal cancers worldwide. FYN, a gene that is differentially expressed in gastric cancer, is considered a critical metastasis regulator in several solid tumors, but its role in gastric cancer is still unclear. This study aimed to evaluate the role of FYN and test whether FYN promotes migration and invasion of gastric cancer cells in vitro and in vivo via STAT3 signaling. FYN was overexpressed in gastric cancer and positively correlated with metastasis. FYN knockdown significantly decreased cancer cell migration and invasion, whereas FYN overexpression increased cancer migration and invasion. Genetic inhibition of FYN decreased the number of metastatic lung nodules in vivo. Several epithelial-mesenchymal transition markers were positively correlated with FYN expression, indicative of FYN involvement in this transition. Furthermore, gene set enrichment analysis of a Cancer Genome Atlas dataset revealed that the STAT3 signaling pathway was positively correlated with FYN expression. STAT3 inhibition reversed the FYN-mediated epithelial-mesenchymal transition and suppressed metastasis. In conclusion, FYN promotes gastric cancer metastasis possibly by activating STAT3-mediated epithelial mesenchymal transition and may be a novel therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Jie Yu
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan 2(nd) Road, Guangzhou, Guangdong 510080, China
| | - ZhiJun Zhou
- Department of Gastrointestinal Surgery, the Seventh Affiliated Hospital of Sun Yat-sen University, 628 Zhenyuan Road, Shenzhen, Guangdong 518000, China
| | - ZheWei Wei
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan 2(nd) Road, Guangzhou, Guangdong 510080, China
| | - Jing Wu
- Department of Gastrointestinal Surgery, the Seventh Affiliated Hospital of Sun Yat-sen University, 628 Zhenyuan Road, Shenzhen, Guangdong 518000, China
| | - Jun OuYang
- Department of Gastrointestinal Surgery, the Seventh Affiliated Hospital of Sun Yat-sen University, 628 Zhenyuan Road, Shenzhen, Guangdong 518000, China
| | - WeiBin Huang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan 2(nd) Road, Guangzhou, Guangdong 510080, China
| | - YuLong He
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan 2(nd) Road, Guangzhou, Guangdong 510080, China; Department of Gastrointestinal Surgery, the Seventh Affiliated Hospital of Sun Yat-sen University, 628 Zhenyuan Road, Shenzhen, Guangdong 518000, China.
| | - ChangHua Zhang
- Department of Gastrointestinal Surgery, the Seventh Affiliated Hospital of Sun Yat-sen University, 628 Zhenyuan Road, Shenzhen, Guangdong 518000, China.
| |
Collapse
|
27
|
Gu Y, Mohammad IS, Liu Z. Overview of the STAT-3 signaling pathway in cancer and the development of specific inhibitors. Oncol Lett 2020; 19:2585-2594. [PMID: 32218808 PMCID: PMC7068531 DOI: 10.3892/ol.2020.11394] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022] Open
Abstract
Signal transducer and activator of transcription (STAT) proteins represent novel therapeutic targets for the treatment of cancer. In particular, STAT-3 serves critical roles in several cellular processes, including the cell cycle, cell proliferation, cellular apoptosis and tumorigenesis. Persistent activation of STAT-3 has been reported in a variety of cancer types, and a poor prognosis of cancer may be associated with the phosphorylation level of STAT-3. Furthermore, elevated STAT-3 activity has been demonstrated in a variety of mammalian cancers, both in vitro and in vivo. This indicates that STAT-3 serves an important role in the progression of numerous cancer types. A significant obstacle in developing STAT-3 inhibitors is the demonstration of the antitumor efficacy in in vivo systems and the lack of animal models for human tumors. Therefore, it is crucial to determine whether available STAT-3 inhibitors are suitable for clinical trials. Moreover, further preclinical studies are necessary to focus on the impact of STAT-3 inhibitors on tumor cells. When considering STAT-3 hyper-activation in human cancer, selective targeting to these proteins holds promise for significant advancement in cancer treatment. In the present study, advances in our knowledge of the structure of STAT-3 protein and its regulatory mechanisms are summarized. Moreover, the STAT-3 signaling pathway and its critical role in malignancy are discussed, in addition to the development of STAT-3 inhibitors in various cancer types.
Collapse
Affiliation(s)
- Yuchen Gu
- Department of Pharmacy, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, P.R. China.,College of Pharmacy, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Imran Shair Mohammad
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Zhe Liu
- Department of Pharmacy, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, P.R. China.,College of Pharmacy, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| |
Collapse
|
28
|
Verhoeven Y, Tilborghs S, Jacobs J, De Waele J, Quatannens D, Deben C, Prenen H, Pauwels P, Trinh XB, Wouters A, Smits EL, Lardon F, van Dam PA. The potential and controversy of targeting STAT family members in cancer. Semin Cancer Biol 2020; 60:41-56. [DOI: 10.1016/j.semcancer.2019.10.002] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/30/2019] [Accepted: 10/04/2019] [Indexed: 12/13/2022]
|
29
|
Mohammadi Kian M, Salemi M, Bahadoran M, Haghi A, Dashti N, Mohammadi S, Rostami S, Chahardouli B, Babakhani D, Nikbakht M. Curcumin Combined with Thalidomide Reduces Expression of STAT3 and Bcl-xL, Leading to Apoptosis in Acute Myeloid Leukemia Cell Lines. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:185-194. [PMID: 32021103 PMCID: PMC6970263 DOI: 10.2147/dddt.s228610] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 12/16/2019] [Indexed: 12/29/2022]
Abstract
Introduction Acute myeloid leukemia (AML) is a type of blood disorder that exhibits uncontrolled growth and reduced ability to undergo apoptosis. Signal transducer and activator of transcription 3 (STAT3) is a family member of transcription factors which promotes carcinogenesis in most human cancers. This effect on AML is accomplished through deregulation of several critical genes, such as B cell lymphoma-extra-large (BCL-XL) which is anti-apoptotic protein. The aim of this study was to evaluate the effect of curcumin (CUR) and thalidomide (THAL) on apoptosis induction and also the alteration of the mRNA expression level of STAT3 and BCL-XL mRNA on AML cell line compounds. Methods The growth inhibitory effects of CUR and THAL and their combination were measured by MTT assay in U937 and KG-1 cell lines. The rates of apoptosis induction and cell cycle analysis were measured by concurrent staining with Annexin V and PI. The mRNA expression level of STAT3 and BCL-XL was evaluated by Real-Time PCR. Results CUR inhibited proliferation and induced apoptosis in both KG-1 and U937 cells and this effect increased by combination with THAL. The expression level of STAT3 and BCL-XL was significantly down-regulated in KG-1 cells after treatment by CUR and THAL and their combination. Conclusion Overall, our findings suggested that down-regulation of STAT3 and BCL-XL mRNA expression in response to CUR and THAL treatment lead to inhibition of cell growth and induction of apoptosis.
