83 results on '"Hongliang Zong"'
Search Results
2. Cranberry A-type proanthocyanidins selectively target acute myeloid leukemia cells
- Author
-
Laura M. Bystrom, Daniel P. Bezerra, Hsiao-Ting Hsu, Hongliang Zong, Luis A. Lara-Martínez, Jeanne P. De Leon, Megan Emmanuel, David Méry, Sara Gardenghi, Duane Hassane, Catherine C. Neto, Susanna Cunningham-Rundles, Michael W. Becker, Stefano Rivella, and Monica L. Guzman
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: Most elderly patients affected with acute myeloid leukemia (AML) will relapse and die of their disease even after achieving complete remission, thus emphasizing the urgent need for new therapeutic approaches with minimum toxicity to normal hematopoietic cells. Cranberry (Vaccinium spp.) extracts have exhibited anticancer and chemopreventive properties that have been mostly attributed to A-type proanthocyanidin (A-PAC) compounds. A-PACs, isolated from a commercially available cranberry extract, were evaluated for their effects on leukemia cell lines, primary AML samples, and normal CD34+ cord blood specimens. Our results indicated potent and specific antileukemia activity in vitro. In addition, the antileukemia activity of A-PACs extended to malignant progenitor and stem cell populations, sparing their normal counterparts. The antileukemia effects of A-PACs were also observed in vivo using patient derived xenografts. Surprisingly, we found that the mechanism of cell death was driven by activation of NF-κB. Overall, our data suggest that A-PACs could be used to improve treatments for AML by targeting leukemia stem cells through a potentially novel pathway.
- Published
- 2019
- Full Text
- View/download PDF
3. Small Molecule Inhibitor of CBFβ-RUNX Binding for RUNX Transcription Factor Driven Cancers
- Author
-
Anuradha Illendula, Jane Gilmour, Jolanta Grembecka, Venkata Sesha Srimath Tirumala, Adam Boulton, Aravinda Kuntimaddi, Charles Schmidt, Lixin Wang, John A. Pulikkan, Hongliang Zong, Mahmut Parlak, Cem Kuscu, Anna Pickin, Yunpeng Zhou, Yan Gao, Lauren Mishra, Mazhar Adli, Lucio H. Castilla, Roger A. Rajewski, Kevin A. Janes, Monica L. Guzman, Constanze Bonifer, and John H. Bushweller
- Subjects
CBFβ ,RUNX ,PPI ,Transcription factor inhibitor ,Leukemia ,Triple negative breast cancer ,Medicine ,Medicine (General) ,R5-920 - Abstract
Transcription factors have traditionally been viewed with skepticism as viable drug targets, but they offer the potential for completely novel mechanisms of action that could more effectively address the stem cell like properties, such as self-renewal and chemo-resistance, that lead to the failure of traditional chemotherapy approaches. Core binding factor is a heterodimeric transcription factor comprised of one of 3 RUNX proteins (RUNX1-3) and a CBFβ binding partner. CBFβ enhances DNA binding of RUNX subunits by relieving auto-inhibition. Both RUNX1 and CBFβ are frequently mutated in human leukemia. More recently, RUNX proteins have been shown to be key players in epithelial cancers, suggesting the targeting of this pathway could have broad utility. In order to test this, we developed small molecules which bind to CBFβ and inhibit its binding to RUNX. Treatment with these inhibitors reduces binding of RUNX1 to target genes, alters the expression of RUNX1 target genes, and impacts cell survival and differentiation. These inhibitors show efficacy against leukemia cells as well as basal-like (triple-negative) breast cancer cells. These inhibitors provide effective tools to probe the utility of targeting RUNX transcription factor function in other cancers.
- Published
- 2016
- Full Text
- View/download PDF
4. A Hyperactive Signalosome in Acute Myeloid Leukemia Drives Addiction to a Tumor-Specific Hsp90 Species
- Author
-
Hongliang Zong, Alexander Gozman, Eloisi Caldas-Lopes, Tony Taldone, Eric Sturgill, Sarah Brennan, Stefan O. Ochiana, Erica M. Gomes-DaGama, Siddhartha Sen, Anna Rodina, John Koren III, Michael W. Becker, Charles M. Rudin, Ari Melnick, Ross L. Levine, Gail J. Roboz, Stephen D. Nimer, Gabriela Chiosis, and Monica L. Guzman
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Acute myeloid leukemia (AML) is a heterogeneous and fatal disease with an urgent need for improved therapeutic regimens given that most patients die from relapsed disease. Irrespective of mutation status, the development of aggressive leukemias is enabled by increasing dependence on signaling networks. We demonstrate that a hyperactive signalosome drives addiction of AML cells to a tumor-specific Hsp90 species (teHsp90). Through genetic, environmental, and pharmacologic perturbations, we demonstrate a direct and quantitative link between hyperactivated signaling pathways and apoptotic sensitivity of AML to teHsp90 inhibition. Specifically, we find that hyperactive JAK-STAT and PI3K-AKT signaling networks are maintained by teHsp90 and, in fact, gradual activation of these networks drives tumors increasingly dependent on teHsp90. Thus, although clinically aggressive AML survives via signalosome activation, this addiction creates a vulnerability that can be exploited with Hsp90-directed therapy.
- Published
- 2015
- Full Text
- View/download PDF
5. Corrigendum to: 'Small Molecule Inhibitor of CBFβ-RUNX Binding for RUNX Transcription Factor Driven Cancers' [EBioMedicine 8 (2016) 117–131]
- Author
-
Anuradha Illendula, Jane Gilmour, Jolanta Grembecka, Venkata Sesha Srimath Tirumala, Adam Boulton, Aravinda Kuntimaddi, Charles Schmidt, Lixin Wang, John A. Pulikkan, Hongliang Zong, Mahmut Parlak, Cem Kuscu, Anna Pickin, Yunpeng Zhou, Yan Gao, Lauren Mishra, Mazhar Adli, Lucio H. Castilla, Roger A. Rajewski, Kevin A. Janes, Monica L. Guzman, Constanze Bonifer, and John H. Bushweller
- Subjects
Medicine ,Medicine (General) ,R5-920 - Published
- 2017
- Full Text
- View/download PDF
6. BCL6 maintains survival and self-renewal of primary human acute myeloid leukemia cells
- Author
-
Mayumi Sugita, Kimihito Cojin Kawabata, Martin Carroll, Sarah Wyman, Bas J. Wouters, Zhengming Chen, Michael Albert, Winnie Yip, Cem Meydan, Srinjoy Goswami, Hongliang Zong, Ruud Delwel, Ari Melnick, Gail J. Roboz, Monica L. Guzman, Christopher E. Mason, and Hematology
- Subjects
0301 basic medicine ,Indoles ,Myeloid ,Apoptosis ,Mice, SCID ,Proto-Oncogene Mas ,Biochemistry ,Mice ,0302 clinical medicine ,Mice, Inbred NOD ,immune system diseases ,hemic and lymphatic diseases ,RNA, Neoplasm ,RNA-Seq ,Cell Self Renewal ,RNA, Small Interfering ,Tumor Stem Cell Assay ,Myeloid Neoplasia ,Gene Expression Regulation, Leukemic ,Cytarabine ,Myeloid leukemia ,Hematology ,BCL6 ,Acquired immune system ,Neoplasm Proteins ,Leukemia, Myeloid, Acute ,Leukemia ,Haematopoiesis ,medicine.anatomical_structure ,Gene Knockdown Techniques ,Radiation Chimera ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Proto-Oncogene Proteins c-bcl-6 ,RNA Interference ,Stem cell ,medicine.drug ,Immunology ,Antineoplastic Agents ,Biology ,03 medical and health sciences ,medicine ,Animals ,Humans ,RNA, Messenger ,Gene Expression Profiling ,Cell Biology ,Hematopoietic Stem Cells ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Cancer research ,Thiazolidinediones - Abstract
B-cell lymphoma 6 (BCL6) is a transcription repressor and proto-oncogene that plays a crucial role in the innate and adaptive immune system and lymphoid neoplasms. However, its role in myeloid malignancies remains unclear. Here, we explored the role of BCL6 in acute myeloid leukemia (AML). BCL6 was expressed at variable and often high levels in AML cell lines and primary AML samples. AMLs with higher levels of BCL6 were generally sensitive to treatment with BCL6 inhibitors, with the exception of those with monocytic differentiation. Gene expression profiling of AML cells treated with a BCL6 inhibitor revealed induction of BCL6-repressed target genes and transcriptional programs linked to DNA damage checkpoints and downregulation of stem cell genes. Ex vivo treatment of primary AML cells with BCL6 inhibitors induced apoptosis and decreased colony-forming capacity, which correlated with the levels of BCL6 expression. Importantly, inhibition or knockdown of BCL6 in primary AML cells resulted in a significant reduction of leukemia-initiating capacity in mice, suggesting ablation of leukemia repopulating cell functionality. In contrast, BCL6 knockout or inhibition did not suppress the function of normal hematopoietic stem cells. Treatment with cytarabine further induced BCL6 expression, and the levels of BCL6 induction were correlated with resistance to cytarabine. Treatment of AML patient-derived xenografts with BCL6 inhibitor plus cytarabine suggested enhanced antileukemia activity with this combination. Hence, pharmacologic inhibition of BCL6 might provide a novel therapeutic strategy for ablation of leukemia-repopulating cells and increased responsiveness to chemotherapy. Key Points: • Primary human AML cells with higher levels of BCL6 expression displayed sensitivity when exposed to BCL6 inhibitors. • BCL6 induction contributed to cytarabine resistance, and inhibition of BCL6 in AML cells decreased leukemia stem cell activity.
- Published
- 2021
- Full Text
- View/download PDF
7. Synthesis and properties of a novel UV/moisture dual-curable polyurethane resin derived from vinyl-based polyether polyol
- Author
-
Zhongxiang Lin, Hongliang Zong, Qiming Yan, Cheng Fang, and Xing Lin
- Subjects
chemistry.chemical_classification ,Materials science ,Moisture ,030206 dentistry ,02 engineering and technology ,Surfaces and Interfaces ,General Chemistry ,021001 nanoscience & nanotechnology ,Surfaces, Coatings and Films ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Polyol ,chemistry ,Chemical engineering ,Mechanics of Materials ,Materials Chemistry ,0210 nano-technology ,Polyurethane - Abstract
A novel UV/moisture dual-curable polyurethane resin has been synthesized via a two-step process. In the first step, methoxysilane-modified polyether polyol (SPOL) was prepared through the thiolene ...
