1
|
Wei X, Wang W, Yin Q, Li H, Ahmed A, Ullah R, Li W, Jing L. In Vivo Chemical Screening in Zebrafish Embryos Identified FDA-Approved Drugs That Induce Differentiation of Acute Myeloid Leukemia Cells. Int J Mol Sci 2024; 25:7798. [PMID: 39063039 PMCID: PMC11277044 DOI: 10.3390/ijms25147798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Acute myeloid leukemia (AML) is characterized by the abnormal proliferation and differentiation arrest of myeloid progenitor cells. The clinical treatment of AML remains challenging. Promoting AML cell differentiation is a valid strategy, but effective differentiation drugs are lacking for most types of AML. In this study, we generated Tg(drl:hoxa9) zebrafish, in which hoxa9 overexpression was driven in hematopoietic cells and myeloid differentiation arrest was exhibited. Using Tg(drl:hoxa9) embryos, we performed chemical screening and identified four FDA-approved drugs, ethacrynic acid, khellin, oxcarbazepine, and alendronate, that efficiently restored myeloid differentiation. The four drugs also induced AML cell differentiation, with ethacrynic acid being the most effective. By an RNA-seq analysis, we found that during differentiation, ethacrynic acid activated the IL-17 and MAPK signaling pathways, which are known to promote granulopoiesis. Furthermore, we found that ethacrynic acid enhanced all-trans retinoic acid (ATRA)-induced differentiation, and both types of signaling converged on the IL-17/MAPK pathways. Inhibiting the IL-17/MAPK pathways impaired ethacrynic acid and ATRA-induced differentiation. In addition, we showed that ethacrynic acid is less toxic to embryogenesis and less disruptive to normal hematopoiesis than ATRA. Thus, the combination of ethacrynic acid and ATRA may have broader clinical applications. In conclusion, through zebrafish-aided screening, our study identified four drugs that can be repurposed to induce AML differentiation, thus providing new agents for AML therapy.
Collapse
Affiliation(s)
- Xiaona Wei
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (X.W.); (Q.Y.); (H.L.); (A.A.); (R.U.)
| | - Wei Wang
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China;
| | - Qianlan Yin
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (X.W.); (Q.Y.); (H.L.); (A.A.); (R.U.)
| | - Hongji Li
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (X.W.); (Q.Y.); (H.L.); (A.A.); (R.U.)
| | - Abrar Ahmed
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (X.W.); (Q.Y.); (H.L.); (A.A.); (R.U.)
| | - Rahat Ullah
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (X.W.); (Q.Y.); (H.L.); (A.A.); (R.U.)
| | - Wei Li
- Core Facility and Technical Service Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lili Jing
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (X.W.); (Q.Y.); (H.L.); (A.A.); (R.U.)
| |
Collapse
|
2
|
Wang W, An J, Zhao R, Geng X, Jiang W, Yan X, Jiang B. Nanozymes: a new approach for leukemia therapy. J Mater Chem B 2024; 12:2459-2470. [PMID: 38345341 DOI: 10.1039/d3tb02819d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Leukemia is a type of clonal disorder of hematopoietic stem and progenitor cells characterized by bone marrow failure, differentiation arrest, and lineage skewing. Despite leukemia being a complex disease and it being difficult to identify a single driving force, redox homeostasis, the balance between reactive oxygen species (ROS) producers and cellular antioxidant systems, is normally impaired during leukemogenesis. In this context, the modulation of ROS in leukemia cells can be harnessed for therapeutic purposes. Nanozymes are functional nanomaterials with enzyme-like characteristics, which address the intrinsic limitations of natural enzymes and exhibit great potential in synergistic antitumor therapy. Nanozymes possess catalytic activities (e.g., peroxidase-like activity, catalase-like activity, superoxide dismutase-like activity, and oxidase-like activity) to regulate ROS levels in vitro and in vivo, making them promising for leukemia therapy. On account of the rapid development of nanozymes recently, their application potentials in leukemia therapy are gradually being explored. To highlight the achievements of nanozymes in the leukemia field, this review summarizes the recent studies of nanozymes with anti-leukemia efficacy and the underlying mechanism. In addition, the challenges and prospects of nanozyme research in leukemia therapy are discussed.
Collapse
Affiliation(s)
- Wei Wang
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Jingyi An
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Runze Zhao
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Xin Geng
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Wei Jiang
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Xiyun Yan
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Nanozyme Laboratory in Zhongyuan, Zhengzhou, Henan, 451163, China
| | - Bing Jiang
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
- Nanozyme Laboratory in Zhongyuan, Zhengzhou, Henan, 451163, China
| |
Collapse
|
3
|
Dao Nyesiga G, Pool L, Englezou PC, Hylander T, Ohlsson L, Appelgren D, Sundstedt A, Tillerkvist K, Romedahl HR, Wigren M. Tolerogenic dendritic cells generated in vitro using a novel protocol mimicking mucosal tolerance mechanisms represent a potential therapeutic cell platform for induction of immune tolerance. Front Immunol 2023; 14:1045183. [PMID: 37901231 PMCID: PMC10613069 DOI: 10.3389/fimmu.2023.1045183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 08/25/2023] [Indexed: 10/31/2023] Open
Abstract
Dendritic cells (DCs) are mediators between innate and adaptive immunity and vital in initiating and modulating antigen-specific immune responses. The most important site for induction of tolerance is the gut mucosa, where TGF-β, retinoic acid, and aryl hydrocarbon receptors collaborate in DCs to induce a tolerogenic phenotype. To mimic this, a novel combination of compounds - the synthetic aryl hydrocarbon receptor (AhR) agonist IGN-512 together with TGF-β and retinoic acid - was developed to create a platform technology for induction of tolerogenic DCs intended for treatment of several conditions caused by unwanted immune activation. These in vitro-generated cells, designated ItolDCs, are phenotypically characterized by their low expression of co-stimulatory and activating molecules along with high expression of tolerance-associated markers such as ILT3, CD103, and LAP, and a weak pro-inflammatory cytokine profile. When co-cultured with T cells and/or B cells, ItolDC-cultures contain higher frequencies of CD25+Foxp3+ regulatory T cells (Tregs), CD49b+LAG3+ 'type 1 regulatory (Tr1) T cells, and IL-10-producing B cells and are less T cell stimulatory compared to cultures with matured DCs. Factor VIII (FVIII) and tetanus toxoid (TT) were used as model antigens to study ItolDC antigen-loading. ItolDCs can take up FVIII, process, and present FVIII peptides on HLA-DR. By loading both ItolDCs and mDCs with TT, antigen-specific T cell proliferation was observed. Cryo-preserved ItolDCs showed a stable tolerogenic phenotype that was maintained after stimulation with LPS, CD40L, or a pro-inflammatory cocktail. Moreover, exposure to other immune cells did not negatively impact ItolDCs' expression of tolerogenic markers. In summary, a novel protocol was developed supporting the generation of a stable population of human DCs in vitro that exhibited a tolerogenic phenotype with an ability to increase proportions of induced regulatory T and B cells in mixed cultures. This protocol has the potential to constitute the base of a tolDC platform for inducing antigen-specific tolerance in disorders caused by undesired antigen-specific immune cell activation.