Collapse
Affiliation(s)
- Mahnaz Mohammadi Kian
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdieh Salemi
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Bahadoran
- Department of Biochemistry, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Atousa Haghi
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Young Researchers & Elite Club Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nasrin Dashti
- Department of Medical Laboratory Sciences, School of Allied Health Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Mohammadi
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahrbano Rostami
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahram Chahardouli
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Babakhani
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Nikbakht
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Huang B, He A, Zhang P, Ma X, Yang Y, Wang J, Wang J, Zhang W. Targeted silencing of genes related to acute monocytic leukaemia by CpG(B)-MLAA-34 siRNA conjugates. J Drug Target 2019; 28:516-524. [PMID: 31718329 DOI: 10.1080/1061186x.2019.1689397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Acute monocytic leukaemia (AML-M5) associated antigen-34 (MLAA-34) is a novel antigen overexpressed in patients with acute monocytic leukaemia. RNA interference is a promising therapy in oncology, especially for refractory acute leukaemia. In this study, we delivered MLAA-34 siRNA into AML-M5 THP-1 cells using CpG(B)-MLAA-34 siRNA conjugates, in the absence of any other transfection reagent. The uptake efficiency and the rate of apoptosis were measured by using flow cytometry. The level of relevant mRNAs was measured by quantitative PCR. THP-1 cell invasion was assessed by transwell assay. Protein expression was analysed by western blotting. The spleen and liver of AML-M5 nude mice were measured and weighted after euthanisation. Spleen sections were analysed by immunohistochemistry. We found that MLAA-34 siRNA was successfully delivered into THP-1 cells and induced MLAA-34 gene silencing via the blockade of JAK2/STAT3 and Wnt/-catenin signalling pathways. In addition, CpG(B)-MLAA-34 siRNA upregulated Gsk3β protein expression, resulting in retraining of the JAK2/STAT3 and Wnt/β-catenin signalling pathways. Importantly, CpG(B)-MLAA-34 siRNA reduced the survival and invasiveness of THP-1 cells. We further demonstrated that CAB39L was effectively downregulated by CpG(B)-MLAA-34 siRNA in vivo. These findings suggested CpG(B)-MLAA-34 siRNA conjugates may provide a novel therapeutic strategy for acute monocytic leukaemia.
Collapse
Affiliation(s)
- Bingqiao Huang
- Department of Haematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Aili He
- Department of Haematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Pengyu Zhang
- Department of Haematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaorong Ma
- Department of Haematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yun Yang
- Department of Haematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jianli Wang
- Department of Haematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Wang
- Department of Haematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wanggang Zhang
- Department of Haematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
31
|
Saw PE, Song EW. Phage display screening of therapeutic peptide for cancer targeting and therapy. Protein Cell 2019; 10:787-807. [PMID: 31140150 PMCID: PMC6834755 DOI: 10.1007/s13238-019-0639-7] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/21/2019] [Indexed: 12/14/2022] Open
Abstract
Recently, phage display technology has been announced as the recipient of Nobel Prize in Chemistry 2018. Phage display technique allows high affinity target-binding peptides to be selected from a complex mixture pool of billions of displayed peptides on phage in a combinatorial library and could be further enriched through the biopanning process; proving to be a powerful technique in the screening of peptide with high affinity and selectivity. In this review, we will first discuss the modifications in phage display techniques used to isolate various cancer-specific ligands by in situ, in vitro, in vivo, and ex vivo screening methods. We will then discuss prominent examples of solid tumor targeting-peptides; namely peptide targeting tumor vasculature, tumor microenvironment (TME) and over-expressed receptors on cancer cells identified through phage display screening. We will also discuss the current challenges and future outlook for targeting peptide-based therapeutics in the clinics.
Collapse
Affiliation(s)
- Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Er-Wei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| |
Collapse
|
32
|
Natural Sesquiterpene Lactones Enhance Chemosensitivity of Tumor Cells through Redox Regulation of STAT3 Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4568964. [PMID: 31781335 PMCID: PMC6855087 DOI: 10.1155/2019/4568964] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/07/2019] [Accepted: 09/05/2019] [Indexed: 12/18/2022]
Abstract
STAT3 is a nuclear transcription factor that regulates genes involved in cell cycle, cell survival, and immune response. Although STAT3 activation drives cells to physiological response, its deregulation is often associated with the development and progression of many solid and hematological tumors as well as with drug resistance. STAT3 is a redox-sensitive protein, and its activation state is related to intracellular GSH levels. Under oxidative conditions, STAT3 activity is regulated by S-glutathionylation, a reversible posttranslational modification of cysteine residues. Compounds able to suppress STAT3 activation and, on the other hand, to modulate intracellular redox homeostasis may potentially improve cancer treatment outcome. Nowadays, about 35% of commercial drugs are natural compounds that derive from plant extracts used in phytotherapy and traditional medicine. Sesquiterpene lactones are an interesting chemical group of plant-derived compounds often employed in traditional medicine against inflammation and cancer. This review focuses on sesquiterpene lactones able to downmodulate STAT3 signaling leading to an antitumor effect and correlates the anti-STAT3 activity with their ability to decrease GSH levels in cancer cells. These properties make them lead compounds for the development of a new therapeutic strategy for cancer treatment.