- Published
- 2020
- Full Text
- View/download PDF
8. A novel polyether polyol contains repeating cyclohexane units and its application on reactive polyurethane adhesive
- Author
-
Zhongxiang Lin, Hongliang Zong, Cheng Fang, Qiming Yan, and Xing Lin
- Subjects
chemistry.chemical_classification ,chemistry.chemical_compound ,Materials science ,Polymers and Plastics ,Cyclohexane ,chemistry ,Polyol ,Polymer chemistry ,Polyurethane adhesive ,Cyclohexene oxide - Published
- 2020
- Full Text
- View/download PDF
9. FDA-approved ferumoxytol displays anti-leukaemia efficacy against cells with low ferroportin levels
- Author
-
Gail J. Roboz, Mohamed O. Rabie, Vicenta Trujillo-Alonso, Valerie A. Longo, Andres Lara-Martinez, Charalambos Kaittanis, Monica L. Guzman, Edwin C. Pratt, Jan Grimm, Michael W. Becker, and Hongliang Zong
- Subjects
Myeloid ,Iron ,Ferroportin ,Biomedical Engineering ,Bioengineering ,02 engineering and technology ,010402 general chemistry ,medicine.disease_cause ,01 natural sciences ,Article ,In vivo ,hemic and lymphatic diseases ,Humans ,Medicine ,General Materials Science ,Electrical and Electronic Engineering ,Cation Transport Proteins ,Leukemia ,biology ,business.industry ,Iron deficiency ,021001 nanoscience & nanotechnology ,Condensed Matter Physics ,medicine.disease ,Ferrosoferric Oxide ,Atomic and Molecular Physics, and Optics ,3. Good health ,0104 chemical sciences ,Ferumoxytol ,medicine.anatomical_structure ,Cancer research ,biology.protein ,Experimental pathology ,0210 nano-technology ,business ,Oxidative stress - Abstract
Acute myeloid leukaemia (AML) is a fatal disease for most patients. We have found that ferumoxytol (Feraheme®), a FDA approved iron oxide nanoparticle for iron deficiency treatment, demonstrates an anti-leukaemia effect in vitro and in vivo. Using leukaemia cell lines and primary AML patient samples, we show that low expression of the iron exporter ferroportin (FPN) results in a susceptibility of these cells by an increase in intracellular iron from ferumoxytol. The reactive oxygen species produced by free ferrous iron leads to increased oxidative stress and cell death. Ferumoxytol treatment results in a significant reduction of disease burden in a murine leukaemia model and patient-derived xenotransplants (PDX) bearing leukaemia cells with low FPN expression. Our findings show how a clinical nanoparticle considered previously largely biologically inert could be rapidly incorporated into clinical trials for patients with leukaemia with low FPN levels., One Sentence Summary: Administration of the clinically approved iron oxide nanoparticle drug ferumoxytol in vitro results in an anti-leukaemia effect and in vivo extended overall survival in part due to the low expression of the iron export protein ferroportin.
- Published
- 2019
- Full Text
- View/download PDF
10. 117 Rapid point-of-care subcutaneous CAR-T from blood draw to injection in 4 hours with modified LV encoding CARs and synthetic driver elements enables efficient CAR-T expansion and tumor regression
- Author
-
Gregory I. Frost, Qun He, Michelle Andraza, Wei Zhang, Anirban Kundu, Ewa Jaruga-Killeen, Frederic Vigant, Gregory Schreiber, Hongliang Zong, and Alissa R. Kerner
- Subjects
biology ,medicine.diagnostic_test ,business.industry ,CD22 ,Epitope ,Chimeric antigen receptor ,CD19 ,Raji cell ,Flow cytometry ,Subcutaneous injection ,medicine.anatomical_structure ,Cancer research ,medicine ,biology.protein ,business ,human activities ,Lymph node - Abstract
Background Adoptive cellular therapy with chimeric antigen receptor (CAR)-T cells has demonstrated remarkable clinical activity in a number of hematologic malignancies, but product chain of custody, individualized manufacturing, preparative chemotherapy, and patient management present technical and logistical hurdles to broader implementation. Methods Lentiviral constructs for CARs (either CD19- or CD22-directed) co-expressed with a synthetic driver domain were identified from a >6 × 10 diversity combinatorial library of proliferative elements, transmembrane domains, leucine zippers, and an EGFR epitope screened for cellular expansion in a lymphoreplete model. Modified serum-free-lentiviral manufacturing process was developed to reduce complexity of CAR-T and to introduce CD3-activating elements into the viral envelope allowing activation and transduction of resting lymphocytes from peripheral blood. Results Four-hour exposure of as little as 1 ml of blood to the CD3-directed CD19-targeted CAR encoding lentivirus followed by subcutaneous injection in NSG mice bearing CD19+/CD22+ Raji cells resulted in tumor regression (figure 1) and robust CAR-T cell expansion as determined by flow cytometry (figure 2) and qPCR (table 1), with peak levels >10,000 CAR-T cells/µl and less than three CAR copies per genome. In contrast, administration of the same products intravenously failed to support significant CAR-T expansion or control tumor growth (figure 3). Regression of established Raji tumors was also observed in NSG-(KbDb) (IA) animals following SC administration of CD19 or CD22 CARs with driver domains. CAR-T cells contracted in peripheral blood following tumor regression. Regression of Raji tumor from the initial median volume of 151 mm3 throughout 40 days post subcutaneous administration of the LV transduced (at MOI 1 or 5) CD19-directed CAR T product (1M or 5M cells) in the NSG mice Conclusions We conclude that through a synthetic subcutaneous lymph node approach with modified lentiviruses and driver domains, rPOC SC may enable CAR-T generation with reduced complexity, while maintaining the ability of CAR-T cells to expand, persist and exert anti-tumor activity. Ethics Approval All animal studies were IACUC approved.
- Published
- 2020
- Full Text
- View/download PDF
11. Allogeneic TCRαβ deficient CAR T-cells targeting CD123 in acute myeloid leukemia
- Author
-
Mayumi Sugita, Roman Galetto, Hongliang Zong, Nathan Ewing-Crystal, Vicenta Trujillo-Alonso, Nuria Mencia-Trinchant, Winnie Yip, Stephanie Filipe, Celine Lebuhotel, Agnès Gouble, Duane C. Hassane, Julianne Smith, Gail J. Roboz, and Monica L. Guzman
- Subjects
Leukemia, Myeloid, Acute ,Mice ,Multidisciplinary ,hemic and lymphatic diseases ,Receptors, Antigen, T-Cell, alpha-beta ,T-Lymphocytes ,Hematopoietic Stem Cell Transplantation ,Interleukin-3 Receptor alpha Subunit ,General Physics and Astronomy ,Animals ,Humans ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology - Abstract
Acute myeloid leukemia (AML) is a disease with high incidence of relapse that is originated and maintained from leukemia stem cells (LSCs). Hematopoietic stem cells can be distinguished from LSCs by an array of cell surface antigens such as CD123, thus a candidate to eliminate LSCs using a variety of approaches, including CAR T cells. Here, we evaluate the potential of allogeneic gene-edited CAR T cells targeting CD123 to eliminate LSCs (UCART123). UCART123 cells are TCRαβneg T cells generated from healthy donors using TALEN® gene-editing technology, decreasing the likelihood of graft vs host disease. As safety feature, cells express RQR8 to allow elimination with Rituximab. UCART123 effectively eliminates AML cells in vitro and in vivo with significant benefits in overall survival of AML-patient derived xenograft mice. Furthermore, UCART123 preferentially target AML over normal cells with modest toxicity to normal hematopoietic stem/progenitor cells. Together these results suggest that UCART123 represents an off-the shelf therapeutic approach for AML.
- Published
- 2020
12. Identification of a new p53 responsive element in the promoter region of anillin
- Author
-
Wenqing Lu, Hongliang Zong, Xinying Liu, Xinxin Shen, Ruixiang Ma, Pengyi Liu, and Jiao Ma
- Subjects
Untranslated region ,Chromatin Immunoprecipitation ,DNA damage ,Down-Regulation ,Biology ,Response Elements ,medicine.disease_cause ,Contractile Proteins ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Luciferases ,Promoter Regions, Genetic ,Gene ,Mutation ,Binding Sites ,Cell Cycle ,Promoter ,General Medicine ,Cell cycle ,HCT116 Cells ,Cell biology ,MCF-7 Cells ,Tumor Suppressor Protein p53 ,Chromatin immunoprecipitation ,Cytokinesis ,DNA Damage ,Protein Binding - Abstract
The expression of anillin mRNA and protein is regulated in a cell cycle‑dependent manner. However, the mechanism underlying this process is unclear. Previous studies analyzing the sequence of the 5'‑untranslated region of anillin have unveiled several putative p53 binding sites. Therefore, the present study hypothesized that the anillin gene may be repressed by p53 and that the commonly observed mutation (or loss of function) of p53 may serve a role in this phenotype. Bioinformatic analysis of the anillin promoter region revealed potential p53 responsive elements. Of those identified, 2 were able to bind p53 protein, as determined via a chromatin immunoprecipitation assay. Although it was hypothesized that DNA damage and resultant p53 expression would repress anillin expression, the results revealed that anillin mRNA and protein expression levels were negatively regulated by DNA damage in the wild‑type p53 cells, but not in the isogenic p53 null cells. Furthermore, DNA sequences encompassing the p53 binding site downregulated luciferase transgenes in a p53 dependent manner. Taken together, these data indicated that anillin was negatively regulated by p53 and that anillin overexpression observed in cancer may be a p53‑mediated phenomenon. The data from the present study provided further evidence for the role of p53 in the biologically crucial process of cytokinesis.
- Published
- 2020
- Full Text
- View/download PDF
13. Abstract 1511: Subcutaneous injection of total nucleated cells rapidly isolated following four-hour peripheral whole blood exposure to CD3-directed CAR-T lentiviruses with a synthetic driver results in robust CAR-T proliferation and anti-tumor immunity
- Author
-
Qun He, Sid P. Kerkar, Hongliang Zong, Ewa Jaruga-Killeen, Michelle Andraza, Wei Zhang, Gregory Schreiber, Anirban Kundu, Gregory I. Frost, Dongming Zhang, Frederic Vigant, and Alissa R. Kerner
- Subjects
Cancer Research ,biology ,medicine.diagnostic_test ,CD3 ,Peripheral blood mononuclear cell ,Molecular biology ,CD19 ,In vitro ,Flow cytometry ,Immunophenotyping ,Oncology ,Cell culture ,biology.protein ,medicine ,Whole blood - Abstract
Background: Adoptive cell therapies using Chimeric Antigen Receptors (CARs) show durable clinical benefit for patients with hematologic malignancies, however, challenges remain for enabling this personalized treatment to be delivered in a timely, cost effective, and logistically friendly manner. Methods: Lentiviral vectors (LV) encoding CD19 and CD22 CARs with a synthetic driver element were packaged with a VSV-G envelope designed with the capability to target and activate CD3pos T cells in whole blood. LV were directly reconstituted in peripheral whole blood for four hours and total nucleated cells (TNC) or peripheral blood mononuclear cells (PBMC) were rapidly isolated utilizing two different closed systems. Immediately following the four-hour exposure event, isolated TNC or PBMC were injected subcutaneously into mice with disseminated Raji luciferase tumor cells. For further characterization, LV-exposed TNC and PBMC were cultured in vitro for six days and functionally examined. Results: Following four-hour exposure to CD3-directed LVs, more than 90% of T cells, including naïve/naïve derived (CCR7pos CD45ROneg) and central memory (CCR7pos CD45ROpos) T cells present within isolated TNC or PBMC exhibited a significant decrease in CD3 surface expression. Subcutaneous injection of gene modified TNC or PBMC resulted in the in vivo generation and expansion of large numbers of circulating CAR-T positive cells with complete eradication of disseminated Raji tumors. In parallel cell culture experiments, TNC or PBMC isolated following four-hour LV whole blood exposure exhibited robust expansion without additional T-cell receptor (TCR) or CD3 stimulation, while TNC or PBMC not exposed to virus did not show any expansion. Following six days in culture, immunophenotyping by flow cytometry demonstrated that more than 90% of the cells were CD8pos and CD4pos T cells with CAR-T expression present on central memory (CCR7pos CD45ROpos) and effector memory (CCR7neg CD45ROpos) T cells. CAR-T antigen specificity to CD19 and CD22 was measured by IFN-gamma release co-culture assays. Conclusion: We conclude that large numbers of functionally active CAR-T positive cells can be generated both in vitro and in vivo following a four-hour peripheral whole blood exposure to CD3-directed LVs encoding for CARs with a synthetic driver element. These results provide the basis for an autologous same-day peripheral blood draw to subcutaneous injection rapid point-of-care (rPOC) approach. Citation Format: Dongming Zhang, Frederic Vigant, Qun He, Anirban Kundu, Wei Zhang, Hongliang Zong, Ewa Jaruga-Killeen, Gregory Schreiber, Michelle Andraza, Alissa R. Kerner, Gregory I. Frost, Sid P. Kerkar. Subcutaneous injection of total nucleated cells rapidly isolated following four-hour peripheral whole blood exposure to CD3-directed CAR-T lentiviruses with a synthetic driver results in robust CAR-T proliferation and anti-tumor immunity [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1511.