Collapse
Affiliation(s)
- Gillian Dao Nyesiga
- Idogen AB, Lund, Sweden
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
| | | | | | | | - Lars Ohlsson
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
| | - Daniel Appelgren
- Department of Health, Medicine and Caring Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | | | | | | | | |
Collapse
|
4
|
Zhu K, Kazim N, Yue J, Yen A. Vacuolin-1 enhances RA-induced differentiation of human myeloblastic leukemia cells: evidence for involvement of a CD11b/FAK/LYN/SLP-76 axis subject to endosomal regulation that drives late differentiation steps. Cell Biosci 2022; 12:179. [PMID: 36329484 PMCID: PMC9635152 DOI: 10.1186/s13578-022-00911-6] [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: 03/19/2022] [Accepted: 10/07/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Retinoic acid(RA), an embryonic morphogen, regulates cell differentiation. Endocytosis regulates receptor signaling that governs such RA-directed cellular processes. Vacuolin-1 is a small molecule that disrupts endocytosis, motivating interest in its effect on RA-induced differentiation/arrest. In HL-60 myeloblastic-leukemia cells, RA causes differentiation evidenced by a progression of cell-surface and functional markers, CD38, CD11b, and finally reactive oxygen species(ROS) production and G1/0 cell cycle arrest in mature cells. RESULTS We found that Vacuolin-1 enhanced RA-induced CD11b, ROS and G1/0 arrest, albeit not CD38. Enhanced CD11b expression was associated with enhanced activation of Focal Adhesion Kinase(FAK). Adding vacuolin-1 enhanced RA-induced tyrosine phosphorylation of FAK, Src Family Kinases(SFKs), and the adaptor protein, SLP-76, expression of which is known to drive RA-induced differentiation. Depleting CD11b cripples late stages of progressive myeloid differentiation, namely G1/0 arrest and inducible ROS production, but not expression of CD38. Loss of NUMB, a protein that supports early endosome maturation, affected RA-induced ROS and G1/0 arrest, but not CD38 expression. CONCLUSION Hence there appears to be a novel CD11b/FAK/LYN/SLP-76 axis subject to endosome regulation which contributes to later stages of RA-induced differentiation. The effects of vacuolin-1 thus suggest a model where RA-induced differentiation consists of progressive stages driven by expression of sequentially-induced receptors.
Collapse
Affiliation(s)
- Kaiyuan Zhu
- grid.448631.c0000 0004 5903 2808Division of Natural and Applied Sciences, Synear Molecular Biology Lab, Duke Kunshan University, Kunshan, China ,grid.464255.4City University of Hong Kong Shenzhen Research Institute, ShenZhen, China
| | - Noor Kazim
- grid.5386.8000000041936877XDepartment of Biomedical Sciences, Cornell University, Ithaca, NY USA
| | - Jianbo Yue
- grid.5386.8000000041936877XDepartment of Biomedical Sciences, Cornell University, Ithaca, NY USA ,grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China ,grid.464255.4City University of Hong Kong Shenzhen Research Institute, ShenZhen, China
| | - Andrew Yen
- grid.5386.8000000041936877XDepartment of Biomedical Sciences, Cornell University, Ithaca, NY USA
| |
Collapse
|
5
|
Shi D, Wu X, Jian Y, Wang J, Huang C, Mo S, Li Y, Li F, Zhang C, Zhang D, Zhang H, Huang H, Chen X, Wang YA, Lin C, Liu G, Song L, Liao W. USP14 promotes tryptophan metabolism and immune suppression by stabilizing IDO1 in colorectal cancer. Nat Commun 2022; 13:5644. [PMID: 36163134 PMCID: PMC9513055 DOI: 10.1038/s41467-022-33285-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 09/08/2022] [Indexed: 12/03/2022] Open
Abstract
Indoleamine 2,3 dioxygenase 1 (IDO1) is an attractive target for cancer immunotherapy. However, IDO1 inhibitors have shown disappointing therapeutic efficacy in clinical trials, mainly because of the activation of the aryl hydrocarbon receptor (AhR). Here, we show a post-transcriptional regulatory mechanism of IDO1 regulated by a proteasome-associated deubiquitinating enzyme, USP14, in colorectal cancer (CRC). Overexpression of USP14 promotes tryptophan metabolism and T-cell dysfunction by stabilizing the IDO1 protein. Knockdown of USP14 or pharmacological targeting of USP14 decreases IDO1 expression, reverses suppression of cytotoxic T cells, and increases responsiveness to anti-PD-1 in a MC38 syngeneic mouse model. Importantly, suppression of USP14 has no effects on AhR activation induced by the IDO1 inhibitor. These findings highlight a relevant role of USP14 in post-translational regulation of IDO1 and in the suppression of antitumor immunity, suggesting that inhibition of USP14 may represent a promising strategy for CRC immunotherapy.
Collapse
Affiliation(s)
- Dongni Shi
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Xianqiu Wu
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Yunting Jian
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
- Department of Pathology, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, 510150, Guangzhou, China
| | - Junye Wang
- Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Chengmei Huang
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Shuang Mo
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 510080, Guangzhou, China
| | - Yue Li
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Fengtian Li
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Chao Zhang
- Department of Pathology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China
| | - Dongsheng Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China
| | - Huizhong Zhang
- Department of Pathology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China
| | - Huilin Huang
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Xin Chen
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Institute of Oncology, Tumor Hospital, Guangzhou Medical University, 511436, Guangzhou, China
| | - Y Alan Wang
- Brown Center for Immunotherapy, Department of Medicine, Indiana University School of Medicine, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, 46202-3082, USA
| | - Chuyong Lin
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Guozhen Liu
- School of Life and Health Sciences, The Chinese University of Hong Kong, 518172, Shenzhen, China.
| | - Libing Song
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China.
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Institute of Oncology, Tumor Hospital, Guangzhou Medical University, 511436, Guangzhou, China.
| | - Wenting Liao
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China.
| |
Collapse
|
6
|
Kazim N, Yen A. Role for Fgr and Numb in retinoic acid-induced differentiation and G0 arrest of non-APL AML cells. Oncotarget 2021; 12:1147-1164. [PMID: 34136084 PMCID: PMC8202776 DOI: 10.18632/oncotarget.27969] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/03/2021] [Indexed: 12/15/2022] Open
Abstract
Retinoic acid (RA) is a fundamental regulator of cell cycle and cell differentiation. Using a leukemic patient-derived in vitro model of a non-APL AML, we previously found that RA evokes activation of a macromolecular signaling complex, a signalosome, built of numerous MAPK-pathway-related signaling molecules; and this signaling enabled Retinoic-Acid-Response-Elements (RAREs) to regulate gene expression that results in cell differentiation/cell cycle arrest. Toward mechanistic insight into the nature of this novel signaling, we now find that the NUMB cell fate determinant protein is an apparent scaffold for the signalosome. Numb exists in the cell bound to an ensemble of signalosome molecules, including Raf, Lyn, Slp-76, and Vav. Addition of RA induces the expression of Fgr. Fgr binds NUMB, which is associated with (p-tyr)phosphorylation of NUMB and enhanced NUMB-binding and (p-tyr)phosphorylation of select signalosome components, thereby betraying signalosome activation. Signalosome activation is associated with cell differentiation along the myeloid lineage and G1/0 cell cycle arrest. If RA-induced Fgr expression is ablated by a CRISPR-KO; then the RA-induced (p-tyr) phosphorylation of NUMB and enhanced NUMB-binding and (p-tyr)phosphorylation of select signalosome components are lost. The cells now fail to undergo RA-induced differentiation or G1/0 arrest. In sum we find that NUMB acts as a scaffold for a signaling machine that functions to propel RA-induced differentiation and G1/0 arrest, and that Fgr binding to NUMB turns the function on. The Numb fate determinant protein thus appears to regulate the retinoic acid embryonic morphogen using the Fgr Src-Family-Kinase. These mechanistic insights suggest therapeutic targets for a hitherto incurable AML.
Collapse
Affiliation(s)
- Noor Kazim
- Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Andrew Yen
- Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
7
|
Han H, Shin DY, Kim D, Kim H, Lee C, Koh Y, Hong J, Yoon SS. Induction of leukemic stem cell differentiation by aryl hydrocarbon receptor agonist and synergy with gilteritinib in FLT3-ITD + acute myeloid leukemia. Leuk Lymphoma 2020; 61:1932-1942. [PMID: 32374198 DOI: 10.1080/10428194.2020.1747062] [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: 10/24/2022]
Abstract
Leukemic stem cells (LSCs) are a major cause of treatment failure and recurrence of acute myeloid leukemia (AML). Targeting LSC is essential to developing a potential cure for patients with relapsed/refractory AML. Here we investigated the effect of aryl hydrocarbon receptor (AhR) signaling on AML stem/progenitor proportion and examined the combined effect of AhR agonist and tyrosine kinase inhibitor. The AhR agonist, 6-formylindolo[3,2-b]carbazole (FICZ), significantly decreased the LSC proportion and clonogenicity and increased differentiation markers in AML primary cells. Synergistic/additive effects of FICZ and gilteritinib, FMS-like tyrosine kinase 3 (FLT3) inhibitor, were confirmed in AML cells with FLT3-ITD. We present evidence that combination of both agents inhibits FLT3 downstream molecules and degrades clonogenicity. Collectively, our results suggest that FICZ not only compels LSC differentiation, but also enhances the efficacy of gilteritinib when combined. Clinical application of this combined approach may pave a new therapeutic strategy for patients with FLT3 mutated AML.