Collapse
|
33
|
Joyce MA, Berry-Wynne KM, dos Santos T, Addison WR, McFarlane N, Hobman T, Tyrrell DL. HCV and flaviviruses hijack cellular mechanisms for nuclear STAT2 degradation: Up-regulation of PDLIM2 suppresses the innate immune response. PLoS Pathog 2019; 15:e1007949. [PMID: 31374104 PMCID: PMC6677295 DOI: 10.1371/journal.ppat.1007949] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 06/29/2019] [Indexed: 12/22/2022] Open
Abstract
Host encounters with viruses lead to an innate immune response that must be rapid and broadly targeted but also tightly regulated to avoid the detrimental effects of unregulated interferon expression. Viral stimulation of host negative regulatory mechanisms is an alternate method of suppressing the host innate immune response. We examined three key mediators of the innate immune response: NF-KB, STAT1 and STAT2 during HCV infection in order to investigate the paradoxical induction of an innate immune response by HCV despite a multitude of mechanisms combating the host response. During infection, we find that all three are repressed only in HCV infected cells but not in uninfected bystander cells, both in vivo in chimeric mouse livers and in cultured Huh7.5 cells after IFNα treatment. We show here that HCV and Flaviviruses suppress the innate immune response by upregulation of PDLIM2, independent of the host interferon response. We show PDLIM2 is an E3 ubiquitin ligase that also acts to stimulate nuclear degradation of STAT2. Interferon dependent relocalization of STAT1/2 to the nucleus leads to PDLIM2 ubiquitination of STAT2 but not STAT1 and the proteasome-dependent degradation of STAT2, predominantly within the nucleus. CRISPR/Cas9 knockout of PDLIM2 results in increased levels of STAT2 following IFNα treatment, retention of STAT2 within the nucleus of HCV infected cells after IFNα stimulation, increased interferon response, and increased resistance to infection by several flaviviruses, indicating that PDLIM2 is a global regulator of the interferon response. The response of cells to an invading pathogen must be swift and well controlled because of the detrimental effects of chronic inflammation. However, viruses often hijack host control mechanisms. HCV and flaviviruses are known to suppress the innate immune response in cells by a variety of mechanisms. This study clarifies and expands a specific cellular mechanism for global control of the antiviral response after the induction of interferon expression. It shows how several viruses hijack this control mechanism to suppress the innate interferon response.
Collapse
Affiliation(s)
- Michael A. Joyce
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
- * E-mail: (MAJ); (DLT)
| | - Karyn M. Berry-Wynne
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Theodore dos Santos
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - William R. Addison
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Nicola McFarlane
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Tom Hobman
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada
| | - D. Lorne Tyrrell
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
- * E-mail: (MAJ); (DLT)
| |
Collapse
|
34
|
Wang H, Zhang Y, Xia F, Zhang W, Chen P, Yang G. Protective effect of silencing Stat1 on high glucose-induced podocytes injury via Forkhead transcription factor O1-regulated the oxidative stress response. BMC Mol Cell Biol 2019; 20:27. [PMID: 31337338 PMCID: PMC6652005 DOI: 10.1186/s12860-019-0209-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 07/09/2019] [Indexed: 02/07/2023] Open
Abstract
Background Podocyte plays an important role in maintaining the integrity and function of the glomerular filtration barrier. Various studies reported that forkhead transcription factor (Fox) O1 played a key role in anti-oxidative signaling. This study aimed to investigate the role of Stat1 in high glucose (HG) -induced podocyte injury. Methods Under normal glucose, hypertonic and HG stimulated podocyte conditions, cell counting kit-8 (CCK-8) assay, flow cytometry and western blot and quantitative real-time polymerase chain reaction (qRT-PCR) were respectively carried out to determine cell viability, apoptosis, reactive oxygen species (ROS) production and related genes expressions. We then respectively used silent Stat1, simultaneous silencing Stat1 and FoxO1 and over-expression of FoxO1, to observe whether they/it could reverse the damage of podocytes induced by HG. Results High glucose attenuated cell survival and promoted cell apoptosis in MPC-5 cells at the same time, and it was also observed to promote the protein expression of Stat1 and the FoxO1 expression inhibition. Silencing Stat1 could reverse HG-induced podocytes injury. Specifically, siStat1 increased cell viability, inhibited cell apoptosis and attenuated ROS level in a high-glucose environment. Cleaved caspase-3 and pro-apoptosis protein Bax was significantly down-regulated, and anti-apoptosis protein Bcl-2 was up-regulated by siStat1. The antioxidant genes Catalase, MnSOD, NQO1 and HO1 were up-regulated by siStat1. We found that silencing FoxO1 reversed the protective effect of siStat1 on the HG-induced podocytes injury. Conclusions Silencing Stat1 could reverse the effects of high glucose-triggered low cell viability, cell apoptosis and ROS release and the functions of Stat1 might be involved in FoxO1 mediated-oxidative stress in nucleus.