- Published
- 2021
- Full Text
- View/download PDF
14. A-type proanthocyanidins selectively target acute myeloid leukemia cells in vitro and in vivo
- Author
-
Monica L. Guzman, Catherine C. Neto, Lal Martinez, Gail J. Roboz, Hongliang Zong, Laura M. Bystrom, and Stefano Rivella
- Subjects
Pharmacology ,Complementary and alternative medicine ,Proanthocyanidin ,Chemistry ,In vivo ,Organic Chemistry ,Drug Discovery ,Cancer research ,Pharmaceutical Science ,Molecular Medicine ,Myeloid leukemia ,In vitro ,Analytical Chemistry - Published
- 2016
- Full Text
- View/download PDF
15. The epichaperome is an integrated chaperome network that facilitates tumour survival
- Author
-
Oscar Lin, Robert Trondl, Brad Beattie, Ari Melnick, Anna Rodina, Alexander Bolaender, Ross L. Levine, Ethel Cesarman, Pat Zanzonico, Gabriela Chiosis, Pengrong Yan, Chenghua Yang, Jason S. Lewis, Sujata Patil, Tony Taldone, John F. Gerecitano, Leandro Cerchietti, Sarah Kishinevsky, Clifford A. Hudis, Radu I Peter, Fumiko Shimizu, Monica L. Guzman, Matthew Riolo, Mohammad F. Farooq, Erica DaGama Gomes, John Koren, Hardik J. Patel, Hediye Erdjument-Bromage, Gail J. Roboz, Nagavarakishore Pillarsetty, Lorenz Studer, Mark Dunphy, Tai Wang, Hongliang Zong, Shanu Modi, Palak Panchal, Eloisi Caldas-Lopes, Mary L. Alpaugh, Christina Pressl, Chao Xu, Steven M. Larson, Feixia Chu, and Adriana D. Corben
- Subjects
0301 basic medicine ,Protein family ,Cell Survival ,Genes, myc ,Cellular homeostasis ,Antineoplastic Agents ,Protein degradation ,Biology ,Bioinformatics ,Article ,Malignant transformation ,Mice ,03 medical and health sciences ,Cell Line, Tumor ,Neoplasms ,Heat shock protein ,Drug Discovery ,Animals ,Humans ,HSP70 Heat-Shock Proteins ,HSP90 Heat-Shock Proteins ,Multidisciplinary ,Drug discovery ,Hsp90 ,Cell biology ,Crosstalk (biology) ,030104 developmental biology ,Organ Specificity ,Multiprotein Complexes ,biology.protein ,Female ,Molecular Chaperones - Abstract
Transient, multi-protein complexes are important facilitators of cellular functions. This includes the chaperome, an abundant protein family comprising chaperones, co-chaperones, adaptors, and folding enzymes—dynamic complexes of which regulate cellular homeostasis together with the protein degradation machinery1–6. Numerous studies have addressed the role of chaperome members in isolation, yet little is known about their relationships regarding how they interact and function together in malignancy7–17. As function is probably highly dependent on endogenous conditions found in native tumours, chaperomes have resisted investigation, mainly due to the limitations of methods needed to disrupt or engineer the cellular environment to facilitate analysis. Such limitations have led to a bottleneck in our understanding of chaperome-related disease biology and in the development of chaperome-targeted cancer treatment. Here we examined the chaperome complexes in a large set of tumour specimens. The methods used maintained the endogenous native state of tumours and we exploited this to investigate the molecular characteristics and composition of the chaperome in cancer, the molecular factors that drive chaperome networks to crosstalk in tumours, the distinguishing factors of the chaperome in tumours sensitive to pharmacologic inhibition, and the characteristics of tumours that may benefit from chaperome therapy. We find that under conditions of stress, such as malignant transformation fuelled by MYC, the chaperome becomes biochemically ‘rewired’ to form a network of stable, survival-facilitating, high-molecular-weight complexes. The chaperones heat shock protein 90 (HSP90) and heat shock cognate protein 70 (HSC70) are nucleating sites for these physically and functionally integrated complexes. The results indicate that these tightly integrated chaperome units, here termed the epichaperome, can function as a network to enhance cellular survival, irrespective of tissue of origin or genetic background. The epichaperome, present in over half of all cancers tested, has implications for diagnostics and also provides potential vulnerability as a target for drug intervention.
- Published
- 2016
- Full Text
- View/download PDF
16. Small Molecule Inhibitor of CBFβ-RUNX Binding for RUNX Transcription Factor Driven Cancers
- Author
-
Lauren Mishra, Mahmut Parlak, Adam Boulton, Cem Kuscu, Venkata Sesha Srimath Tirumala, Mazhar Adli, Yunpeng Zhou, John H. Bushweller, Anuradha Illendula, Yan Gao, Anna Pickin, Charles Schmidt, Jane Gilmour, Kevin A. Janes, John Anto Pulikkan, Roger A. Rajewski, Aravinda Kuntimaddi, Jolanta Grembecka, Lucio H. Castilla, Hongliang Zong, Constanze Bonifer, Monica L. Guzman, and Lixin Wang
- Subjects
0301 basic medicine ,Core Binding Factor alpha Subunits ,PPI ,lcsh:Medicine ,Biology ,Core binding factor ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,chemistry.chemical_compound ,Medicine, General & Internal ,hemic and lymphatic diseases ,medicine ,Triple negative breast cancer ,Transcription factor ,Gene ,Triple-negative breast cancer ,lcsh:R5-920 ,Leukemia ,CBFβ ,lcsh:R ,General Medicine ,medicine.disease ,Molecular biology ,RUNX ,3. Good health ,Cell biology ,030104 developmental biology ,RUNX1 ,chemistry ,Stem cell ,Transcription factor inhibitor ,lcsh:Medicine (General) - Abstract
Transcription factors have traditionally been viewed with skepticism as viable drug targets, but they offer the potential for completely novel mechanisms of action that could more effectively address the stem cell like properties, such as self-renewal and chemo-resistance, that lead to the failure of traditional chemotherapy approaches. Core binding factor is a heterodimeric transcription factor comprised of one of 3 RUNX proteins (RUNX1-3) and a CBFβ binding partner. CBFβ enhances DNA binding of RUNX subunits by relieving auto-inhibition. Both RUNX1 and CBFβ are frequently mutated in human leukemia. More recently, RUNX proteins have been shown to be key players in epithelial cancers, suggesting the targeting of this pathway could have broad utility. In order to test this, we developed small molecules which bind to CBFβ and inhibit its binding to RUNX. Treatment with these inhibitors reduces binding of RUNX1 to target genes, alters the expression of RUNX1 target genes, and impacts cell survival and differentiation. These inhibitors show efficacy against leukemia cells as well as basal-like (triple-negative) breast cancer cells. These inhibitors provide effective tools to probe the utility of targeting RUNX transcription factor function in other cancers.
- Published
- 2016
17. Cranberry A-type proanthocyanidins selectively target acute myeloid leukemia cells
- Author
-
Monica L. Guzman, Luis A. Lara-Martinez, Stefano Rivella, Hsiao-Ting Hsu, Sara Gardenghi, Jeanne P. De Leon, Hongliang Zong, Daniel P. Bezerra, Laura M. Bystrom, Duane C. Hassane, Michael W. Becker, David E Mery, Susanna Cunningham-Rundles, Catherine C. Neto, and Megan Emmanuel
- Subjects
0301 basic medicine ,RM ,Myeloid ,Cell Survival ,CD34 ,RV ,Q1 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Proanthocyanidins ,Dose-Response Relationship, Drug ,business.industry ,Plant Extracts ,Myeloid leukemia ,Hematology ,medicine.disease ,R1 ,Antineoplastic Agents, Phytogenic ,Xenograft Model Antitumor Assays ,Stimulus Report ,humanities ,Leukemia ,Haematopoiesis ,Disease Models, Animal ,Leukemia, Myeloid, Acute ,030104 developmental biology ,medicine.anatomical_structure ,Vaccinium macrocarpon ,Cell culture ,030220 oncology & carcinogenesis ,Cord blood ,Cancer research ,Stem cell ,business ,RB - Abstract
Most elderly patients affected with acute myeloid leukemia (AML) will relapse and die of their disease even after achieving complete remission, thus emphasizing the urgent need for new therapeutic approaches with minimum toxicity to normal hematopoietic cells. Cranberry (Vaccinium spp.) extracts have exhibited anticancer and chemopreventive properties that have been mostly attributed to A-type proanthocyanidin (A-PAC) compounds. A-PACs, isolated from a commercially available cranberry extract, were evaluated for their effects on leukemia cell lines, primary AML samples, and normal CD34(+) cord blood specimens. Our results indicated potent and specific antileukemia activity in vitro. In addition, the antileukemia activity of A-PACs extended to malignant progenitor and stem cell populations, sparing their normal counterparts. The antileukemia effects of A-PACs were also observed in vivo using patient derived xenografts. Surprisingly, we found that the mechanism of cell death was driven by activation of NF-κB. Overall, our data suggest that A-PACs could be used to improve treatments for AML by targeting leukemia stem cells through a potentially novel pathway.
- Published
- 2018
18. In vivo targeting of leukemia stem cells by directing parthenolide-loaded nanoparticles to the bone marrow niche
- Author
-
Mauro Ferrari, Siddhartha Sen, Chaofeng Mu, Hongliang Zong, David G. Gorenstein, Monica L. Guzman, Xuewu Liu, Peter A. Crooks, Guodong Zhang, Gail J. Roboz, Zaineb A.F. Albayati, and Haifa Shen
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Pathology ,Myeloid ,Antineoplastic Agents ,Mice, SCID ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Delivery Systems ,Bone Marrow ,Mice, Inbred NOD ,In vivo ,Cell Line, Tumor ,Internal medicine ,Animals ,Humans ,Medicine ,Parthenolide ,Stem Cell Niche ,Hematology ,business.industry ,medicine.disease ,Xenograft Model Antitumor Assays ,3. Good health ,Leukemia, Myeloid, Acute ,Leukemia ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,chemistry ,Neoplastic Stem Cells ,Cancer research ,Nanoparticles ,Bone marrow ,Stem cell ,business ,Sesquiterpenes - Abstract
In vivo targeting of leukemia stem cells by directing parthenolide-loaded nanoparticles to the bone marrow niche
- Published
- 2015
- Full Text
- View/download PDF
19. Synthesis, anticancer activity and molecular docking studies on a series of heterocyclic trans-cyanocombretastatin analogues as antitubulin agents
- Author
-
Narsimha Reddy Penthala, Venumadhav Janganati, Hongliang Zong, Robert L. Eoff, Peter A. Crooks, Amit Ketkar, Nikhil Reddy Madadi, and Monica L. Guzman
- Subjects
Stereochemistry ,Antineoplastic Agents ,Article ,Tubulin binding ,Structure-Activity Relationship ,symbols.namesake ,chemistry.chemical_compound ,Heterocyclic Compounds ,Tubulin ,Cell Line, Tumor ,Stilbenes ,Drug Discovery ,Humans ,Colchicine ,Molecule ,Moiety ,Cell Proliferation ,Pharmacology ,Combretastatin ,Dose-Response Relationship, Drug ,Molecular Structure ,biology ,Organic Chemistry ,General Medicine ,Tubulin Modulators ,Molecular Docking Simulation ,chemistry ,Cell culture ,biology.protein ,symbols ,Drug Screening Assays, Antitumor ,van der Waals force - Abstract
A series of heterocyclic combretastatin analogues have been synthesized and evaluated for their anticancer activity against a panel of 60 human cancer cell lines. The most potent compounds were two 3,4,5-trimethoxy phenyl analogues containing either an (Z)-indol-2-yl (8) or (Z)-benzo[b]furan-2-yl (12) moiety; these compounds exhibited GI50 values of 50%. The binding modes of the three most active compounds (8, 12 and 29) to tubulin were also investigated utilizing molecular docking studies. All three molecules were observed to bind in the same hydrophobic pocket at the interface of α- and β-tubulin that is occupied by colchicine, and were stablized by van der Waals’ interactions with surrounding tubulin residues. The results from the tubulin polymerization and molecular docking studies indicate that compounds 8 and 29 are the most potent anti-leukemic compounds in this structural class, and are considered lead compounds for further development as anti-leukemic drugs.