Collapse
Affiliation(s)
- Heejoo Han
- Seoul National University College of Medicine, Seoul, Republic of Korea.,Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong-Yeop Shin
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dongchan Kim
- Seoul National University College of Medicine, Seoul, Republic of Korea.,Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyungsuk Kim
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chansup Lee
- Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Youngil Koh
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Junshik Hong
- Seoul National University College of Medicine, Seoul, Republic of Korea.,Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung-Soo Yoon
- Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
8
|
Ly M, Rentas S, Vujovic A, Wong N, Moreira S, Xu J, Holzapfel N, Bhatia S, Tran D, Minden MD, Draper JS, Hope KJ. Diminished AHR Signaling Drives Human Acute Myeloid Leukemia Stem Cell Maintenance. Cancer Res 2019; 79:5799-5811. [PMID: 31519687 DOI: 10.1158/0008-5472.can-19-0274] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 07/31/2019] [Accepted: 09/10/2019] [Indexed: 11/16/2022]
Abstract
Eliminating leukemic stem cells (LSC) is a sought after therapeutic paradigm for the treatment of acute myeloid leukemia (AML). While repression of aryl hydrocarbon receptor (AHR) signaling has been shown to promote short-term maintenance of primitive AML cells in culture, no work to date has examined whether altered AHR signaling plays a pathologic role in human AML or whether it contributes at all to endogenous LSC function. Here, we show AHR signaling is repressed in human AML blasts and preferentially downregulated in LSC-enriched populations within leukemias. A core set of AHR targets are uniquely repressed in LSCs across diverse genetic AML subtypes. In vitro and in vivo administration of the specific AHR agonist FICZ significantly impaired leukemic growth, promoted differentiation, and repressed self-renewal. Furthermore, LSCs suppressed a set of FICZ-responsive AHR target genes that function as tumor suppressors and promoters of differentiation. FICZ stimulation did not impair normal hematopoietic stem and progenitor (HSPC) function, and failed to upregulate a prominent LSC-specific AHR target in HSPCs, suggesting that differential mechanisms govern FICZ-induced AHR signaling manifestations in HSCs versus LSCs. Altogether, this work highlights AHR signaling suppression as a key LSC-regulating control mechanism and provides proof of concept in a preclinical model that FICZ-mediated AHR pathway activation enacts unique transcriptional programs in AML that identify it as a novel chemotherapeutic approach to selectively target human LSCs. SIGNIFICANCE: The AHR pathway is suppressed in leukemic stem cells (LSC), therefore activating AHR signaling is a potential therapeutic option to target LSCs and to treat acute myeloid leukemia.
Collapse
Affiliation(s)
- Michelle Ly
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Stefan Rentas
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Ana Vujovic
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Nicholas Wong
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Steven Moreira
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Joshua Xu
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Nicholas Holzapfel
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Sonam Bhatia
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Damian Tran
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Mark D Minden
- Department of Medicine, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan S Draper
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Kristin J Hope
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
9
|
Günther J, Däbritz J, Wirthgen E. Limitations and Off-Target Effects of Tryptophan-Related IDO Inhibitors in Cancer Treatment. Front Immunol 2019; 10:1801. [PMID: 31417567 PMCID: PMC6682646 DOI: 10.3389/fimmu.2019.01801] [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: 04/30/2019] [Accepted: 07/17/2019] [Indexed: 12/11/2022] Open
Abstract
Immunooncology is still a growing area in cancer therapy. Drugs within this therapeutic approach do not directly target/attack the tumor but interfere with immune checkpoints and target or reprogram key metabolic pathways critical for anti-cancer immune defense. Indolamine 2,3-dioxygenase 1 (IDO1) and the tryptophan (TRP)-kynurenine pathway were identified as critical mechanisms in cancer immune escape and their inhibition as an approach with promising therapeutic potential. Particularly, a multitude of IDO1 inhibiting tryptophan analogs are widely applied in several clinical trials. However, this therapy results in a variety of implications for the patient's physiology. This is not only due to the inhibition of an enzyme important in almost every organ and tissue in the body but also because of the general nature of the inhibitor as an analog of a proteinogenic amino acid as well as the initiation of cellular detoxification known to affect inflammatory pathways. In this review we provide a deeper insight into the physiological consequences of an IDO1 inhibiting therapy based on TRP related molecules. We discuss potential side and off-target effects that contribute to the interpretation of unexpected positive as well as negative results of ongoing or discontinued clinical studies while we also highlight the potential of these inhibitors independent of the IDO1 signaling pathway.
Collapse
Affiliation(s)
- Juliane Günther
- Research Group Epigenetics, Metabolism and Longevity, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Jan Däbritz
- Department of Pediatrics, Rostock University Medical Center, Rostock, Germany
| | - Elisa Wirthgen
- Department of Pediatrics, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
10
|
Yang D, Li T, Li Y, Zhang S, Li W, Liang H, Xing Z, Du L, He J, Kuang C, Yang Q. H 2S suppresses indoleamine 2, 3-dioxygenase 1 and exhibits immunotherapeutic efficacy in murine hepatocellular carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:88. [PMID: 30777103 PMCID: PMC6380069 DOI: 10.1186/s13046-019-1083-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/06/2019] [Indexed: 12/16/2022]
Abstract
Background Over-expression and over-activation of immunosuppressive enzyme indoleamine 2, 3 -dioxygenase 1 (IDO1) is a key mechanism of cancer immune escape. However, the regulation of IDO1 has not been fully studied. The relation between hydrogen sulfide (H2S) and IDO1 is unclear. Methods The influences of endogenous and exogenous H2S on the expression of IDO1, iNOS and NF-κB and STAT3 signaling proteins were investigated using qPCR or western blot, and the production of nitric oxide (NO) was analyzed by nitrate/nitrite assay in Cse−/− mice and MCF-7 and SGC-7901 cells. The effect of H2S on IDO1 activity was investigated by HPLC and in-vitro enzymatic assay. The effect of H2S on tryptophan metabolism was tested by luciferase reporter assay in MCF-7 and SGC-7901 cells. The correlation between H2S-generating enzyme CSE and IDO1 was investigated by immunostaining and heatmaps analysis in clinical specimens and tissue arrays of hepatocellular carcinoma (HCC) patients. The immunotherapeutic effects of H2S on H22 HCC-bearing mice were investigated. Results Using Cse−/− mice, we found that H2S deficiency increased IDO1 expression and activity, stimulated NF-κB and STAT3 pathways and decreased the expression of NO-generating enzyme Inos. Using IDO1-expressing MCF-7 and SGC-7901 cells, we found that exogenous H2S inhibited IDO1 expression by blocking STAT3 and NF-κB pathways, and decreased IDO1 activity via H2S/NO crosstalk, and combinedly decreased the tryptophan metabolism. The negative correlation between H2S-generating enzyme CSE and IDO1 was further validated in clinical specimens and tissue arrays of HCC patients. Additionally, H2S donors effectively restricted the tumor development in H22 HCC-bearing mice via downregulating IDO1 expression, inducing T-effector cells and inhibiting MDSCs. Conclusions Thus, H2S, as a novel negative regulator of IDO1, shows encouraging antitumor immunotherapeutic effects and represents a novel therapeutic target in cancer therapy. Electronic supplementary material The online version of this article (10.1186/s13046-019-1083-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dan Yang
- State Key Laboratory of Genetic Engineering, Department of Biochemistry, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Tianqi Li
- State Key Laboratory of Genetic Engineering, Department of Biochemistry, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Yinlong Li
- State Key Laboratory of Genetic Engineering, Department of Biochemistry, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Shengnan Zhang
- State Key Laboratory of Genetic Engineering, Department of Biochemistry, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Weirui Li
- State Key Laboratory of Genetic Engineering, Department of Biochemistry, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Heng Liang
- State Key Laboratory of Genetic Engineering, Department of Biochemistry, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Zikang Xing
- State Key Laboratory of Genetic Engineering, Department of Biochemistry, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Lisha Du
- State Key Laboratory of Genetic Engineering, Department of Biochemistry, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Jinchao He
- State Key Laboratory of Genetic Engineering, Department of Biochemistry, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Chunxiang Kuang
- Department of Chemistry, Tongji University, Siping Road 1239, Shanghai, 200092, China
| | - Qing Yang
- State Key Laboratory of Genetic Engineering, Department of Biochemistry, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China.