Collapse
Affiliation(s)
- Hongkun Wang
- Department of Nephrology, The First Affiliated Hospital of Baotou Medical College Inner Mongolia University of Science and Technology, Baotou, China
| | - Yanhui Zhang
- Department of Nephrology, The First Affiliated Hospital of Baotou Medical College Inner Mongolia University of Science and Technology, Baotou, China
| | - Fangfang Xia
- Department of Nephrology, North Hospital, Baotou, China
| | - Wei Zhang
- Central Laboratory, The First Affiliated Hospital of Baotou Medical College Inner Mongolia University of Science and Technology, No.41 Linyin Road, Kundulun District, Baotou, 014010, Inner Mongolia, China
| | - Peng Chen
- Department of Nutriology, The First Affiliated Hospital of Baotou Medical College Inner Mongolia University of Science and Technology, Baotou, China
| | - Guoan Yang
- Central Laboratory, The First Affiliated Hospital of Baotou Medical College Inner Mongolia University of Science and Technology, No.41 Linyin Road, Kundulun District, Baotou, 014010, Inner Mongolia, China.
| |
Collapse
|
35
|
Duval R, Bui LC, Mathieu C, Nian Q, Berthelet J, Xu X, Haddad I, Vinh J, Dupret JM, Busi F, Guidez F, Chomienne C, Rodrigues-Lima F. Benzoquinone, a leukemogenic metabolite of benzene, catalytically inhibits the protein tyrosine phosphatase PTPN2 and alters STAT1 signaling. J Biol Chem 2019; 294:12483-12494. [PMID: 31248982 DOI: 10.1074/jbc.ra119.008666] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/21/2019] [Indexed: 12/12/2022] Open
Abstract
Protein tyrosine phosphatase, nonreceptor type 2 (PTPN2) is mainly expressed in hematopoietic cells, where it negatively regulates growth factor and cytokine signaling. PTPN2 is an important regulator of hematopoiesis and immune/inflammatory responses, as evidenced by loss-of-function mutations of PTPN2 in leukemia and lymphoma and knockout mice studies. Benzene is an environmental chemical that causes hematological malignancies, and its hematotoxicity arises from its bioactivation in the bone marrow to electrophilic metabolites, notably 1,4-benzoquinone, a major hematotoxic benzene metabolite. Although the molecular bases for benzene-induced leukemia are not well-understood, it has been suggested that benzene metabolites alter topoisomerases II function and thereby significantly contribute to leukemogenesis. However, several studies indicate that benzene and its hematotoxic metabolites may also promote the leukemogenic process by reacting with other targets and pathways. Interestingly, alterations of cell-signaling pathways, such as Janus kinase (JAK)/signal transducer and activator of transcription (STAT), have been proposed to contribute to benzene-induced malignant blood diseases. We show here that 1,4-benzoquinone directly impairs PTPN2 activity. Mechanistic and kinetic experiments with purified human PTPN2 indicated that this impairment results from the irreversible formation (k inact = 645 m-1·s-1) of a covalent 1,4-benzoquinone adduct at the catalytic cysteine residue of the enzyme. Accordingly, cell experiments revealed that 1,4-benzoquinone exposure irreversibly inhibits cellular PTPN2 and concomitantly increases tyrosine phosphorylation of STAT1 and expression of STAT1-regulated genes. Our results provide molecular and cellular evidence that 1,4-benzoquinone covalently modifies key signaling enzymes, implicating it in benzene-induced malignant blood diseases.
Collapse
Affiliation(s)
- Romain Duval
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | - Linh-Chi Bui
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | - Cécile Mathieu
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | - Qing Nian
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | | | - Ximing Xu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Iman Haddad
- ESPCI Paris, PSL Université, USR 3149, CNRS, F-75005 Paris, France
| | - Joelle Vinh
- ESPCI Paris, PSL Université, USR 3149, CNRS, F-75005 Paris, France
| | | | - Florent Busi
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | - Fabien Guidez
- Université de Paris, Institut de Recherche Saint-Louis, UMRS 1131, INSERM, F-75010 Paris, France
| | - Christine Chomienne
- Université de Paris, Institut de Recherche Saint-Louis, UMRS 1131, INSERM, F-75010 Paris, France; Service de Biologie Cellulaire, Assistance Publique des Hôpitaux de Paris (AP-HP), Hôpital Saint Louis, F-75010 Paris, France
| | | |
Collapse
|
36
|
Nian Q, Berthelet J, Zhang W, Bui LC, Liu R, Xu X, Duval R, Ganesan S, Leger T, Chomienne C, Busi F, Guidez F, Dupret JM, Rodrigues Lima F. T-Cell Protein Tyrosine Phosphatase Is Irreversibly Inhibited by Etoposide-Quinone, a Reactive Metabolite of the Chemotherapy Drug Etoposide. Mol Pharmacol 2019; 96:297-306. [DOI: 10.1124/mol.119.116319] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 06/07/2019] [Indexed: 11/22/2022] Open
|
37
|
Butturini E, Boriero D, Carcereri de Prati A, Mariotto S. STAT1 drives M1 microglia activation and neuroinflammation under hypoxia. Arch Biochem Biophys 2019; 669:22-30. [PMID: 31121156 DOI: 10.1016/j.abb.2019.05.011] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/07/2019] [Accepted: 05/15/2019] [Indexed: 12/25/2022]
Abstract
Microglia are resident immune cells that act as the first active defence in the central nervous system. These cells constantly monitor the tissue microenvironment and rapidly react in response to hypoxia, infection and injuries. Hypoxia in the brain has been detected in several neurodegenerative disorders such as Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease and Huntington's disease. Hypoxic conditions activate microglia cells towards M1 phenotype resulting in oxidative stress and the release of pro-inflammatory cytokines. Recently, we have demonstrated that oxidative stress induces S-glutathionylation of the STAT1 and hyper-activates its signaling in microglia BV2 cells pointing out the importance of this transcription factor in neuroinflammation. In this paper we analyse the cellular mechanisms that drive M1 microglia activation in BV2 cells in response to hypoxia correlating it to STAT1 activation. The analysis of the molecular mechanism of STAT1 signaling reveals that hypoxia generates oxidative stress and induces both phosphorylation and S-glutathionylation of STAT1 that are responsible of its aberrant activation. The silencing of STAT1 protein expression counteracts hypoxia-M1 microglia phenotype suggesting the strong link between hypoxia-STAT1 and STAT1-microglia activation.