- Published
- 2015
- Full Text
- View/download PDF
20. A small-molecule inhibitor of the aberrant transcription factor CBFβ-SMMHC delays leukemia in mice
- Author
-
Roger A. Rajewski, Hongliang Zong, Adam Boulton, Yunpeng Zhou, John Anto Pulikkan, Aravinda Kuntimaddi, Anuradha Illendula, John H. Bushweller, Yan Gao, Liting Xue, Monica L. Guzman, Siddhartha Sen, Jolanta Grembecka, and Lucio H. Castilla
- Subjects
Myeloid ,Oncogene Proteins, Fusion ,Antineoplastic Agents ,Biology ,Core binding factor ,Article ,Small Molecule Libraries ,Mice ,chemistry.chemical_compound ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Protein Interaction Maps ,Transcription factor ,Multidisciplinary ,Myeloid leukemia ,medicine.disease ,Fusion protein ,Mice, Inbred C57BL ,Leukemia, Myeloid, Acute ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,RUNX1 ,chemistry ,Core Binding Factor Alpha 2 Subunit ,Immunology ,Cancer research ,Benzimidazoles ,Female - Abstract
Toward drugging the undruggable in cancer Many human cancers are characterized by inappropriate activity of transcription factors. These proteins are attractive drug targets in principle, but normalizing their function requires drugs that modulate specific protein-protein interactions, a goal that has been challenging. In acute myeloid leukemia, a chromosomal translocation creates an aberrant form of the transcription factor CBF-beta, which outcompetes “normal” CBF-beta for binding to another transcription factor called RUNX1, thereby deregulating its activity. Illendula et al. identified and chemically optimized a small molecule that selectively disrupts the interaction between the aberrant CBF-beta and RUNX1 (see the Perspective by Koehler and Chen). This molecule restored normal gene expression patterns and delayed leukemia progression in mice. Thus, transcription factors may not be as undruggable as once thought. Science , this issue p. 779 ; see also p. 713
- Published
- 2015
- Full Text
- View/download PDF
21. Synthesis and evaluation of a series of resveratrol analogues as potent anti-cancer agents that target tubulin
- Author
-
Robert L. Eoff, Shobanbabu Bommagani, Hongliang Zong, Amit Ketkar, Chen Zheng, Narsimha Reddy Penthala, Nikhil Reddy Madadi, Peter A. Crooks, Venumadhav Janganati, and Monica L. Guzman
- Subjects
Pharmacology ,biology ,Chemistry ,Organic Chemistry ,Pharmaceutical Science ,Myeloid leukemia ,Cancer ,Resveratrol ,medicine.disease ,Biochemistry ,Article ,3. Good health ,chemistry.chemical_compound ,Tubulin ,Cell culture ,Microtubule ,Drug Discovery ,Cancer research ,biology.protein ,medicine ,Molecular Medicine ,Cytotoxic T cell ,Binding site - Abstract
A series of novel diarylacrylonitrile and trans-stilbene analogues of resveratrol has been synthesized and evaluated for their anticancer activities against a panel of 60 human cancer cell lines. The diarylacrylonitrile analogues 3b and 4a exhibited the most potent anticancer activity of all the analogues synthesized in this study, with GI50 values of < 10 nM against almost all the cell lines in the human cancer cell panel. Compounds 3b and 4a were also screened against the acute myeloid leukemia (AML) cell line, MV4-11, and were found to have potent cytotoxic properties that are likely mediated through inhibition of tubulin polymerization. Results from molecular docking studies indicate a common binding site for 4a and 3b on the 3,3-tubulin heterodimer, with a slightly more favorable binding for 3b compared to 4a; this is consistent with the results from the microtubule assays, which demonstrate that 4a is more potent than 3b in inhibiting tubulin polymerization in MV4-11 cells. Taken together, these data suggest that diarylacrylonitriles 3b and 4a may have potential as antitubulin therapeutics for treatment of both solid and hematological tumors.
- Published
- 2015
- Full Text
- View/download PDF
22. Affinity-based proteomics reveal cancer-specific networks coordinated by Hsp90
- Author
-
Kamalika Moulick, Len Neckers, Tony Taldone, Jason S. Lewis, Monica L. Guzman, Xinyang Zhao, James H. Ahn, Leandro Cerchietti, Fabiana Perna, Steven S. Gross, Peter Smith-Jones, Kristin Beebe, Danuta Zatorska, Gabriela Chiosis, Hediye Erdjument-Bromage, Nagavarakishore Pillarsetty, Ly P. Vu, Anna Rodina, Mary L. Alpaugh, Ari Melnick, Stephen D. Nimer, Gomes-Dagama Erica M, Hongliang Zong, Eloisi Caldas-Lopes, Katerina Hatzi, Steven M. Larson, Thomas Ku, and Ross L. Levine
- Subjects
Proteomics ,Antineoplastic Agents ,Biology ,Interactome ,Article ,Cell Line, Tumor ,Neoplasms ,Drug Discovery ,medicine ,Animals ,Humans ,Benzodioxoles ,HSP90 Heat-Shock Proteins ,Molecular Biology ,Regulation of gene expression ,Drug discovery ,Computational Biology ,Myeloid leukemia ,Cancer ,Cell Biology ,medicine.disease ,Cell biology ,Gene Expression Regulation, Neoplastic ,Purines ,Proteome ,Signal transduction ,Signal Transduction - Abstract
Most cancers are characterized by multiple molecular alterations, but identification of the key proteins involved in these signaling pathways is currently beyond reach. We show that the inhibitor PU-H71 preferentially targets tumor-enriched Hsp90 complexes and affinity captures Hsp90-dependent oncogenic client proteins. We have used PU-H71 affinity capture to design a proteomic approach that, when combined with bioinformatic pathway analysis, identifies dysregulated signaling networks and key oncoproteins in chronic myeloid leukemia. The identified interactome overlaps with the well-characterized altered proteome in this cancer, indicating that this method can provide global insights into the biology of individual tumors, including primary patient specimens. In addition, we show that this approach can be used to identify previously uncharacterized oncoproteins and mechanisms, potentially leading to new targeted therapies. We further show that the abundance of the PU-H71-enriched Hsp90 species, which is not dictated by Hsp90 expression alone, is predictive of the cell’s sensitivity to Hsp90 inhibition.
- Published
- 2011
- Full Text
- View/download PDF
23. AGEs, RAGE, and Diabetic Retinopathy
- Author
-
Micheal S. Ward, Hongliang Zong, and Alan W. Stitt
- Subjects
Glycation End Products, Advanced ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Receptor for Advanced Glycation End Products ,Inflammation ,Models, Biological ,RAGE (receptor) ,Proinflammatory cytokine ,Glycation ,Diabetes mellitus ,Internal medicine ,Internal Medicine ,medicine ,Animals ,Humans ,Receptors, Immunologic ,Retina ,Diabetic Retinopathy ,business.industry ,Diabetic retinopathy ,medicine.disease ,medicine.anatomical_structure ,Endocrinology ,medicine.symptom ,business ,Signal Transduction ,Retinopathy - Abstract
Diabetic retinopathy is a major diabetic complication with a highly complex etiology. Although there are many pathways involved, it has become established that chronic exposure of the retina to hyperglycemia gives rise to accumulation of advanced glycation end products (AGEs) that play an important role in retinopathy. In addition, the receptor for AGEs (RAGE) is ubiquitously expressed in various retinal cells and is upregulated in the retinas of diabetic patients, resulting in activation of pro-oxidant and proinflammatory signaling pathways. This AGE-RAGE axis appears to play a central role in the sustained inflammation, neurodegeneration, and retinal microvascular dysfunction occurring during diabetic retinopathy. The nature of AGE formation and RAGE signaling bring forward possibilities for therapeutic intervention. The multiple components of the AGE-RAGE axis, including signal transduction, formation of ligands, and the end-point effectors, may be promising targets for strategies to treat diabetic retinopathy.
- Published
- 2011
- Full Text
- View/download PDF
24. Thr-370 Is Responsible for CDK11p58 Autophosphorylation, Dimerization, and Kinase Activity
- Author
-
Hongliang Zong, Xiaojing Yun, Yi Hong, Jianxin Gu, Chun-Yi Zhang, Weiying Zou, Yanlin Wang, Yayun Chi, Haiou Liu, and Xiangfei Kong
- Subjects
Transcriptional Activation ,Mutation, Missense ,Apoptosis ,Biology ,Mitogen-activated protein kinase kinase ,Biochemistry ,MAP2K7 ,Humans ,Phosphorylation ,Kinase activity ,Molecular Biology ,MAP kinase kinase kinase ,Cyclin-dependent kinase 4 ,Cyclin-dependent kinase 2 ,Autophosphorylation ,Tryptophan ,Cell Biology ,Cyclin-Dependent Kinases ,Cell biology ,Amino Acid Substitution ,biology.protein ,Cyclin-dependent kinase 9 ,Protein Multimerization ,HeLa Cells ,Signal Transduction - Abstract
CDK11(p58), a member of the p34(cdc2)-related kinase family, is associated with cell cycle progression, tumorigenesis, and proapoptotic signaling. It is also required for the maintenance of chromosome cohesion, the maturation of centrosome, the formation of bipolar spindle, and the completion of mitosis. Here we identified that CDK11(p58) interacted with itself to form homodimers in cells, whereas D224N, the kinase-dead mutant, failed to form homodimers. CDK11(p58) was autophosphorylated, and the main functions of CDK11(p58), such as kinase activity, transactivation of nuclear receptors, and proapoptotic signal transduction, were dependent on its autophosphorylation. Furthermore, the in vitro kinase assay indicated that CDK11(p58) was autophosphorylated at Thr-370. By mutagenesis, we created CDK11(p58) T370A and CDK11(p58) T370D, which mimic the dephosphorylated and phosphorylated forms of CDK11(p58), respectively. The T370A mutant could not form dimers and be phosphorylated by the wild type CDK11(p58) and finally lost the kinase activity. Further functional research revealed that T370A failed to repress the transactivation of androgen receptor and enhance the cell apoptosis. Overall, our data indicated that Thr-370 is responsible for the autophosphorylation, dimerization, and kinase activity of CDK11(p58). Moreover, Thr-370 mutants might affect CDK11(p58)-mediated signaling pathways.