| |
Collapse
|
11
|
Bunaciu RP, MacDonald RJ, Jensen HA, Gao F, Wang X, Johnson L, Varner JD, Yen A. Retinoic acid and 6-formylindolo(3,2-b)carbazole (FICZ) combination therapy reveals putative targets for enhancing response in non-APL AML. Leuk Lymphoma 2018; 60:1697-1708. [PMID: 30570341 DOI: 10.1080/10428194.2018.1543880] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In non-acute promyelotic leukemia (APL)- non myelocytic leukemia (AML), identification of a signaling signature would predict potentially actionable targets to enhance differentiation effects of all-trans-retinoic acid (RA) and make combination differentiation therapy realizable. Components of such a signaling machine/signalsome found to drive RA-induced differentiation discerned in a FAB M2 cell line/model (HL-60) were further characterized and then compared against AML patient expression profiles. FICZ, known to enhance RA-induced differentiation, was used to experimentally augment signaling for analysis. FRET revealed novel signalsome protein associations: CD38 with pS376SLP76 and caveolin-1 with CD38 and AhR. The signaling molecules driving differentiation in HL-60 cluster in non-APL AML de novo samples, too. Pearson correlation coefficients for this molecular ensemble are nearer 1 in the FAB M2 subtype than in non-APL AML. SLP76 correlation to RXRα and p47phox were conserved in FAB M2 model and patient subtype but not in general non-APL AML. The signalsome ergo identifies potential actionable targets in AML.
Collapse
Affiliation(s)
- Rodica P Bunaciu
- a Department of Biomedical Sciences , Cornell University , Ithaca , NY , USA
| | - Robert J MacDonald
- a Department of Biomedical Sciences , Cornell University , Ithaca , NY , USA
| | - Holly A Jensen
- a Department of Biomedical Sciences , Cornell University , Ithaca , NY , USA.,b Robert Frederick Smith School of Chemical and Biomolecular Engineering , Cornell University , Ithaca , NY , USA
| | - Feng Gao
- a Department of Biomedical Sciences , Cornell University , Ithaca , NY , USA.,c Department of Biomedical Sciences , City University of Hong Kong , Hong Kong , China
| | - Xin Wang
- c Department of Biomedical Sciences , City University of Hong Kong , Hong Kong , China
| | - Lynn Johnson
- d Cornell Statistical Unit , Cornell University , Ithaca , NY , USA
| | - Jeffrey D Varner
- b Robert Frederick Smith School of Chemical and Biomolecular Engineering , Cornell University , Ithaca , NY , USA
| | - Andrew Yen
- a Department of Biomedical Sciences , Cornell University , Ithaca , NY , USA
| |
Collapse
|
12
|
Rannug A, Rannug U. The tryptophan derivative 6-formylindolo[3,2-b]carbazole, FICZ, a dynamic mediator of endogenous aryl hydrocarbon receptor signaling, balances cell growth and differentiation. Crit Rev Toxicol 2018; 48:555-574. [PMID: 30226107 DOI: 10.1080/10408444.2018.1493086] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The aryl hydrocarbon receptor (AHR) is not essential to survival, but does act as a key regulator of many normal physiological events. The role of this receptor in toxicological processes has been studied extensively, primarily employing the high-affinity ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). However, regulation of physiological responses by endogenous AHR ligands remains to be elucidated. Here, we review developments in this field, with a focus on 6-formylindolo[3,2-b]carbazole (FICZ), the endogenous ligand with the highest affinity to the receptor reported to date. The binding of FICZ to different isoforms of the AHR seems to be evolutionarily well conserved and there is a feedback loop that controls AHR activity through metabolic degradation of FICZ via the highly inducible cytochrome P450 1A1. Several investigations provide strong evidence that FICZ plays a critical role in normal physiological processes and can ameliorate immune diseases with remarkable efficiency. Low levels of FICZ are pro-inflammatory, providing resistance to pathogenic bacteria, stimulating the anti-tumor functions, and promoting the differentiation of cancer cells by repressing genes in cancer stem cells. In contrast, at high concentrations FICZ behaves in a manner similar to TCDD, exhibiting toxicity toward fish and bird embryos, immune suppression, and activation of cancer progression. The findings are indicative of a dual role for endogenously activated AHR in barrier tissues, aiding clearance of infections and suppressing immunity to terminate a vicious cycle that might otherwise lead to disease. There is not much support for the AHR ligand-specific immune responses proposed, the differences between FICZ and TCDD in this context appear to be explained by the rapid metabolism of FICZ.
Collapse
Affiliation(s)
- Agneta Rannug
- a Karolinska Institutet, Institute of Environmental Medicine , Stockholm , Sweden
| | - Ulf Rannug
- b Department of Molecular Biosciences , The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| |
Collapse
|
13
|
Janosik T, Rannug A, Rannug U, Wahlström N, Slätt J, Bergman J. Chemistry and Properties of Indolocarbazoles. Chem Rev 2018; 118:9058-9128. [PMID: 30191712 DOI: 10.1021/acs.chemrev.8b00186] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The indolocarbazoles are an important class of nitrogen heterocycles which has evolved significantly in recent years, with numerous studies focusing on their diverse biological effects, or targeting new materials with potential applications in organic electronics. This review aims at providing a broad survey of the chemistry and properties of indolocarbazoles from an interdisciplinary point of view, with particular emphasis on practical synthetic aspects, as well as certain topics which have not been previously accounted for in detail, such as the occurrence, formation, biological activities, and metabolism of indolo[3,2- b]carbazoles. The literature of the past decade forms the basis of the text, which is further supplemented with older key references.