Collapse
Affiliation(s)
- Elena Butturini
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, Verona, Italy.
| | - Diana Boriero
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, Verona, Italy
| | - Alessandra Carcereri de Prati
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, Verona, Italy
| | - Sofia Mariotto
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, Verona, Italy
| |
Collapse
|
38
|
YL064 directly inhibits STAT3 activity to induce apoptosis of multiple myeloma cells. Cell Death Discov 2018; 4:44. [PMID: 30302278 PMCID: PMC6170385 DOI: 10.1038/s41420-018-0108-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 09/02/2018] [Indexed: 01/07/2023] Open
Abstract
Aberrant activation of signal transducer and activator of transcription 3 (STAT3) plays a critical role in the proliferation and survival of multiple myeloma. And inactivation of STAT3 is considered a promising strategy for the treatment of multiple myeloma. Here we show that the sinomenine derivative YL064 could selectively reduce the cell viability of multiple myeloma cell lines and primary multiple myeloma cells. Moreover, YL064 also induces cell death of myeloma cells in the presence of stromal cells. Western blot analysis showed that YL064 inhibited the constitutive activation and IL-6-induced activation of STAT3, reflected by the decreased phosphorylation of STAT3 on Tyr705. Consistent with this, YL064 inhibited the nuclear translocation of STAT3 and the expression of STAT3 target genes, such as cyclin D1 and Mcl-1. Using biotin- and FITC-labeled YL064, we found that YL064 could pull-down STAT3 from myeloma cells and colocalized with STAT3, suggesting that YL064 directly targets STAT3. Cellular thermal shift assay further demonstrated the engagement of YL064 to STAT3 in cells. Molecular docking studies indicated that YL064 may interact with STAT3 in its SH2 domain, thereby inhibiting the dimerization of STAT3. Finally, YL064 inhibited the growth of human myeloma xenograft in vivo. Taken together, this study demonstrated that YL064 may be a promising candidate compound for the treatment of multiple myeloma by directly targeting STAT3.
Collapse
|
39
|
Arora L, Kumar AP, Arfuso F, Chng WJ, Sethi G. The Role of Signal Transducer and Activator of Transcription 3 (STAT3) and Its Targeted Inhibition in Hematological Malignancies. Cancers (Basel) 2018; 10:cancers10090327. [PMID: 30217007 PMCID: PMC6162647 DOI: 10.3390/cancers10090327] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 12/22/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3), a member of the STAT protein family, can be phosphorylated by receptor-associated Janus kinases (JAKs) in response to stimulation by cytokines and growth factors. It forms homo- or heterodimers that can translocate to the cell nucleus where they act as transcription activators. Constitutive activation of STAT3 has been found to be associated with initiation and progression of various cancers. It can exert proliferative as well as anti-apoptotic effects. This review focuses on the role of STAT3 in pathogenesis i.e., proliferation, differentiation, migration, and apoptosis of hematological malignancies viz. leukemia, lymphoma and myeloma, and briefly highlights the potential therapeutic approaches developed against STAT3 activation pathway.
Collapse
Affiliation(s)
- Loukik Arora
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
- Cancer Science Institute of Singapore, Centre for Translational Medicine, 14 Medical Drive, #11-01M, Singapore 117599, Singapore.
- Medical Science Cluster, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia.
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore.
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
| | - Wee Joo Chng
- Cancer Science Institute of Singapore, Centre for Translational Medicine, 14 Medical Drive, #11-01M, Singapore 117599, Singapore.
- Department of Hematology-Oncology, National University Cancer Institute, National University Health System, Singapore 119074, Singapore.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6009, Australia.
| |
Collapse
|
40
|
Alasseiri M, Ahmed AU, Williams BRG. Mechanisms and consequences of constitutive activation of integrin-linked kinase in acute myeloid leukemia. Cytokine Growth Factor Rev 2018; 43:1-7. [PMID: 29903521 DOI: 10.1016/j.cytogfr.2018.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 05/31/2018] [Accepted: 06/05/2018] [Indexed: 12/22/2022]
Abstract
Integrin-linked kinase (ILK) has emerged as a critical adaptor and mediator protein in cell signaling pathways that is commonly deregulated in acute myeloid leukemia (AML). This has led to the expectation that therapeutic targeting of ILK may be a useful option in treating leukemia. Although ILK can regulate many cellular processes, including cell differentiation, survival, migration, apoptosis and production of pro-inflammatory cytokines, its role in promoting AML is still unclear. However, its ability to mediate phosphorylation and regulate the important hematopoietic stem cell regulators protein kinase B (AKT) and glycogen synthase kinase-3β supports ILK as an attractive target for the development of novel anticancer therapeutics. In this review, we summarize the existing knowledge of ILK signaling and its impact on cytokines, paying particular attention to the relevance of ILK signaling in AML. We also discuss the rationale for targeting ILK in the treatment of AML and conclude with perspectives on the future of ILK-targeted therapy in AML.