- Published
- 2011
- Full Text
- View/download PDF
25. Hyperglycaemia-induced pro-inflammatory responses by retinal Müller glia are regulated by the receptor for advanced glycation end-products (RAGE)
- Author
-
Hongliang Zong, Phaik Har Yong, Ga Limb, Angelina Madden, Micheal S. Ward, Alan W. Stitt, and Tim M. Curtis
- Subjects
Male ,MAPK/ERK pathway ,medicine.medical_specialty ,endocrine system diseases ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Receptor for Advanced Glycation End Products ,Biology ,Retina ,Streptozocin ,Cell Line ,Diabetes Mellitus, Experimental ,RAGE (receptor) ,Rats, Sprague-Dawley ,Diabetic Neuropathies ,Downregulation and upregulation ,Glycation ,Internal medicine ,Internal Medicine ,medicine ,Animals ,Humans ,cardiovascular diseases ,Receptors, Immunologic ,Receptor ,Diabetic Retinopathy ,Glial fibrillary acidic protein ,Retinitis ,nutritional and metabolic diseases ,Rats ,Glucose ,Endocrinology ,Cytokine ,medicine.anatomical_structure ,Hyperglycemia ,cardiovascular system ,biology.protein ,Inflammation Mediators ,Neuroglia ,human activities ,Muller glia - Abstract
Up-regulation of the receptor for AGEs (RAGE) and its ligands in diabetes has been observed in various tissues. Here, we sought to determine levels of RAGE and one of its most important ligands, S100B, in diabetic retina, and to investigate the regulatory role of S100B and RAGE in Muller glia. Streptozotocin-diabetes was induced in Sprague–Dawley rats. RAGE, S100B and glial fibrillary acidic protein (GFAP) were detected in retinal cryosections. In parallel, the human retinal Muller cell line, MIO-M1, was maintained in normal glucose (5.5 mmol/l) or high glucose (25 mmol/l). RAGE knockdown was achieved using small interfering RNA (siRNA), while soluble RAGE was used as a competitive inhibitor of RAGE ligand binding. RAGE, S100B and cytokines were detected using quantitative RT-PCR, western blotting, cytokine protein arrays or ELISA. Activation of mitogen-activated protein kinase (MAPK) by RAGE was determined by western blotting. Compared with non-diabetic controls, RAGE and S100B were significantly elevated in the diabetic retina with apparent localisation in the Muller glia, occurring concomitantly with upregulation of GFAP. Exposure of MIO-M1 cells to high glucose induced increased production of RAGE and S100B. RAGE signalling via MAPK pathway was linked to cytokine production. Blockade of RAGE prevented cytokine responses induced by high glucose and S100B in Muller glia. Hyperglycaemia in vivo and in vitro exposure to high glucose induce upregulation of RAGE and its ligands, leading to RAGE signalling, which links to pro-inflammatory responses by retinal Muller glia. These data shed light on the potential clinical application of RAGE blockade to inhibit the progression of diabetic retinopathy.
- Published
- 2010
- Full Text
- View/download PDF
26. Cdc34-mediated Degradation of ATF5 Is Blocked by Cisplatin
- Author
-
Jin Zhou, Xiaojing Yun, Jianxin Gu, Yuanyan Wei, Xiaoning Chen, Si Zhang, Hongliang Zong, Dan Liu, and Jianhai Jiang
- Subjects
Cytoplasm ,Proteasome Endopeptidase Complex ,Active Transport, Cell Nucleus ,Activating transcription factor ,Down-Regulation ,Antineoplastic Agents ,Apoptosis ,Protein tag ,Ubiquitin-conjugating enzyme ,CREB ,Biochemistry ,Anaphase-Promoting Complex-Cyclosome ,Cell Line ,Ubiquitin ,Humans ,E2F1 ,Molecular Biology ,Transcription factor ,Cell Nucleus ,biology ,Ubiquitination ,Ubiquitin-Protein Ligase Complexes ,Cell Differentiation ,Cell Biology ,Activating Transcription Factors ,Ubiquitin-Conjugating Enzymes ,biology.protein ,Cisplatin - Abstract
ATF5, a member of activating transcription factor (ATF)/cAMP-response element-binding protein (CREB) family of b-ZIP transcription factors, contributes to neural cell differentiation and is involved in cell apoptosis in response to cisplatin and a number of environment factors. However, the mechanisms governing the regulation of ATF5 protein during apoptosis are largely unknown. In this study we reported that ATF5 protein was a substrate of the ubiquitin-proteasome pathway. Interestingly, the ubiquitin-dependent degradation of exogenous ATF5 protein was independent of lysine residues. Instead, the addition of a large N-terminal enhanced green fluorescence protein tag increased the stability of ATF5 protein, and the free amino acid group of the N-terminal methionine of ATF5 protein was a site for ubiquitinylation, indicating that exogenous ATF5 was degraded via the ubiquitin-proteasome system through N-terminal ubiquitinylation. Furthermore, cisplatin increased ATF5 protein expression via preventing its ubiquitin-dependent degradation, which might be associated with its promoting the nucleus-to-cytoplasm translocation of E2 ubiquitin-conjugating enzyme Cdc34 and reducing the interaction between ATF5 and Cdc34. In summary, a down-regulation of proteasome-mediated degradation of ATF5 might contribute to cisplatin-induced apoptosis, providing a new mechanism of cisplatin-induced apoptosis.
- Published
- 2008
- Full Text
- View/download PDF
27. Cyclin-dependent kinase 11p58interacts with HBO1 and enhances its histone acetyltransferase activity
- Author
-
Xiaojing Yun, Yayun Chi, Jianhai Jiang, Hanzhou Wang, Xiaoyun Shen, Liyun Liu, Yi Hong, Jianxin Gu, Zejuan Li, Hongliang Zong, Yun Hu, and Ziyue Niu
- Subjects
PITSLRE ,Transcription, Genetic ,Biophysics ,Protein Serine-Threonine Kinases ,Biology ,Biochemistry ,Transcriptional regulation ,Acetyltransferases ,Structural Biology ,Cyclin-dependent kinase ,Two-Hybrid System Techniques ,Protein Interaction Mapping ,Tumor Cells, Cultured ,Genetics ,medicine ,Humans ,Histone acetyltransferase activity ,Molecular Biology ,Gene Library ,Histone Acetyltransferases ,Sequence Deletion ,Cell Nucleus ,Cyclin-dependent kinase 1 ,Eukaryotic transcription ,Cell Biology ,Histone acetyltransferase ,Molecular biology ,Cyclin-Dependent Kinases ,Cell biology ,Cell nucleus ,Histone ,medicine.anatomical_structure ,HBO1 ,biology.protein ,Protein Kinases ,CDK11p58 - Abstract
CDK11(p58), a 58kDa protein of the PITSLRE kinase family, plays an important role in cell cycle progression, and is closely related to cell apoptosis. To gain further insight into the function of CDK11(p58), we screened a human fetal liver cDNA library for its interacting proteins using the yeast two-hybrid system. Here we report that histone acetyltransferase (HAT) HBO1, a MYST family protein, interacts with CDK11(p58) in vitro and in vivo. CDK11(p58) and HBO1 colocalize in the cell nucleus. Recombinant CDK11(p58) enhances the HAT activity of HBO1 significantly in vitro. Meanwhile, overexpression of CDK11(p58) in mammalian cells leads to the enhanced HAT activity of HBO1 towards free histones. Thus, we conclude that CDK11(p58) is a new interacting protein and a novel regulator of HBO1. Both of the proteins may be involved in the regulation of eukaryotic transcription.
- Published
- 2005
- Full Text
- View/download PDF
28. Elevated β1,4-Galactosyltransferase I in Highly Metastatic Human Lung Cancer Cells
- Author
-
Hongliang Zong, Zejuan Li, Xiaoning Chen, Hailian Shen, Jianxin Gu, Xiaoyu Zhu, Ziyue Niu, Jianhai Jiang, Weicheng Liu, Hanzhou Wang, Yanzhong Yang, and Yun Hu
- Subjects
Small interfering RNA ,Mutation ,Expression vector ,Cell ,Cell Biology ,Transfection ,Biology ,medicine.disease_cause ,Biochemistry ,Molecular biology ,medicine.anatomical_structure ,Epidermal growth factor ,medicine ,Electrophoretic mobility shift assay ,Cell adhesion ,Molecular Biology - Abstract
The elevated levels of β1,4-galactosyltransferase I (GalT I; EC 2.4.1.38) are detected in highly metastatic lung cancer PGBE1 cells compared with its less metastatic partner PGLH7 cells. Decreasing the GalT I surface expression by small interfering RNA or interfering with the surface of GalT I function by mutation inhibited cell adhesion on laminin, the invasive potential in vitro, and tyrosine phosphorylation of focal adhesion kinase. The mechanism by which GalT I activity is up-regulated in highly metastatic cells remains unclear. To investigate the regulation of GalT I expression, we cloned the 5′-region flanking the transcription start point of the GalT I gene (–1653 to +52). Cotransfection of the GalT I promoter/luciferase reporter and the Ets family protein E1AF expression plasmid increased the luciferase reporter activity in a dose-dependent manner. By deletion and mutation analyses, we identified an Ets-binding site between nucleotides –205 and –200 in the GalT I promoter that was critical for responsiveness to E1AF. It was identified that E1AF could bind to and activate the GalT I promoter by electrophoretic mobility shift assay in PGLH7 cells and COS1 cells. A stronger affinity of E1AF for DNA has contributed to the elevated expression of GalT I in PGBE1 cells. Stable transfection of the E1AF expression plasmid resulted in increased GalT I expression in PGLH7 cells, and stable transfectants migrated faster than control cells. Meanwhile, the content of the β1,4-Gal branch on the cell surface was increased in stably transfected PGLH7 cells. GalT I expression can also be induced by epidermal growth factor and dominant active Ras, JNK1, and ERK1. These data suggest an essential role for E1AF in the activation of the human GalT I gene in highly metastatic lung cancer cells.
- Published
- 2005
- Full Text
- View/download PDF
29. Downregulation of β1,4-galactosyltransferase 1 inhibits CDK11p58-mediated apoptosis induced by cycloheximide
- Author
-
Jianxin Gu, Qing Sun, Jianhai Jiang, Zejuan Li, Xiangfei Kong, Hongliang Zong, and Hanzhou Wang
- Subjects
Cell ,Biophysics ,Down-Regulation ,Apoptosis ,Cycloheximide ,Mitochondrion ,Biochemistry ,chemistry.chemical_compound ,Downregulation and upregulation ,Cell Line, Tumor ,medicine ,Humans ,Viability assay ,Molecular Biology ,biology ,Caspase 3 ,Kinase ,Cytochrome c ,Cytochromes c ,Cell Biology ,Galactosyltransferases ,Caspase Inhibitors ,Molecular biology ,Cyclin-Dependent Kinases ,Mitochondria ,Cell biology ,Isoenzymes ,Molecular Weight ,medicine.anatomical_structure ,chemistry ,Caspases ,biology.protein ,RNA Interference ,Poly(ADP-ribose) Polymerases - Abstract
Cyclin-dependent kinase 11 (CDK11; also named PITSLRE) is part of the large family of p34(cdc2)-related kinases whose functions appear to be linked with cell cycle progression, tumorigenesis, and apoptotic signaling. The mechanism that CDK11(p58) induces apoptosis is not clear. Some evidences suggested beta1,4-galactosyltransferase 1 (beta1,4-GT 1) might participate in apoptosis induced by CDK11(p58). In this study, we demonstrated that ectopically expressed beta1,4-GT 1 increased CDK11(p58)-mediated apoptosis induced by cycloheximide (CHX). In contrast, RNAi-mediated knockdown of beta1,4-GT 1 effectively inhibited apoptosis induced by CHX in CDK11(p58)-overexpressing cells. For example, the cell morphological and nuclear changes were reduced; the loss of cell viability was prevented and the number of cells in sub-G1 phase was decreased. Knock down of beta1,4-GT 1 also inhibited the release of cytochrome c from mitochondria and caspase-3 processing. Therefore, the cleavage of CDK11(p58) by caspase-3 was reduced. We proposed that beta1,4-GT 1 might contribute to the pro-apoptotic effect of CDK11(p58). This may represent a new mechanism of beta1,4-GT 1 in CHX-induced apoptosis of CDK11(p58)-overexpressing cells.