Collapse
Affiliation(s)
- Tomasz Janosik
- Research Institutes of Sweden , Bioscience and Materials, RISE Surface, Process and Formulation , SE-151 36 Södertälje , Sweden
| | - Agneta Rannug
- Institute of Environmental Medicine , Karolinska Institutet , SE-171 77 Stockholm , Sweden
| | - Ulf Rannug
- Department of Molecular Biosciences, The Wenner-Gren Institute , Stockholm University , SE-106 91 Stockholm , Sweden
| | | | - Johnny Slätt
- Department of Chemistry, Applied Physical Chemistry , KTH Royal Institute of Technology , SE-100 44 Stockholm , Sweden
| | - Jan Bergman
- Karolinska Institutet , Department of Biosciences and Nutrition , SE-141 83 Huddinge , Sweden
| |
Collapse
|
14
|
Amanzadeh A, Molla-Kazemiha V, Samani S, Habibi-Anbouhi M, Azadmanesh K, Abolhassani M, Shokrgozar MA. New synergistic combinations of differentiation-inducing agents in the treatment of acute promyelocytic leukemia cells. Leuk Res 2018; 68:98-104. [PMID: 29602066 DOI: 10.1016/j.leukres.2018.01.007] [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: 10/16/2017] [Revised: 12/29/2017] [Accepted: 01/14/2018] [Indexed: 10/18/2022]
Abstract
Acute promyelocytic leukemia (APL) was considered to be one of the most lethal forms of leukemia in adults before the introduction of the vitamin A metabolite all-trans retinoic acid (ATRA). Surprisingly, it has been confirmed that FICZ (6-Formylindolo (3, 2-b) carbazole) enhances ATRA-induced differentiation. Moreover, a number of studies have demonstrated that anti CD44 monoclonal antibody (mAb) induces to bring back differentiation blockage the leukemic stem cells. The level of differentiation markers including CD11b and CD11c in NB4 cells was assessed by flow cytometry. The induction of apoptosis was also evaluated. We estimated the induction potential of a triple compound of ATRA-FICZ, anti-CD44 maps. The cells showed the gradually increased expression levels of CD11b and CD11c. A mixture of a "CD44 mAb, ATRA and FICZ effectively promoted granulocytic maturation resulting in increased rates of apoptosis. The differences in expression of CD11b and CD11c at 5 μg/ml and 10 μg/ml were significant. These phenomena were highest at 10 μg/ml CD44 mAb concentrations. Synergistic induction differentiation and apoptosis of APL cells by using a co-treatment with novel triple compound are more effective for eradicating blasts and controlling the metastasis. Our results show that the addition of anti-CD44 mAb improves "ATRA-FICZ"-induced differentiation and has potential to reduce usual chemotherapy based treatments. Taken together, this compound may lead to novel clinical applications of differentiation-based approaches for APL and other types of leukemia. Further clinical studies would be recommended to clarify the clinical efficacy.
Collapse
Affiliation(s)
- Amir Amanzadeh
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | | | - Saeed Samani
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Mohsen Abolhassani
- Department of Immunology, Hybridoma Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
15
|
MacDonald RJ, Bunaciu RP, Ip V, Dai D, Tran D, Varner JD, Yen A. Src family kinase inhibitor bosutinib enhances retinoic acid-induced differentiation of HL-60 leukemia cells. Leuk Lymphoma 2018; 59:2941-2951. [PMID: 29569971 DOI: 10.1080/10428194.2018.1452213] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The acute promyelocytic leukemia (APL) has been treated with all-trans retinoic acid (RA) for decades. While RA has largely been ineffective in non-APL AML subtypes, co-treatments combining RA and other agents are currently in clinical trials. Using the RA-responsive non-APL AML cell line HL-60, we tested the efficacy of the Src family kinase (SFK) inhibitor bosutinib on RA-induced differentiation. HL-60 has been recently shown to bear fidelity to a subtype of AML that respond to RA. We found that co-treatment with RA and bosutinib enhanced differentiation evidenced by increased CD11b expression, G1/G0 cell cycle arrest, and respiratory burst. Expression of the SFK members Fgr and Lyn was enhanced, while SFK activation was inhibited. Phosphorylation of several sites of c-Raf was increased and expression of AhR and p85 PI3K was enhanced. Expression of c-Cbl and mTOR was decreased. Our study suggests that SFK inhibition enhances RA-induced differentiation and may have therapeutic value in non-APL AML.
Collapse
Affiliation(s)
- Robert J MacDonald
- a Department of Biomedical Sciences , Cornell University , Ithaca , NY , USA
| | - Rodica P Bunaciu
- a Department of Biomedical Sciences , Cornell University , Ithaca , NY , USA
| | - Victoria Ip
- a Department of Biomedical Sciences , Cornell University , Ithaca , NY , USA
| | - David Dai
- b Robert Frederick Smith School of Chemical and Biomolecular Engineering , Cornell University , Ithaca , NY , USA
| | - David Tran
- a Department of Biomedical Sciences , Cornell University , Ithaca , NY , USA
| | - Jeffrey D Varner
- b Robert Frederick Smith School of Chemical and Biomolecular Engineering , Cornell University , Ithaca , NY , USA
| | - Andrew Yen
- a Department of Biomedical Sciences , Cornell University , Ithaca , NY , USA
| |
Collapse
|
16
|
Bunaciu RP, MacDonald RJ, Gao F, Johnson LM, Varner JD, Wang X, Nataraj S, Guzman ML, Yen A. Potential for subsets of wt-NPM1 primary AML blasts to respond to retinoic acid treatment. Oncotarget 2017; 9:4134-4149. [PMID: 29423110 PMCID: PMC5790527 DOI: 10.18632/oncotarget.23642] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/09/2017] [Indexed: 01/16/2023] Open
Abstract
Acute myeloid leukemia (AML) has high mortality rates, perhaps reflecting a lack of understanding of the molecular diversity in various subtypes and a lack of known actionable targets. There are currently 12 open clinical trials for AML using combination therapeutic modalities including all-trans retinoic acid (RA). Mutant nucleophosmin-1, proposed as a possible marker for RA response, is the criterion for recruiting patients in three active RA phase 3 clinical trials. We tested the ability of RA alone or in combination with either bosutinib (B) or 6-formylindolo(3,2-b) carbazole (F) to induce conversion of 12 de novo AML samples toward a more differentiated phenotype. We assessed levels of expression of cell surface markers associated with differentiation, aldehyde dehydrogenase activity, and glucose uptake activity. Colony formation capacity was reduced with the combined treatment of RA and B or F, and correlated with modulation of a c-Cbl/Lyn/c-Raf-centered signalsome. Combination treatment was in most cases more effective than RA alone. Based on their responses to the treatments, some primary leukemic samples cluster closer to HL-60 cells than to other primary samples, suggesting that they may represent a hitherto undefined AML subtype that is potentially responsive to RA in a combination differentiation therapy.
Collapse
Affiliation(s)
- Rodica P Bunaciu
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
| | | | - Feng Gao
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA.,Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA.,Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Lynn M Johnson
- Cornell Statistical Unit, Cornell University, Ithaca, NY, USA
| | - Jeffrey D Varner
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA
| | - Xin Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Sarah Nataraj
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Monica L Guzman
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Andrew Yen
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
| |
Collapse
|
17
|
An Effective Model of the Retinoic Acid Induced HL-60 Differentiation Program. Sci Rep 2017; 7:14327. [PMID: 29085021 PMCID: PMC5662654 DOI: 10.1038/s41598-017-14523-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/11/2017] [Indexed: 12/17/2022] Open
Abstract
In this study, we present an effective model All-Trans Retinoic Acid (ATRA)-induced differentiation of HL-60 cells. The model describes reinforcing feedback between an ATRA-inducible signalsome complex involving many proteins including Vav1, a guanine nucleotide exchange factor, and the activation of the mitogen activated protein kinase (MAPK) cascade. We decomposed the effective model into three modules; a signal initiation module that sensed and transformed an ATRA signal into program activation signals; a signal integration module that controlled the expression of upstream transcription factors; and a phenotype module which encoded the expression of functional differentiation markers from the ATRA-inducible transcription factors. We identified an ensemble of effective model parameters using measurements taken from ATRA-induced HL-60 cells. Using these parameters, model analysis predicted that MAPK activation was bistable as a function of ATRA exposure. Conformational experiments supported ATRA-induced bistability. Additionally, the model captured intermediate and phenotypic gene expression data. Knockout analysis suggested Gfi-1 and PPARg were critical to the ATRAinduced differentiation program. These findings, combined with other literature evidence, suggested that reinforcing feedback is central to hyperactive signaling in a diversity of cell fate programs.