Collapse
Affiliation(s)
- Mohammed Alasseiri
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC, 3168, Australia; Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Afsar U Ahmed
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC, 3168, Australia
| | - Bryan R G Williams
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC, 3168, Australia.
| |
Collapse
|
41
|
A novel small molecular STAT3 inhibitor, 5Br-6b, induces apoptosis and inhibits migration in colorectal cancer cells. Anticancer Drugs 2018; 29:402-410. [DOI: 10.1097/cad.0000000000000605] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
42
|
Li CW, Lai TY, Chen BS. Changes of signal transductivity and robustness of gene regulatory network in the carcinogenesis of leukemic subtypes via microarray sample data. Oncotarget 2018; 9:23636-23660. [PMID: 29805763 PMCID: PMC5955113 DOI: 10.18632/oncotarget.25318] [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: 11/11/2016] [Accepted: 04/11/2018] [Indexed: 11/25/2022] Open
Abstract
Mutation accumulation and epigenetic alterations in genes are important for carcinogenesis. Because leukemogenesis-related signal pathways have been investigated and microarray sample data have been produced in acute myeloid leukemia (AML), myelodysplastic syndromes (MDS) and normal cells, systems analysis in coupling pathways becomes possible. Based on system modeling and identification, we could construct the coupling pathways and their associated gene regulatory networks using microarray sample data. By applying system theory to the estimated system model in coupling pathways, we can then obtain transductivity sensitivity, basal sensitivity and error sensitivity of each protein to identify the potential impact of genetic mutations, epigenetic alterations and the coupling of other pathways from the perspective of energy, respectively. By comparing the results in AML, MDS and normal cells, we investigated the potential critical genetic mutations and epigenetic alterations that activate or repress specific cellular functions to promote MDS or AML leukemogenesis. We suggested that epigenetic modification of β-catenin and signal integration of CSLs, AP-2α, STATs, c-Jun and β-catenin could contribute to cell proliferation at AML and MDS. Epigenetic regulation of ERK and genetic mutation of p53 could lead to the repressed apoptosis, cell cycle arrest and DNA repair in leukemic cells. Genetic mutation of JAK, epigenetic regulation of ERK, and signal integration of C/EBPα could result in the promotion of MDS cell differentiation. According to the results, we proposed three drugs, decitabine, genistein, and monorden for preventing AML leukemogenesis, while three drugs, decitabine, thalidomide, and geldanamycin, for preventing MDS leukemogenesis.
Collapse
Affiliation(s)
- Cheng-Wei Li
- Laboratory of Control and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Tzu-Ying Lai
- Laboratory of Control and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Bor-Sen Chen
- Laboratory of Control and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
43
|
Discovery of new benzensulfonamide derivatives as tripedal STAT3 inhibitors. Eur J Med Chem 2018; 151:752-764. [DOI: 10.1016/j.ejmech.2018.03.053] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 01/08/2023]
|
44
|
Hu W, Lv J, Han M, Yang Z, Li T, Jiang S, Yang Y. STAT3: The art of multi-tasking of metabolic and immune functions in obesity. Prog Lipid Res 2018; 70:17-28. [DOI: 10.1016/j.plipres.2018.04.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/04/2018] [Accepted: 04/06/2018] [Indexed: 02/07/2023]
|
45
|
Abstract
The IL-6/JAK/STAT3 pathway is aberrantly hyperactivated in many types of cancer, and such hyperactivation is generally associated with a poor clinical prognosis. In the tumour microenvironment, IL-6/JAK/STAT3 signalling acts to drive the proliferation, survival, invasiveness, and metastasis of tumour cells, while strongly suppressing the antitumour immune response. Thus, treatments that target the IL-6/JAK/STAT3 pathway in patients with cancer are poised to provide therapeutic benefit by directly inhibiting tumour cell growth and by stimulating antitumour immunity. Agents targeting IL-6, the IL-6 receptor, or JAKs have already received FDA approval for the treatment of inflammatory conditions or myeloproliferative neoplasms and for the management of certain adverse effects of chimeric antigen receptor T cells, and are being further evaluated in patients with haematopoietic malignancies and in those with solid tumours. Novel inhibitors of the IL-6/JAK/STAT3 pathway, including STAT3-selective inhibitors, are currently in development. Herein, we review the role of IL-6/JAK/STAT3 signalling in the tumour microenvironment and the status of preclinical and clinical investigations of agents targeting this pathway. We also discuss the potential of combining IL-6/JAK/STAT3 inhibitors with currently approved therapeutic agents directed against immune-checkpoint inhibitors.
Collapse
Affiliation(s)
- Daniel E. Johnson
- Department of Otolaryngology – Head and Neck Surgery, University of California, San Francisco, CA, USA
| | - Rachel A. O’Keefe
- Department of Otolaryngology – Head and Neck Surgery, University of California, San Francisco, CA, USA
| | - Jennifer R. Grandis
- Department of Otolaryngology – Head and Neck Surgery, University of California, San Francisco, CA, USA
| |
Collapse
|
46
|
Butturini E, Cozzolino F, Boriero D, Carcereri de Prati A, Monti M, Rossin M, Canetti D, Cellini B, Pucci P, Mariotto S. S-glutathionylation exerts opposing roles in the regulation of STAT1 and STAT3 signaling in reactive microglia. Free Radic Biol Med 2018; 117:191-201. [PMID: 29427792 DOI: 10.1016/j.freeradbiomed.2018.02.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 02/02/2018] [Accepted: 02/03/2018] [Indexed: 10/18/2022]
Abstract
STAT1 and STAT3 are two transcription factors involved in a lot of cellular functions such as immune response, proliferation, apoptosis, and cell survival. A number of literature evidences described a yin-yang relationship between activation of STAT1 and STAT3 in neurodegenerative disorders where STAT1 exerts a pro-apoptotic effect whereas STAT3 shows neuroprotective properties through the inhibition of apoptosis. Although the role of oxidative-stress in the pathogenesis of neurodegeneration is clearly described, its influence in the regulation of these pathways is poorly understood. Herein, we demonstrate that H2O2 rapidly induces phosphorylation of STAT1 whereas it is not able to influence phosphorylation of STAT3 in mouse microglia BV2 cells. The analysis of the molecular mechanism of STATs signaling reveals that H2O2 induces S-glutathionylation of both STAT1 and STAT3. The same post-translational event exerts an opposing role in the regulation of STAT1 and STAT3 signaling. These data not only confirm redox sensibility of STAT3 signaling but also reveal for the first time that STAT1 is susceptible to redox regulation. A deep study of the molecular mechanism of STAT1 redox regulation, identifies Cys324 and Cys492 as the main targets of S-glutathionylation and confirms that S-glutathionylation does not impair JAK2 mediated STAT1 tyrosine phosphorylation. These results demonstrate that both phosphorylation and glutathionylation contribute to activation of STAT1 during oxidative stress and underline that the same post-translation event exerts an opposing role in the regulation of STAT1 and STAT3 signaling in microglia cells.