- Published
- 2005
- Full Text
- View/download PDF
30. Overexpression of small glutamine-rich TPR-containing protein promotes apoptosis in 7721 cells
- Author
-
Jianhai Jiang, Zejuan Li, Hongyan Yin, Yanlin Wang, Hanzhou Wang, Jianxin Gu, Hongliang Zong, and Hailian Shen
- Subjects
Biophysics ,Gene Expression ,Apoptosis ,TPR ,Caspase 3 ,Biochemistry ,Structural Biology ,Cell Line, Tumor ,Chlorocebus aethiops ,Gene expression ,Genetics ,Animals ,Humans ,Heat shock protein 70 ,Molecular Biology ,Cell Proliferation ,Regulation of gene expression ,Gene knockdown ,Heat shock cognate protein 70 ,biology ,Cytochrome c ,Cell Cycle ,Cytochromes c ,Proteins ,Cell Biology ,Cell cycle ,Molecular biology ,Mitochondria ,Gene Expression Regulation, Neoplastic ,Small glutamine-rich TPR-containing protein ,Caspases ,biology.protein ,Ectopic expression ,Poly(ADP-ribose) Polymerases ,Carrier Proteins ,Molecular Chaperones - Abstract
It is known that small glutamine-rich TPR-containing protein (SGT) is the member of TPR motif family. However, the biological functions of SGT remain unclear. In this paper, we report that SGT plays a role in apoptotic signaling. Ectopic expression of SGT enhances DNA fragment and nucleus breakage after the induction of apoptosis. Increasing mRNA level of SGT is also observed in 7721 cells undergoing apoptosis, knockdown the expression of endogenous SGT contributes to the decrease of apoptosis of 7721 cells. Deletion analysis reveals that TPR domain is critical to pro-apoptotic function of SGT. Furthermore, we demonstrated that the PARP cleavage and cytochrome c release are enhanced when SGT is overexpressed in 7721 cells during apoptosis. Collectively, our results indicate that SGT is a new pro-apoptotic factor.
- Published
- 2005
- Full Text
- View/download PDF
31. Identification of the p28 subunit of eukaryotic initiation factor 3(eIF3k) as a new interaction partner of cyclin D3
- Author
-
Jianxin Gu, Maoyun Sun, Yanzhong Yang, Hongliang Zong, Weicheng Liu, Jianhai Jiang, and Xiaoyun Shen
- Subjects
Cyclin E ,Interaction ,Eukaryotic Initiation Factor-3 ,Cyclin D ,Molecular Sequence Data ,Cyclin A ,Biophysics ,Cyclin B ,Biochemistry ,Structural Biology ,Cyclin-dependent kinase ,Cyclins ,Two-Hybrid System Techniques ,Translation initiation ,Genetics ,Humans ,Amino Acid Sequence ,Cyclin D3 ,Molecular Biology ,Binding Sites ,biology ,Cell Biology ,Molecular biology ,eIF3k ,Protein Subunits ,Protein Transport ,Protein Biosynthesis ,biology.protein ,Cyclin-dependent kinase complex ,Sequence Alignment ,Cyclin A2 ,HeLa Cells ,Protein Binding - Abstract
Cyclin D3 is found to play a crucial role not only in progression through the G1 phase as a regulatory subunit of cyclin-dependent kinase 4 (CDK 4) and CDK 6, but also in many other aspects such as cell cycle, cell differentiation, transcriptional regulation and apoptosis. In this work, we screened a human fetal liver cDNA library using human cyclin D3 as bait and identified human eukaryotic initiation factor 3 p28 protein (eIF3k) as a partner of cyclin D3. The association of cyclin D3 with eIF3k was further confirmed by in vitro binding assay, in vivo coimmunoprecipitation, and confocal microscopic analysis. We found that cyclin D3 specifically interacted with eIF3k through its C-terminal domain. Immunofluorescence experiments showed that eIF3k distributed both in nucleus and cytoplasm and colocalized with cyclin D3. In addition, the cellular translation activity in HeLa cells was upregulated by cyclin D3 overexpression and the mRNA levels are constant. These data provide a new clue to our understanding of the cellular function of cyclin D3.
- Published
- 2004
- Full Text
- View/download PDF
32. The β-(1→6)-branched β-(1→3) glucohexaose and its analogues containing an α-(1→3)-linked bond have similar stimulatory effects on the mouse spleen as Lentinan
- Author
-
Jun Yan, Hongliang Zong, Xiaosong Gu, Xianglei Yin, Aiguo Shen, Jianxin Gu, She Chen, Xiaoyun Shen, and Weicheng Liu
- Subjects
Ratón ,Immunology ,Lentinan ,Shiitake Mushrooms ,Gene Expression ,Oligosaccharides ,Spleen ,Drug Administration Schedule ,Mice ,chemistry.chemical_compound ,Concanavalin A ,medicine ,Splenocyte ,Animals ,Immunology and Allergy ,RNA, Messenger ,Northern blot ,Hematoxylin ,Pharmacology ,Mice, Inbred BALB C ,Staining and Labeling ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Tumor Necrosis Factor-alpha ,Organ Size ,Blotting, Northern ,In vitro ,Blot ,Glucose ,medicine.anatomical_structure ,Carbohydrate Sequence ,Biochemistry ,chemistry ,biology.protein ,Cytokines ,Eosine Yellowish-(YS) ,Female ,Cell Division ,Injections, Intraperitoneal - Abstract
The stimulatory effects of the synthetic beta-(1-->6)-branched beta-(1-->3) glucohexaose and its analogues containing an alpha-(1-->3)-linked bond on the mouse spleen were studied for elucidation of the mechanism of their antitumor activity, and their stimulatory effects were compared with Lentinan. The mouse spleen's weight was increased after the intraperitoneal (i.p.) injection of the oligosaccharides compared with the saline group. In addition, routinely hematoxylin and eosin (HE)-stained spleen sections showed that the injection also changed the spleen's histopathology. RNA samples were isolated from splenocytes of oligosaccharides, Lentinan or saline-injected mice. Reverse transcription-polymerase chain reaction (RT-PCR) and Northern blot showed that the administration of the oligosaccharides or Lentinan enhanced mouse spleen mRNA production of TNF-alpha but not IL-2. The injection also enhanced Concanavalin A (Con A)-induced mouse splenocytes proliferation, but the in vitro administration of the oligosaccharides did not have the proliferation-enhancing effect. Taken together, these results suggest that the synthetic beta-(1-->6)-branched beta-(1-->3) glucohexaose and its analogues containing an alpha-(1-->3)-linked bond have similar stimulatory effects as Lentinan. Additionally, they may exert their antitumor effects through the induction of splenocytes mediated immune responses.
- Published
- 2003
- Full Text
- View/download PDF
33. The C-terminal Kinase Domain of the p34cdc2-related PITSLRE Protein Kinase (p110C) Associates with p21-activated Kinase 1 and Inhibits Its Activity during Anoikis
- Author
-
Xiaoyu Zhu, Xianglei Yin, Shuying Ji, Chun Chen, Jun Yan, Jianxin Gu, She Chen, Zhenghong Yuan, Mingmei Cai, Zonghou Shen, Yun Hu, Hongliang Zong, and Songwen Zhang
- Subjects
Molecular Sequence Data ,CHO Cells ,Protein Serine-Threonine Kinases ,Mitogen-activated protein kinase kinase ,Biochemistry ,MAP2K7 ,Mice ,Cricetinae ,Animals ,Anoikis ,ASK1 ,c-Raf ,Molecular Biology ,DNA Primers ,Microscopy, Confocal ,Base Sequence ,MAP kinase kinase kinase ,biology ,Cyclin-dependent kinase 2 ,3T3 Cells ,Cell Biology ,Molecular biology ,Cyclin-Dependent Kinases ,Cell biology ,p21-Activated Kinases ,biology.protein ,Cyclin-dependent kinase 9 ,Signal Transduction - Abstract
The PITSLRE protein kinases are parts of the large family of p34cdc2-related kinases. During apoptosis induced by some stimuli, specific PITSLRE isoforms are cleaved by caspase to produce a protein that contains the C-terminal kinase domain of the PITSLRE proteins (p110C). The p110C induces apoptosis when it is ectopically expressed in Chinese hamster ovary cells. In our study, similar induction of this p110C was observed during anoikis in NIH3T3 cells. To investigate the molecular mechanism of apoptosis mediated by p110C, we used the yeast two-hybrid system to screen a human fetal liver cDNA library and identified p21-activated kinase 1 (PAK1) as an interacting partner of p110C. The association of p110C with PAK1 was further confirmed by in vitro binding assay, in vivo coimmunoprecipitation, and confocal microscope analysis. The interaction of p110C with PAK1 occurred within the residues 210-332 of PAK1. Neither association between p58PITSLRE or p110PITSLRE and PAK1 nor association between p110C and PAK2 or PAK3 was observed. Anoikis was increased and PAK1 activity was inhibited when NIH3T3 cells were transfected with p110C. Furthermore, the binding of p110C with PAK1 and inhibition of PAK1 activity were also observed during anoikis. Taken together, these data suggested that PAK1 might participate in the apoptotic pathway mediated by p110C.
- Published
- 2003
- Full Text
- View/download PDF
34. Corrigendum to: 'Small Molecule Inhibitor of CBFβ-RUNX Binding for RUNX Transcription Factor Driven Cancers' [EBioMedicine 8 (2016) 117–131]
- Author
-
Mahmut Parlak, Aravinda Kuntimaddi, Lucio H. Castilla, John Anto Pulikkan, Anuradha Illendula, Kevin A. Janes, Anna Pickin, Adam Boulton, Jane Gilmour, Hongliang Zong, Lixin Wang, Constanze Bonifer, Roger A. Rajewski, Cem Kuscu, Lauren Mishra, Monica L. Guzman, John H. Bushweller, Venkata Sesha Srimath Tirumala, Mazhar Adli, Yan Gao, Jolanta Grembecka, Charles Schmidt, and Yunpeng Zhou
- Subjects
Models, Molecular ,PPI ,Molecular Conformation ,lcsh:Medicine ,Antineoplastic Agents ,General Biochemistry, Genetics and Molecular Biology ,Core Binding Factor beta Subunit ,Structure-Activity Relationship ,Text mining ,Allosteric Regulation ,hemic and lymphatic diseases ,Cell Line, Tumor ,Neoplasms ,Drug Discovery ,Humans ,Triple negative breast cancer ,Protein Interaction Domains and Motifs ,Transcription factor ,Nuclear Magnetic Resonance, Biomolecular ,lcsh:R5-920 ,Leukemia ,business.industry ,Chemistry ,CBFβ ,lcsh:R ,Core Binding Factor alpha Subunits ,Cell Differentiation ,General Medicine ,Small molecule ,RUNX ,Gene Expression Regulation, Neoplastic ,Gene Knockdown Techniques ,Mutation ,Cancer research ,Drug Screening Assays, Antitumor ,Protein Multimerization ,Transcription factor inhibitor ,lcsh:Medicine (General) ,business ,Corrigendum ,Research Paper ,Protein Binding ,Signal Transduction - Abstract
Transcription factors have traditionally been viewed with skepticism as viable drug targets, but they offer the potential for completely novel mechanisms of action that could more effectively address the stem cell like properties, such as self-renewal and chemo-resistance, that lead to the failure of traditional chemotherapy approaches. Core binding factor is a heterodimeric transcription factor comprised of one of 3 RUNX proteins (RUNX1-3) and a CBFβ binding partner. CBFβ enhances DNA binding of RUNX subunits by relieving auto-inhibition. Both RUNX1 and CBFβ are frequently mutated in human leukemia. More recently, RUNX proteins have been shown to be key players in epithelial cancers, suggesting the targeting of this pathway could have broad utility. In order to test this, we developed small molecules which bind to CBFβ and inhibit its binding to RUNX. Treatment with these inhibitors reduces binding of RUNX1 to target genes, alters the expression of RUNX1 target genes, and impacts cell survival and differentiation. These inhibitors show efficacy against leukemia cells as well as basal-like (triple-negative) breast cancer cells. These inhibitors provide effective tools to probe the utility of targeting RUNX transcription factor function in other cancers., Graphical Abstract Image 10, Highlights • Small molecule inhibitors of CBFβ-RUNX protein-protein interaction developed. • Inhibitors alter occupancy of RUNX1 on target genes and alter their expression. • Inhibitors show efficacy against leukemia cell lines and basal-like (triple negative) breast cancer cell lines. Transcription factors are proteins that bind to DNA and regulate how much of other proteins are made. We describe the development of inhibitors of the interaction between two transcription factors, CBFβ and RUNX. Both these transcription factors are the targets of alterations in human leukemia as well as in a number of solid tumors. The inhibitor changes the behavior of the RUNX transcription factor and alters the levels of proteins it regulates. We show these inhibitors may have potential utility for leukemia as well as one specific type of breast cancer which has a very poor prognosis.