Collapse
|
18
|
Hammerschmidt-Kamper C, Biljes D, Merches K, Steiner I, Daldrup T, Bol-Schoenmakers M, Pieters RHH, Esser C. Indole-3-carbinol, a plant nutrient and AhR-Ligand precursor, supports oral tolerance against OVA and improves peanut allergy symptoms in mice. PLoS One 2017; 12:e0180321. [PMID: 28666018 PMCID: PMC5493375 DOI: 10.1371/journal.pone.0180321] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/14/2017] [Indexed: 12/14/2022] Open
Abstract
In general, dietary antigens are tolerated by the gut associated immune system. Impairment of this so-called oral tolerance is a serious health risk. We have previously shown that activation of the ligand-dependent transcription factor aryl hydrocarbon receptor (AhR) by the environmental pollutant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) affects both oral tolerance and food allergy. In this study, we determine whether a common plant-derived, dietary AhR-ligand modulates oral tolerance as well. We therefore fed mice with indole-3-carbinole (I3C), an AhR ligand that is abundant in cruciferous plants. We show that several I3C metabolites were detectable in the serum after feeding, including the high-affinity ligand 3,3´-diindolylmethane (DIM). I3C feeding robustly induced the AhR-target gene CYP4501A1 in the intestine; I3C feeding also induced the aldh1 gene, whose product catalyzes the formation of retinoic acid (RA), an inducer of regulatory T cells. We then measured parameters indicating oral tolerance and severity of peanut-induced food allergy. In contrast to the tolerance-breaking effect of TCDD, feeding mice with chow containing 2 g/kg I3C lowered the serum anti-ovalbumin IgG1 response in an experimental oral tolerance protocol. Moreover, I3C feeding attenuated symptoms of peanut allergy. In conclusion, the dietary compound I3C can positively influence a vital immune function of the gut.
Collapse
Affiliation(s)
| | - Daniel Biljes
- IUF – Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Katja Merches
- IUF – Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Irina Steiner
- Institute of Legal Medicine, Department of Forensic Toxicology, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Thomas Daldrup
- Institute of Legal Medicine, Department of Forensic Toxicology, University Hospital of Düsseldorf, Düsseldorf, Germany
| | | | - Raymond H. H. Pieters
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Charlotte Esser
- IUF – Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
19
|
Brem R, Macpherson P, Guven M, Karran P. Oxidative stress induced by UVA photoactivation of the tryptophan UVB photoproduct 6-formylindolo[3,2-b]carbazole (FICZ) inhibits nucleotide excision repair in human cells. Sci Rep 2017; 7:4310. [PMID: 28655934 PMCID: PMC5487344 DOI: 10.1038/s41598-017-04614-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 05/17/2017] [Indexed: 01/11/2023] Open
Abstract
Potentially mutagenic DNA lesions induced by UVB (wavelengths 280–320 nm) are important risk factors for solar ultraviolet (UV) radiation-induced skin cancer. The carcinogenicity of the more abundant UVA (320–400 nm) is less well understood but is generally regarded to reflect its interaction with cellular chromophores that act as photosensitisers. The arylhydrocarbon receptor agonist 6-formylindolo[3,2-b] carbazole (FICZ), is a UVB photoproduct of tryptophan and a powerful UVA chromophore. Combined with UVA, FICZ generates reactive oxygen species (ROS) and induces oxidative DNA damage. Here we demonstrate that ROS generated by FICZ/UVA combinations also cause extensive protein damage in HaCaT human keratinocytes. We show that FICZ/UVA-induced oxidation significantly inhibits the removal of potentially mutagenic UVB-induced DNA photolesions by nucleotide excision repair (NER). DNA repair inhibition is due to FICZ/UVA-induced oxidation damage to the NER proteome and DNA excision repair is impaired in extracts prepared from FICZ/UVA-treated cells. NER protects against skin cancer. As a likely UVB photoproduct of intracellular tryptophan, FICZ represents a de facto endogenous UVA photosensitiser in sun-exposed skin. FICZ formation may increase the risk of solar UV-induced skin cancer by promoting photochemical damage to the NER proteome and thereby preventing the removal of UVB-induced DNA lesions.
Collapse
Affiliation(s)
- Reto Brem
- The Francis Crick Institute, Midland Road, London, NW1 1AT, UK
| | | | - Melisa Guven
- The Francis Crick Institute, Midland Road, London, NW1 1AT, UK
| | - Peter Karran
- The Francis Crick Institute, Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
20
|
Mulero-Navarro S, Fernandez-Salguero PM. New Trends in Aryl Hydrocarbon Receptor Biology. Front Cell Dev Biol 2016; 4:45. [PMID: 27243009 PMCID: PMC4863130 DOI: 10.3389/fcell.2016.00045] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 04/28/2016] [Indexed: 12/28/2022] Open
Abstract
Traditionally considered as a critical intermediate in the toxic and carcinogenic response to dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin, TCDD), the Aryl hydrocarbon/Dioxin receptor (AhR) has proven to be also an important regulator of cell physiology and organ homeostasis. AhR has become an interesting and actual area of research mainly boosted by a significant number of recent studies analyzing its contribution to the proper functioning of the immune, hepatic, cardiovascular, vascular and reproductive systems. At the cellular level, AhR establishes functional interactions with signaling pathways governing cell proliferation and cell cycle, cell morphology, cell adhesion and cell migration. Two exciting new aspects in AhR biology deal with its implication in the control of cell differentiation and its more than likely involvement in cell pluripotency and stemness. In fact, it is possible that AhR could help modulate the balance between differentiation and pluripotency in normal and transformed tumor cells. At the molecular level, AhR regulates an increasingly large array of physiologically relevant genes either by traditional transcription-dependent mechanisms or by unforeseen processes involving genomic insulators, chromatin dynamics and the transcription of mobile genetic elements. AhR is also closely related to epigenetics, not only from the point of view of target gene expression but also with respect to its own regulation by promoter methylation. It is reasonable to consider that deregulation of these many functions could have a causative role, or at least contribute to, human disease. Consequently, several laboratories have proposed that AhR could be a valuable tool as diagnostic marker and/or therapeutic target in human pathologies. An additional point of interest is the possibility of regulating AhR activity by endogenous non-toxic low weight molecules agonist or antagonist molecules that could be present or included in the diet. In this review, we will address these molecular and functional features of AhR biology within physiological and pathological contexts.
Collapse
Affiliation(s)
- Sonia Mulero-Navarro
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura Badajoz, Spain
| | - Pedro M Fernandez-Salguero
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura Badajoz, Spain
| |
Collapse
|
21
|
Park JH, Choi AJ, Kim SJ, Cheong SW, Jeong SY. AhR activation by 6-formylindolo[3,2-b]carbazole and 2,3,7,8-tetrachlorodibenzo-p-dioxin inhibit the development of mouse intestinal epithelial cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 43:44-53. [PMID: 26950395 DOI: 10.1016/j.etap.2016.02.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/06/2016] [Accepted: 02/09/2016] [Indexed: 06/05/2023]
Abstract
The intestinal epithelium plays a central role in immune homeostasis in the intestine. AhR, a ligand-activated transcription factor, plays an important role in diverse physiological processes. The intestines are exposed to various exogenous and endogenous AhR ligands. Thus, AhR may regulate the intestinal homeostasis, directly acting on the development of intestinal epithelial cells (IEC). In this study, we demonstrated that 6-formylindolo[3,2-b]carbazole (FICZ) and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) inhibited the in vitro development of mouse intestinal organoids. The number of Paneth cells in the small intestine and the depth of crypts of the small and large intestines were reduced in mice administrated with FICZ. Immunohistochemical and flow cytometric assays revealed that AhR was highly expressed in Lgr5(+) stem cells. FICZ inhibited Wnt signaling lowering the level of β-catenin protein. Gene expression analyses demonstrated that FICZ increased expression of Lgr5, Math1, BMP4, and Indian Hedgehog while inhibiting that of Lgr4.