Collapse
Affiliation(s)
- Elena Butturini
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, Strada Le Grazie, 8, 37134 Verona, Italy
| | - Flora Cozzolino
- CEINGE Biotecnologie Avanzate and Department of Chemical Science, University of Naples "Federico II, Naples, Italy
| | - Diana Boriero
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, Strada Le Grazie, 8, 37134 Verona, Italy
| | - Alessandra Carcereri de Prati
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, Strada Le Grazie, 8, 37134 Verona, Italy
| | - Maria Monti
- CEINGE Biotecnologie Avanzate and Department of Chemical Science, University of Naples "Federico II, Naples, Italy
| | - Michele Rossin
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, Strada Le Grazie, 8, 37134 Verona, Italy; CEINGE Biotecnologie Avanzate and Department of Chemical Science, University of Naples "Federico II, Naples, Italy; Department of Experimental Medicine, University of Perugia, Perugia, Italy3
| | - Diana Canetti
- CEINGE Biotecnologie Avanzate and Department of Chemical Science, University of Naples "Federico II, Naples, Italy
| | - Barbara Cellini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy3
| | - Piero Pucci
- CEINGE Biotecnologie Avanzate and Department of Chemical Science, University of Naples "Federico II, Naples, Italy
| | - Sofia Mariotto
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, Strada Le Grazie, 8, 37134 Verona, Italy.
| |
Collapse
|
47
|
Yilmaz AF, Kaymaz B, Aktan Ç, Soyer N, Kosova B, Güneş A, Şahin F, Cömert M, Saydam G, Vural F. Determining expression of miRNAs that potentially regulate STAT5A and 5B in dasatinib-sensitive K562 cells. Turk J Biol 2017; 41:926-934. [PMID: 30814857 DOI: 10.3906/biy-1705-66] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In the era of tyrosine kinase inhibitors, resistance still constitutes a problem in chronic myeloid leukemia (CML) patients; thus, new pathway-specific inhibitors like miRNAs have become important in the treatment of refractory patients. There are no satisfying data regarding the miRNAs and anti-miRNA treatment targeting STAT5A and 5B. In this study, we first researched the effect of dasatinib on apoptosis in the CML cell line K562. The expressions of miRNAs possibly targeting both STAT5A and 5B were then determined. The down- and upregulation of the miRNAs were compared using the ΔΔCT method. At the last stage of the study, we used a new primer probe in order to validate the results. The level of hsa-miR-940 was decreased 4.4 times and the levels of hsa-miR-527 and hsa-miR-518a-5p were increased 12.1 and 8 times, respectively, in the dasatinib-treated group when compared to the control group. We detected similar results in the validation step. As a conclusion, we determined the expression profiles of miRNAs targeting STAT5A and 5B that had an important role in the pathogenesis of CML. The data obtained could lead to determining new therapeutic targets for CML patients.
Collapse
Affiliation(s)
- Asu Fergün Yilmaz
- Department of Hematology, İzmir Kâtip Çelebi University Atatürk Training and Research Hospital , İzmir , Turkey
| | - Burçin Kaymaz
- Department of Medical Biology, Ege University Hospital , İzmir , Turkey
| | - Çağdaş Aktan
- Department of Medical Biology, School of Medicine, Beykent University , İstanbul , Turkey
| | - Nur Soyer
- Department of Hematology, Internal Medicine, Ege University Hospital , İzmir , Turkey
| | - Buket Kosova
- Department of Medical Biology, Ege University Hospital , İzmir , Turkey
| | - Ajda Güneş
- Department of Hematology, Sivas Numune Hospital , Sivas , Turkey
| | - Fahri Şahin
- Department of Hematology, Internal Medicine, Ege University Hospital , İzmir , Turkey
| | - Melda Cömert
- Department of Hematology, Internal Medicine, İnönü University Hospital , Malatya , Turkey
| | - Güray Saydam
- Department of Hematology, Internal Medicine, Ege University Hospital , İzmir , Turkey
| | - Filiz Vural
- Department of Hematology, Internal Medicine, Ege University Hospital , İzmir , Turkey
| |
Collapse
|
48
|
Huang Y, Wang J, Cao F, Jiang H, Li A, Li J, Qiu L, Shen H, Chang W, Zhou C, Pan Y, Lu Y. SHP2 associates with nuclear localization of STAT3: significance in progression and prognosis of colorectal cancer. Sci Rep 2017; 7:17597. [PMID: 29242509 PMCID: PMC5730547 DOI: 10.1038/s41598-017-17604-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/28/2017] [Indexed: 12/11/2022] Open
Abstract
Tyrosine phosphatase SHP2, encoded by PTPN11, has been implicated in many physiologic and pathologic processes in neoplastic progression. However, controversies are emerging from many studies, indicating SHP2 has a dual role in different types of tumors. We aimed to explore the role of SHP2 in progression and prognosis of colorectal cancer (CRC). SHP2 inhibited CRC cell proliferation and migration, and the phosphorylation of STAT3 was negatively regulated by SHP2 in CRC. SHP2 and nuclear STAT3 were examined in 270 CRC tissues. SHP2 was significantly correlated with nuclear STAT3 (Spearman’s rho = −0.408, P ≤ 0.001). Based on Cox regression analysis, patients with high levels of SHP2 and low levels of nuclear STAT3 had longer disease-specific survival (DSS) (HR, 0.362; 95% CI, 0.165–0.794) and disease-free survival (DFS) (HR, 0.447; 95% CI, 0.227–0.877). Further, low levels of SHP2 and high levels of nuclear STAT3 were independently associated with adverse outcomes in the whole cohort (DFS; HR, 2.353; 95% CI, 1.199–4.619). These results suggest that combination of SHP2 and nuclear STAT3 is a strong prognostic predictor in CRC.