- Published
- 2017
35. Pharmacologic inhibition of the Menin-MLL interaction blocks progression of MLL leukemia in vivo
- Author
-
Jay L. Hess, Ting Zhao, Trupta Purohit, Morgan Jones, Bo Wen, Gwenn Danet-Desnoyers, Tomasz Cierpicki, Shihan He, Dale L. Bixby, Katarzyna Kempińska, Jingya Wang, Jennifer Chase, Monica L. Guzman, Hongliang Zong, Bhavna Malik, Hongzhi Miao, Jonathan Pollock, Dmitry Borkin, Andrew G. Muntean, Jolanta Grembecka, Duxin Sun, Ivan Maillard, and Moshe Talpaz
- Subjects
Cancer Research ,Oncogene Proteins ,Oncogene Proteins, Fusion ,Drug Evaluation, Preclinical ,Chromosomal translocation ,Antineoplastic Agents ,Biology ,Leukemogenic ,Mice ,In vivo ,hemic and lymphatic diseases ,Proto-Oncogene Proteins ,medicine ,Tumor Cells, Cultured ,Effective treatment ,Animals ,Humans ,neoplasms ,Cell Biology ,Histone-Lysine N-Methyltransferase ,medicine.disease ,Fusion protein ,Molecular biology ,3. Good health ,Hematopoiesis ,Mice, Inbred C57BL ,Leukemia ,Oncology ,Cancer research ,Disease Progression ,Myeloid-Lymphoid Leukemia Protein ,Female - Abstract
SummaryChromosomal translocations affecting mixed lineage leukemia gene (MLL) result in acute leukemias resistant to therapy. The leukemogenic activity of MLL fusion proteins is dependent on their interaction with menin, providing basis for therapeutic intervention. Here we report the development of highly potent and orally bioavailable small-molecule inhibitors of the menin-MLL interaction, MI-463 and MI-503, and show their profound effects in MLL leukemia cells and substantial survival benefit in mouse models of MLL leukemia. Finally, we demonstrate the efficacy of these compounds in primary samples derived from MLL leukemia patients. Overall, we demonstrate that pharmacologic inhibition of the menin-MLL interaction represents an effective treatment for MLL leukemias in vivo and provide advanced molecular scaffold for clinical lead identification.
- Published
- 2014
36. Characterization of ibrutinib-sensitive and -resistant mantle lymphoma cells
- Author
-
Shuhua Cheng, Pin Lu, Peter Martin, Morton Coleman, Y. Lynn Wang, Hongliang Zong, Jiao Ma, and Ailin Guo
- Subjects
Cell Survival ,MAP Kinase Signaling System ,Antineoplastic Agents ,Lymphoma, Mantle-Cell ,Biology ,chemistry.chemical_compound ,Piperidines ,immune system diseases ,hemic and lymphatic diseases ,medicine ,Agammaglobulinaemia Tyrosine Kinase ,Tumor Cells, Cultured ,Bruton's tyrosine kinase ,Humans ,Phosphorylation ,Protein kinase B ,Cell Proliferation ,Cell Death ,Adenine ,Cell Cycle ,breakpoint cluster region ,Hematology ,Protein-Tyrosine Kinases ,medicine.disease ,Pyrimidines ,chemistry ,Cell culture ,Drug Resistance, Neoplasm ,Ibrutinib ,Gene Knockdown Techniques ,Immunology ,Cancer research ,Refractory Mantle Cell Lymphoma ,biology.protein ,Pyrazoles ,Mantle cell lymphoma - Abstract
Ibrutinib inhibits Bruton tyrosine kinase (BTK), a key component of early B-cell receptor (BCR) signalling pathways. A multicentre phase 2 trial of ibrutinib in patients with relapsed/refractory mantle cell lymphoma (MCL) demonstrated a remarkable response rate. However, approximately one-third of patients have primary resistance to the drug while other patients appear to lose response and develop secondary resistance. Understanding the molecular mechanisms underlying ibrutinib sensitivity is of paramount importance. In this study, we investigated cell lines and primary MCL cells that display differential sensitivity to ibrutinib. We found that the primary cells display a higher BTK activity than normal B cells and MCL cells show differential sensitivity to BTK inhibition. Genetic knockdown of BTK inhibits the growth, survival and proliferation of ibrutinib-sensitive but not resistant MCL cell lines, suggesting that ibrutinib acts through BTK to produce its anti-tumour activities. Interestingly, inhibition of ERK1/2 and AKT, but not BTK phosphorylation per se, correlates well with cellular response to BTK inhibition in cell lines as well as in primary tumours. Our study suggests that, to prevent primary resistance or to overcome secondary resistance to BTK inhibition, a combinatory strategy that targets multiple components or multiple pathways may represent the most effective approach.
- Published
- 2014
37. Allogeneic Tcrα/β Deficient CAR T-Cells Targeting CD123 Prolong Overall Survival of AML Patient-Derived Xenografts
- Author
-
Linda Lam, Monica L. Guzman, Nathan Ewing-Crystal, Duane C. Hassane, Nuria Mencia-Trinchant, Agnès Gouble, Vicenta Trujillo-Alonso, Gail J. Roboz, Mayumi Sugita, Roman Galetto, Julianne Smith, Nicole M. Cruz, and Hongliang Zong
- Subjects
0301 basic medicine ,business.industry ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,03 medical and health sciences ,Leukemia ,Haematopoiesis ,030104 developmental biology ,0302 clinical medicine ,Graft-versus-host disease ,medicine.anatomical_structure ,Cytokine ,030220 oncology & carcinogenesis ,Cord blood ,medicine ,Cancer research ,Interleukin-3 receptor ,Bone marrow ,Stem cell ,business - Abstract
Acute myeloid leukemia (AML) is a disease with a high incidence of relapse and mortality. Relapse is attributed to the inability of current chemotherapeutic agents to eliminate leukemia stem cells (LSCs). Thus, to improve leukemia therapy, it is critical to identify agents that effectively target LSCs, e.g. via unique cell surface antigens. A target of major interest is CD123, the transmembrane alpha chain of the interleukin-3 receptor, expressed on blasts, leukemic progenitor and LSCs in the majority of AML patients. We have developed an allogeneic chimeric antigen receptor (CAR) T-cell platform using T-cells from third-party healthy donors to generate engineered T-cells targeting CD123 (UCART123). UCART123 cells no longer express a TCR, having undergone a disruption of the TCRα gene using TALEN¨ gene editing technology followed by elimination of TCRα/β-positive cells, thus minimizing the potential for engineered T-cells to cause graft versus host disease (GvHD). We tested the activity of UCART123 cells in vitro using primary AML samples, normal bone marrow (nBM) and cord blood (CB) cells. Additionally, we established patient derived xenograft (PDX), nBM- or CB- humanized xenografts (HuX) and a competitive nBM/AML xenograft model to evaluate the in vivo potential of UCART123 cells to preferentially eliminate AML over normal BM cells. In vitro studies reveled that UCART123 cells eliminate AML cells and had minimum effect on normal cells at effector:target ratios as low as 0.5:1. Next, we evaluated the in vivo activity of UCART123 against PDX (AML37, TP53 mutant relapsed AML and AML20, FLT3-ITD+ and TP53 mutant AML) and normal-HuX mice (n=3). At 3 weeks post T-cell injection we found that UCART123 treatment eliminated the leukemic cells when using 10M or 3M UCART123 cells per mouse and no significant difference between PBS or TCR-deficient T-cells (TCRkoT; 10M/mouse). Toxicity to normal cells was dose dependent, doses of 2.5M UCART123 cells did not significantly affect hematopoietic cells. T-cells were detected in the BM at day 14 after treatment, without evidence of GvHD. Since we found complete elimination of human AML cells in the BM of the PDX precluding serial transplantation to evaluate LSC activity, we initiated two new sets of PDX-AML mice [AML2 (NPM1+FLT3-ITD+) and AML37 (TP53 mutant)] to evaluate long-term survival, and time to relapse. Animals were treated with PBS, UCART123 (2.5M or 1M), TCRkoT (2.5M), or Ara-C (60mg/kg 5 days). Animal weight and peripheral blood (PB) was monitored. Cytokines changes were evaluated at day 2. We found that the cytokine release and the kinetics of AML targeting by UCART123 were dose dependent. We found a significant overall survival (OS) benefit with UCART123 in both PDX tested. For example, all PDX-AML2 mice treated with UCART123 are alive to date (day 167; updates will be presented). All other cohorts were lost (PBS day124, TCRkoT day126, AraC day144) (Figure 1A). Finally, to determine selectivity of UCART123 cells for AML cells over nBM cells, we generated a competitive model bearing both nBM and AML (NPM1+FLT3-ITD+). With PB monitoring, treatment with 1M UCAR123 cells resulted in selective elimination of AML cells. Untreated and TCRkoT treated mice showed a rapid progression of AML, while treated mice showed normal hematopoiesis (Figure 1B). NPM1 transcripts were also monitored in the mice and confirmed molecular remission in mice. Taken together, our data show that UCART123, an "off-the-shelf" allogeneic engineered CAR-T product targeting CD123 potently eliminates AML cells in vivo, prevents relapse, and improves OS in PDX mice. Also, UCART123 cells preferentially targets AML cells in a competitive BM/AML model. A phase I trial of UCART123 in AML is under development. Disclosures Guzman: Cellectis: Research Funding. Sugita:Cellectis: Research Funding. Galetto:Cellectis SA: Employment. Gouble:Cellectis: Employment. Smith:Cellectis SA: Employment. Roboz:Agios, Amgen, Amphivena, Astex, AstraZeneca, Boehringer Ingelheim, Celator, Celgene, Genoptix, Janssen, Juno, MEI Pharma, MedImmune, Novartis, Onconova, Pfizer, Roche/Genentech, Sunesis, Teva: Consultancy; Cellectis: Research Funding.