Collapse
Affiliation(s)
- Joo-Hung Park
- Department of Biology, Changwon National University, Changwon, Kyungnam, 641-773, Korea.
| | - Ah-Jeong Choi
- Department of Biology, Changwon National University, Changwon, Kyungnam, 641-773, Korea
| | - Soo-Ji Kim
- Department of Biology, Changwon National University, Changwon, Kyungnam, 641-773, Korea
| | - Seon-Woo Cheong
- Department of Biology, Changwon National University, Changwon, Kyungnam, 641-773, Korea
| | - So-Yeon Jeong
- Department of Biology, Changwon National University, Changwon, Kyungnam, 641-773, Korea
| |
Collapse
|
22
|
Wirthgen E, Hoeflich A. Endotoxin-Induced Tryptophan Degradation along the Kynurenine Pathway: The Role of Indolamine 2,3-Dioxygenase and Aryl Hydrocarbon Receptor-Mediated Immunosuppressive Effects in Endotoxin Tolerance and Cancer and Its Implications for Immunoparalysis. JOURNAL OF AMINO ACIDS 2015; 2015:973548. [PMID: 26881062 PMCID: PMC4736209 DOI: 10.1155/2015/973548] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 10/28/2015] [Accepted: 12/06/2015] [Indexed: 12/16/2022]
Abstract
The degradation of tryptophan (TRP) along the kynurenine pathway plays a crucial role as a neuro- and immunomodulatory mechanism in response to inflammatory stimuli, such as lipopolysaccharides (LPS). In endotoxemia or sepsis, an enhanced activation of the rate-limiting enzyme indoleamine 2,3-dioxygenase (IDO) is associated with a higher mortality risk. It is assumed that IDO induced immunosuppressive effects provoke the development of a protracted compensatory hypoinflammatory phase up to a complete paralysis of the immune system, which is characterized by an endotoxin tolerance. However, the role of IDO activation in the development of life-threatening immunoparalysis is still poorly understood. Recent reports described the impact of inflammatory IDO activation and aryl hydrocarbon receptor- (AhR-) mediated pathways on the development of LPS tolerance and immune escape of cancer cells. These immunosuppressive mechanisms offer new insights for a better understanding of the development of cellular dysfunctions in immunoparalysis. This review provides a comprehensive update of significant biological functions of TRP metabolites along the kynurenine pathway and the complex regulation of LPS-induced IDO activation. In addition, the review focuses on the role of IDO-AhR-mediated immunosuppressive pathways in endotoxin tolerance and carcinogenesis revealing the significance of enhanced IDO activity for the establishment of life-threatening immunoparalysis in sepsis.
Collapse
Affiliation(s)
- Elisa Wirthgen
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology, Germany
| | - Andreas Hoeflich
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology, Germany
| |
Collapse
|
23
|
Esser C, Rannug A. The aryl hydrocarbon receptor in barrier organ physiology, immunology, and toxicology. Pharmacol Rev 2015; 67:259-79. [PMID: 25657351 DOI: 10.1124/pr.114.009001] [Citation(s) in RCA: 364] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is an evolutionarily old transcription factor belonging to the Per-ARNT-Sim-basic helix-loop-helix protein family. AhR translocates into the nucleus upon binding of various small molecules into the pocket of its single-ligand binding domain. AhR binding to both xenobiotic and endogenous ligands results in highly cell-specific transcriptome changes and in changes in cellular functions. We discuss here the role of AhR for immune cells of the barrier organs: skin, gut, and lung. Both adaptive and innate immune cells require AhR signaling at critical checkpoints. We also discuss the current two prevailing views-namely, 1) AhR as a promiscuous sensor for small chemicals and 2) a role for AhR as a balancing factor for cell differentiation and function, which is controlled by levels of endogenous high-affinity ligands. AhR signaling is considered a promising drug and preventive target, particularly for cancer, inflammatory, and autoimmune diseases. Therefore, understanding its biology is of great importance.
Collapse
Affiliation(s)
- Charlotte Esser
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany (C.E.); and Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden (A.R.)
| | - Agneta Rannug
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany (C.E.); and Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden (A.R.)
| |
Collapse
|
24
|
Bunaciu RP, Jensen HA, MacDonald RJ, LaTocha DH, Varner JD, Yen A. 6-Formylindolo(3,2-b)Carbazole (FICZ) Modulates the Signalsome Responsible for RA-Induced Differentiation of HL-60 Myeloblastic Leukemia Cells. PLoS One 2015; 10:e0135668. [PMID: 26287494 PMCID: PMC4545789 DOI: 10.1371/journal.pone.0135668] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 07/24/2015] [Indexed: 12/20/2022] Open
Abstract
6-Formylindolo(3,2-b)carbazole (FICZ) is a photoproduct of tryptophan and an endogenous high affinity ligand for aryl hydrocarbon receptor (AhR). It was previously reported that, in patient-derived HL-60 myeloblastic leukemia cells, retinoic acid (RA)-induced differentiation is driven by a signalsome containing c-Cbl and AhR. FICZ enhances RA-induced differentiation, assessed by expression of the membrane differentiation markers CD38 and CD11b, cell cycle arrest and the functional differentiation marker, inducible oxidative metabolism. Moreover, FICZ augments the expression of a number of the members of the RA-induced signalsome, such as c-Cbl, Vav1, Slp76, PI3K, and the Src family kinases Fgr and Lyn. Pursuing the molecular signaling responsible for RA-induced differentiation, we characterized, using FRET and clustering analysis, associations of key molecules thought to drive differentiation. Here we report that, assayed by FRET, AhR interacts with c-Cbl upon FICZ plus RA-induced differentiation, whereas AhR constitutively interacts with Cbl-b. Moreover, correlation analysis based on the flow cytometric assessment of differentiation markers and western blot detection of signaling factors reveal that Cbl-b, p-p38α and pT390-GSK3β, are not correlated with other known RA-induced signaling components or with a phenotypic outcome. We note that FICZ plus RA elicited signaling responses that were not typical of RA alone, but may represent alternative differentiation-driving pathways. In clusters of signaling molecules seminal to cell differentiation, FICZ co-administered with RA augments type and intensity of the dynamic changes induced by RA. Our data suggest relevance for FICZ in differentiation-induction therapy. The mechanism of action includes modulation of a SFK and MAPK centered signalsome and c-Cbl-AhR association.
Collapse
Affiliation(s)
- Rodica P. Bunaciu
- Department of Biomedical Sciences, Cornell University, Ithaca, New York, 14853, United States of America
| | - Holly A. Jensen
- Department of Biomedical Sciences, Cornell University, Ithaca, New York, 14853, United States of America
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, United States of America
| | - Robert J. MacDonald
- Department of Biomedical Sciences, Cornell University, Ithaca, New York, 14853, United States of America
| | - Dorian H. LaTocha
- Flow Cytometry Core Facility, Cornell University, Ithaca, New York, 14853, United States of America
| | - Jeffrey D. Varner
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, 14853, United States of America
| | - Andrew Yen
- Department of Biomedical Sciences, Cornell University, Ithaca, New York, 14853, United States of America
- Flow Cytometry Core Facility, Cornell University, Ithaca, New York, 14853, United States of America
- * E-mail:
| |
Collapse
|
25
|
Jensen HA, Bunaciu RP, Varner JD, Yen A. GW5074 and PP2 kinase inhibitors implicate nontraditional c-Raf and Lyn function as drivers of retinoic acid-induced maturation. Cell Signal 2015; 27:1666-75. [PMID: 25817574 PMCID: PMC4529126 DOI: 10.1016/j.cellsig.2015.03.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/05/2015] [Accepted: 03/16/2015] [Indexed: 02/07/2023]
Abstract
The multivariate nature of cancer necessitates multi-targeted therapy, and kinase inhibitors account for a vast majority of approved cancer therapeutics. While acute promyelocytic leukemia (APL) patients are highly responsive to retinoic acid (RA) therapy, kinase inhibitors have been gaining momentum as co-treatments with RA for non-APL acute myeloid leukemia (AML) differentiation therapies, especially as a means to treat relapsed or refractory AML patients. In this study GW5074 (a c-Raf inhibitor) and PP2 (a Src-family kinase inhibitor) enhanced RA-induced maturation of t(15;17)-negative myeloblastic leukemia cells and rescued response in RA-resistant cells. PD98059 (a MEK inhibitor) and Akti-1/2 (an Akt inhibitor) were less effective, but did tend to promote maturation-uncoupled G1/G0 arrest, while wortmannin (a PI3K inhibitor) did not enhance differentiation surface marker expression or growth arrest. PD98059 and Akti-1/2 did not enhance differentiation markers and have potential, antagonistic off-targets effects on the aryl hydrocarbon receptor (AhR), but neither could the AhR agonist 6-formylindolo(3,2-b)carbazole (FICZ) rescue differentiation events in the RA-resistant cells. GW5074 rescued early CD38 expression in RA-resistant cells exhibiting an early block in differentiation before CD38 expression, while for RA-resistant cells with differentiation blocked later, PP2 rescued the later differentiation marker CD11b; but surprisingly, the combination of the two was not synergistic. Kinases c-Raf, Src-family kinases Lyn and Fgr, and PI3K display highly correlated signaling changes during RA treatment, while activation of traditional downstream targets (Akt, MEK/ERK), and even the surface marker CD38, were poorly correlated with c-Raf or Lyn during differentiation. This suggests that an interrelated kinase module involving c-Raf, PI3K, Lyn and perhaps Fgr functions in a nontraditional way during RA-induced maturation or during rescue of RA induction therapy using inhibitor co-treatment in RA-resistant leukemia cells.