Collapse
Affiliation(s)
- Yan Huang
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Jie Wang
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Fuao Cao
- Department of colorectal surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Hailong Jiang
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - An Li
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Jianzhong Li
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Lei Qiu
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Hao Shen
- Department of Environmental Hygiene, Second Military Medical University, Shanghai, 200433, China
| | - Wenjun Chang
- Department of Environmental Hygiene, Second Military Medical University, Shanghai, 200433, China
| | - Chuanxiang Zhou
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Yamin Pan
- Department of Digestive Endoscopy, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yiming Lu
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
49
|
He MH, Zhang Q, Shu G, Lin JC, Zhao L, Liang XX, Yin L, Shi F, Fu HL, Yuan ZX. Dihydromyricetin sensitizes human acute myeloid leukemia cells to retinoic acid-induced myeloid differentiation by activating STAT1. Biochem Biophys Res Commun 2017; 495:1702-1707. [PMID: 29225170 DOI: 10.1016/j.bbrc.2017.12.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 12/06/2017] [Indexed: 11/30/2022]
Abstract
The success of all-trans retinoic acid (ATRA) in differentiation therapy for patients with acute promyelocytic leukemia (APL) highly encourages researches to apply a new combination therapy based on ATRA. Therefore, research strategies to further sensitize cells to retinoids are urgently needed. In this study, we showed that Dihydromyricetin (DMY), a 2,3-dihydroflavonol compound, exhibited a strong synergy with ATRA to promote APL NB4 cell differentiation. We observed that DMY sensitized the NB4 cells to ATRA-induced cell growth inhibition, CD11b expression, NBT reduction and myeloid regulator expression. PML-RARα might not be essential for DMY-enhanced differentiation when combined with ATRA, while the enhanced differentiation was dependent on the activation of p38-STAT1 signaling pathway. Taken together, our study is the first to evaluate the synergy of DMY and ATRA in NB4 cell differentiation and to assess new opportunities for the combination of DMY and ATRA as a promising approach for future differentiation therapy.
Collapse
Affiliation(s)
- Ming-Hui He
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Qiang Zhang
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Gang Shu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Ju-Chun Lin
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Ling Zhao
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Xiao-Xia Liang
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Lizi Yin
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Fei Shi
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Hua-Lin Fu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Zhi-Xiang Yuan
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| |
Collapse
|
50
|
Namasu CY, Katzerke C, Bräuer-Hartmann D, Wurm AA, Gerloff D, Hartmann JU, Schwind S, Müller-Tidow C, Hilger N, Fricke S, Christopeit M, Niederwieser D, Behre G. ABR, a novel inducer of transcription factor C/EBPα, contributes to myeloid differentiation and is a favorable prognostic factor in acute myeloid leukemia. Oncotarget 2017; 8:103626-103639. [PMID: 29262589 PMCID: PMC5732755 DOI: 10.18632/oncotarget.22093] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/22/2017] [Indexed: 11/29/2022] Open
Abstract
Active BCR related (ABR) gene deactivates ras-related C3 botulinum toxin substrate 1 (RAC1), which plays an essential role in regulating normal hematopoiesis and in leukemia. BCR gene, closely related to ABR, acts as a tumor suppressor in chronic myeloid leukemia and has overlapping functions with ABR. Evidence for a putative tumor suppressor role of ABR has been shown in several solid tumors, in which deletion of ABR is present. Our results show downregulation of ABR in AML. A block of ABR prevents myeloid differentiation and leads to repression of the myeloid transcription factor C/EBPα, a major regulator of myeloid differentiation and functionally impaired in leukemia. Conversely, stable overexpression of ABR enhances myeloid differentiation. Inactivation of the known ABR target RAC1 by treatment with the RAC1 inhibitor NSC23766 resulted in an increased expression of C/EBPα in primary AML samples and in AML cell lines U937 and MV4;11. Finally, AML patients with high ABR expression at diagnosis showed a significant longer overall survival and patients who respond to azacitidine therapy showed a significant higher ABR expression. This is the first report showing that ABR expression plays a critical role in both myelopoiesis and AML. Our data indicate the tumor suppressor potential of ABR and underline its potential role in leukemia therapeutic strategies.
Collapse
Affiliation(s)
| | - Christiane Katzerke
- Division of Hematology and Oncology, University Hospital Leipzig, Leipzig, Germany
| | | | | | - Dennis Gerloff
- Division of Dermatology and Venereology, University Hospital Halle, Halle, Germany
| | - Jens-Uwe Hartmann
- Division of Hematology and Oncology, University Hospital Leipzig, Leipzig, Germany
| | - Sebastian Schwind
- Division of Hematology and Oncology, University Hospital Leipzig, Leipzig, Germany
| | - Carsten Müller-Tidow
- Division of Hematology and Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Nadja Hilger
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Stephan Fricke
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Maximilian Christopeit
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dietger Niederwieser
- Division of Hematology and Oncology, University Hospital Leipzig, Leipzig, Germany
| | - Gerhard Behre
- Division of Hematology and Oncology, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|