Collapse
Affiliation(s)
- Holly A Jensen
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, United States
| | - Rodica P Bunaciu
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States
| | - Jeffrey D Varner
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, United States
| | - Andrew Yen
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States
| |
Collapse
|
26
|
Tryptophan derivatives regulate the transcription of Oct4 in stem-like cancer cells. Nat Commun 2015; 6:7209. [PMID: 26059097 PMCID: PMC4490363 DOI: 10.1038/ncomms8209] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/17/2015] [Indexed: 12/27/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor that responds to environmental toxicants, is increasingly recognized as a key player in embryogenesis and tumorigenesis. Here we show that a variety of tryptophan derivatives that act as endogenous AhR ligands can affect the transcription level of the master pluripotency factor Oct4. Among them, ITE enhances the binding of the AhR to the promoter of Oct4 and suppresses its transcription. Reduction of endogenous ITE levels in cancer cells by tryptophan deprivation or hypoxia leads to Oct4 elevation, which can be reverted by administration with synthetic ITE. Consequently, synthetic ITE induces the differentiation of stem-like cancer cells and reduces their tumorigenic potential in both subcutaneous and orthotopic xenograft tumour models. Thus, our results reveal a role of tryptophan derivatives and the AhR signalling pathway in regulating cancer cell stemness and open a new therapeutic avenue to target stem-like cancer cells.
Collapse
|
27
|
Ibabao CN, Bunaciu RP, Schaefer DMW, Yen A. The AhR agonist VAF347 augments retinoic acid-induced differentiation in leukemia cells. FEBS Open Bio 2015; 5:308-18. [PMID: 25941627 PMCID: PMC4412882 DOI: 10.1016/j.fob.2015.04.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 03/24/2015] [Accepted: 04/02/2015] [Indexed: 01/19/2023] Open
Abstract
In binary cell-fate decisions, driving one lineage and suppressing the other are conjoined. We have previously reported that aryl hydrocarbon receptor (AhR) promotes retinoic acid (RA)-induced granulocytic differentiation of lineage bipotent HL-60 myeloblastic leukemia cells. VAF347, an AhR agonist, impairs the development of CD14(+)CD11b(+) monocytes from granulo-monocytic (GM) stage precursors. We thus hypothesized that VAF347 propels RA-induced granulocytic differentiation and impairs D3-induced monocytic differentiation of HL-60 cells. Our results show that VAF347 enhanced RA-induced cell cycle arrest, CD11b integrin expression and neutrophil respiratory burst. Granulocytic differentiation is known to be driven by MAPK signaling events regulated by Fgr and Lyn Src-family kinases, the CD38 cell membrane receptor, the Vav1 GEF, the c-Cbl adaptor, as well as AhR, all of which are embodied in a putative signalsome. We found that the VAF347 AhR ligand regulates the signalsome. VAF347 augments RA-induced expression of AhR, Lyn, Vav1, and c-Cbl as well as p47(phox). Several interactions of partners in the signalsome appear to be enhanced: Fgr interaction with c-Cbl, CD38, and with pS259c-Raf and AhR interaction with c-Cbl and Lyn. Thus, we report that, while VAF347 impedes monocytic differentiation induced by 1,25-dihydroxyvitamin D3, VAF347 promotes RA-induced differentiation. This effect seems to involve but not to be limited to Lyn, Vav1, c-Cbl, AhR, and Fgr.
Collapse
Key Words
- APC, allophycocyanin
- APL, acute promyelocytic leukemia
- AhR, aryl hydrocarbon receptor
- D3, 1,25-dihydroxyvitamin D3
- Differentiation
- FICZ, 6-formylindolo (3,2-b) carbazole
- GM, granulo-monocytic
- Leukemia
- PE, phycoerythrin
- RA, all trans-retinoic acid
- Retinoic acid
- TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin
- VAF347
- VAF347, (4-(3-Chloro-phenyl)-pyrimidin-2-yl)-(4-trifluoromethyl-phenyl)-amine
Collapse
Affiliation(s)
| | - Rodica P Bunaciu
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Deanna M W Schaefer
- Department of Population Medicine and Diagnostic Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Andrew Yen
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
28
|
Jensen HA, Bunaciu RP, Ibabao CN, Myers R, Varner JD, Yen A. Retinoic acid therapy resistance progresses from unilineage to bilineage in HL-60 leukemic blasts. PLoS One 2014; 9:e98929. [PMID: 24922062 PMCID: PMC4055670 DOI: 10.1371/journal.pone.0098929] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 05/08/2014] [Indexed: 02/06/2023] Open
Abstract
Emergent resistance can be progressive and driven by global signaling aberrations. All-trans retinoic acid (RA) is the standard therapeutic agent for acute promyelocytic leukemia, but 10-20% of patients are not responsive, and initially responsive patients relapse and develop retinoic acid resistance. The patient-derived, lineage-bipotent acute myeloblastic leukemia (FAB M2) HL-60 cell line is a potent tool for characterizing differentiation-induction therapy responsiveness and resistance in t(15;17)-negative cells. Wild-type (WT) HL-60 cells undergo RA-induced granulocytic differentiation, or monocytic differentiation in response to 1,25-dihydroxyvitamin D3 (D3). Two sequentially emergent RA-resistant HL-60 cell lines, R38+ and R38-, distinguishable by RA-inducible CD38 expression, do not arrest in G1/G0 and fail to upregulate CD11b and the myeloid-associated signaling factors Vav1, c-Cbl, Lyn, Fgr, and c-Raf after RA treatment. Here, we show that the R38+ and R38- HL-60 cell lines display a progressive reduced response to D3-induced differentiation therapy. Exploiting the biphasic dynamic of induced HL-60 differentiation, we examined if resistance-related defects occurred during the first 24 h (the early or "precommitment" phase) or subsequently (the late or "lineage-commitment" phase). HL-60 were treated with RA or D3 for 24 h, washed and retreated with either the same, different, or no differentiation agent. Using flow cytometry, D3 was able to induce CD38, CD11b and CD14 expression, and G1/G0 arrest when present during the lineage-commitment stage in R38+ cells, and to a lesser degree in R38- cells. Clustering analysis of cytometry and quantified Western blot data indicated that WT, R38+ and R38- HL-60 cells exhibited decreasing correlation between phenotypic markers and signaling factor expression. Thus differentiation induction therapy resistance can develop in stages, with initial partial RA resistance and moderate vitamin D3 responsiveness (unilineage maturation block), followed by bilineage maturation block and progressive signaling defects, notably the reduced expression of Vav1, Fgr, and c-Raf.
Collapse
Affiliation(s)
- Holly A Jensen
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, United States of America
| | - Rodica P Bunaciu
- Department of Biomedical Sciences, Cornell University, Ithaca, New York, United States of America
| | - Christopher N Ibabao
- Department of Biology, Cornell University, Ithaca, New York, United States of America
| | - Rebecca Myers
- Department of Biology, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Jeffrey D Varner
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, United States of America
| | - Andrew Yen
- Department of Biomedical Sciences, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|