- Published
- 2016
- Full Text
- View/download PDF
38. Abstract 1232: A-type proanthocyanidins selectively target acute myeloid leukemia cells in vitro and in vivo
- Author
-
Sabrina Martinez, Monica L. Guzman, Bernardo Gomel, Luis A. Lara-Martinez, Hongliang Zong, Stefano Rivella, Burak Isal, Catherine C. Neto, and Laura M. Bystrom
- Subjects
Cancer Research ,business.industry ,CD33 ,Myeloid leukemia ,Pharmacology ,medicine.disease ,medicine.disease_cause ,In vitro ,Myelogenous ,Leukemia ,Oncology ,In vivo ,Cytarabine ,medicine ,business ,Oxidative stress ,medicine.drug - Abstract
Acute myelogenous leukemia (AML) is often a fatal disease where after strong induction therapy most patients relapse and die. A-type proanthocyanidins (A-PACs) are a unique class of compounds found in cranberries (Vaccinium macrocarpon Ait.) that we have found to be effective against several leukemia cell lines and primary AML samples in vitro. Moreover, A-PACs possess a unique ether bond and have ortho-hydroxyl phenolic groups that have the potential to bind to iron, alter redox status, and other biological effects. We found that pre-treatment with antioxidants or holo-transferrin (iron-saturated transferrin) partially protected AML cells from A-PAC induced cell death (p AML-PDX mice (n = 15), were treated for 2.5 weeks via intraperitoneal injections of A-PACs (25 or 50 mg/kg dose every 3 days) or a vehicle control (PBS every 3 days). Mice were sacrificed and leukemia engraftment was evaluated using anti-human CD45 and CD33. Moreover, primary cells treated with A-PACs were assessed for effects on iron metabolism, oxidative stress, cytokine response, and survival pathways by gene expression analysis or flow cytometry. Administration of A-PACs to AML-PDX tumors reduced tumor burden. Mice that were treated with the vehicle control had engraftment of AML primary cells equivalent to 12.51% (95% CI: 4.9, 20.11; n = 5), whereas the mice treated with the 50 mg/kg and 25 mg/kg A-PACs showed a level of engraftment of 5.2% (95% CI: 1.5, 8.9; n = 5) and 5.4% (95% CI: 2.3, 8.5; n = 5), respectively. These results indicated more than a 50% reduction in engraftment, which was better or equal to the effects we observed in mice treated with high-dose cytarabine, a standard care drug. Moreover, no toxic effects were observed in the mice. It was also found that both cells and mice treated with A-PACs lead to the production of specific subset of cytokines. Global gene expression data showed consistent upregulation of some of these cytokines, and also upregulation of NF-κB and enzymes indicative of oxidative stress. The results indicate that A-PACs not only target primary AML cells in vitro, but are also effective in vivo by a potentially novel mechanism. Further elucidation of this mechanism may uncover new vulnerabilities of this disease. Citation Format: Laura M. Bystrom, Luis Andres Lara-Martinez, Bernardo Gomel, Burak Isal, Hongliang Zong, Sabrina Martinez, Catherine Neto, Stefano Rivella, Monica L. Guzman. A-type proanthocyanidins selectively target acute myeloid leukemia cells in vitro and in vivo. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1232.
- Published
- 2016
- Full Text
- View/download PDF
39. Abstract 3992: Allogeneic TCRαβ-deficient CAR T-cells targeting CD123 effectively eliminate myeloid leukemia cells in vitro and in vivo PDX mice
- Author
-
Gail J. Roboz, Hongliang Zong, Céline Lebuhotel, Roman Galetto, Agnès Gouble, Alexander Bank, Monica L. Guzman, Mayumi Sugita, Nicole M. Cruz, Laura M. Bystrom, Julianne Smith, and Luis A. Lara-Martinez
- Subjects
Cancer Research ,business.industry ,CD34 ,Myeloid leukemia ,medicine.disease ,Haematopoiesis ,Leukemia ,Oncology ,B-cell leukemia ,Immunology ,Cancer research ,medicine ,Cytotoxic T cell ,Progenitor cell ,Stem cell ,business - Abstract
Acute myeloid leukemia (AML) is incurable in the majority of patients. While allogeneic stem cell transplantation remains the most effective therapy for AML to date, other types of cellular therapy have not yet been successful in this disease. The success of autologous T-cells expressing chimeric antigen receptors (CARs) in patients with advanced B cell leukemia and lymphomas has encouraged the investigation of CAR technology for the treatment of AML by targeting distinct tumor-specific antigens. We have developed an allogeneic CAR-T cell platform using T-cells from third-party healthy donors to generate T-cells targeting CD123, the transmembrane alpha chain of the interleukin-3 receptor, which is expressed on blasts, leukemic progenitor and leukemic stem cells from the majority of patients with acute myeloid leukemia (AML). Transcription Activator-Like Effector Nuclease (TALEN) gene-editing technology was used to inactivate the TCRα constant (TRAC) gene, eliminating the potential for engineered T-cells to cause graft versus host disease (GvHD). Using leukemia cell lines in both in vitro and in vivo models, we confirmed that TCR-deficient T-cells expressing an anti-CD123 CAR display significant antitumor activity. We next evaluated the in vitro cytotoxic activity of TCR KO CD123-CAR T-cells (UCART123) in primary AML (n = 6) samples and normal cells (normal bone and umbilical cord blood; n = 3) using T-cell:AML cell ratios of 5:1, 2:1 and 0.5:1; and T-cell:normal cell ratio of 10:1 and 1:1. Degranulation and IFNγ release assays revealed potent activation of UCART123 cells when exposed to CD123 leukemia cells but not to normal hematopoietic cells. Cytotoxicity of UCART123 cells was observed as early as 4 hours upon initiation of the co-cultures. However, at 24 hours, more than 80% of leukemic cells (blasts, progenitors and stem cells) were eliminated at all ratios tested, whereas only approximately 20% CAR-independent cell death was observed with the TCR-deficient T-cells. Normal hematopoietic cells showed an average of 30% cell death when exposed to either UCART123 or TCR-deficient T-cells, demonstrating the selectivity of CD123 CAR-T cells toward leukemic cells. Finally, we evaluated the in vivo activity of the CAR-T cells against established patient derived xenografts (PDX) using AML and normal CB CD34+ cells. We found no significant difference between PBS, TCR-deficient (10e6/mouse) and CAR T-cell treatments (10 e6/mouse) in PDX mice transplanted with normal CD34+ CB. Strikingly, 14 days of treatment eliminated most of the leukemic cells from the AML-PDX mice. T-cells were still detected at day 14 after treatment with UCART123 cells (mean 52% CD123 CAR and mean 1.5% TCR-), without evidence of GVHD. Efforts are underway to develop allogeneic CD123 CAR-T cells for clinical trials in AML. Citation Format: Monica L. Guzman, Hongliang Zong, Mayumi Sugita, Luis A. Lara-Martinez, Laura M. Bystrom, Nicole M. Cruz, Roman Galetto, Agnès Gouble, Céline Lebuhotel, Alexander Bank, Julianne Smith, Gail J. Roboz. Allogeneic TCRαβ-deficient CAR T-cells targeting CD123 effectively eliminate myeloid leukemia cells in vitro and in vivo PDX mice. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3992.
- Published
- 2016
- Full Text
- View/download PDF
40. Abstract 3834: scFv antibodies against EphA2 receptor as a therapeutic strategy for AML
- Author
-
Mayumi Sugita, Hongliang Zong, Monica L. Guzman, Juha P. Himanen, and Michael Albert
- Subjects
Cancer Research ,education.field_of_study ,biology ,medicine.drug_class ,Population ,Erythropoietin-producing hepatocellular (Eph) receptor ,Monoclonal antibody ,EPH receptor A2 ,medicine.disease ,Raji cell ,Leukemia ,Oncology ,Cell culture ,biology.protein ,medicine ,Cancer research ,Antibody ,education - Abstract
The ephrin (Eph) receptors have been sought as therapeutic targets in liquid and solid tumors. Eph receptors are a subcategory of receptor tyrosine kinases and play vital parts in cell survival and function. Recently, monoclonal antibodies have been designed to bind to the ligand-binding cavity, that block ephrin to bind to ephrin type-A receptor 2 (EphA2). The potential clinical importance has been demonstrated in lymphomas, and the ability of the antibody to reduce proliferation and induce apoptosis (Goldgur et. al 2014). Acute myelogenous leukemia (AML) is a disease with dismal outcomes and new therapeutic targets have been explored. EphA2 has been described to be critical for the survival in EphA2-positive mouse leukemia models and not essential for normal hematopoiesis. Thus, we examined cell surface expression of EphA2 in peripheral blood, and bone marrow samples from AML patients. We screened 30 primary AML samples and 5 normal samples and assessed their expression of EphA2 using flow cytometry using the lymphoma cell line (Raji) as a positive control, as reported to express EphA2. The EphA2 expression was evaluated under culture conditions as well as without culture. Prior to culture, 22 (73.3%) of 30 samples were EphA2 positive in more than 90% of blasts. The mean cell surface expression of EphA2 in blasts was 95.4% (range: 64.4-99.7%) with a mean fluorescence intensity (MFI) of 201.5 (range: 18.4-675.0) with a MFI of 4.4 for isotype control. Interestingly, we observed 13.3% increase in EphA2 expression post-culture, bringing the proportion of EphA2-high samples from 73.3% to 86.6% of total samples. In addition to the increase of EphA2-high population, the average MFI of this cohort was also increased by 47.6% post-culture relative to pre-culture. Furthermore, we saw a more significant increase in the CD34+ population and little or no change in the CD34- population. These data shows that many AML express EphA2, and that culturing with cytokines can induce robust EphA2 expression. After validating EphA2 expression in leukemia cells, we have tested a set of single-chain antibodies (scFv) against EphA2 that have shown activity against Raji cells (Goldgur et. al 2014). We tested two single-chain variable fragments (G2 or D2) on samples with low or high expression of EphA2. The samples were incubated in the serum free IMDM medium supplemented with cytokines. The scFv antibodies were tested at 0.5ug and 1.0ug and evaluated after 24hr- or 72hr-culture. We found that exposure to D2 resulted in a significant decrease in cell number after 72 hr-culture, 52.6% and 99.3% relative to vehicle control treatment for 0.5ug and 1.0ug respectively in EphA2-high AML cells. These data suggests that the scFv antibodies against EphA2 are lethal to AML cells that express high level of EphA2. Taken together, our data suggests that more than 70% of AML cells highly express EphA2 and can be targeted using scFv antibodies specific to EphA2. Citation Format: Michael Albert, Mayumi Sugita, Hongliang Zong, Juha Himanen, Monica L. Guzman. scFv antibodies against EphA2 receptor as a therapeutic strategy for AML. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3834.
- Published
- 2016
- Full Text
- View/download PDF
41. Design, synthesis, and evaluation of small molecule Hsp90 probes
- Author
-
Hongliang Zong, James H. Ahn, Monica L. Guzman, Pallav D. Patel, Anna Rodina, Danuta Zatorska, Kamalika Moulick, Tony Taldone, and Gabriela Chiosis
- Subjects
Molecular model ,Clinical Biochemistry ,Pharmaceutical Science ,Antineoplastic Agents ,Biochemistry ,Chemical synthesis ,Binding, Competitive ,Article ,Hsp90 inhibitor ,chemistry.chemical_compound ,Drug Discovery ,polycyclic compounds ,Humans ,HSP90 Heat-Shock Proteins ,Molecular Biology ,Natural product ,biology ,Organic Chemistry ,Geldanamycin ,Hsp90 ,Small molecule ,chemistry ,Docking (molecular) ,Drug Design ,Molecular Probes ,biology.protein ,Molecular Medicine - Abstract
A number of compounds from different chemical classes are known to bind competitively to the ATP-pocket of Hsp90 and inhibit its chaperone function. The natural product geldanamycin was the first reported inhibitor of Hsp90 and since then synthetic inhibitors from purine, isoxazole and indazol-4-one chemical classes have been discovered and are currently or soon to be in clinical trials for the treatment of cancer. In spite of a similar binding mode to Hsp90, distinct biological profiles were demonstrated amongst these molecules, both in vitro and in vivo. To better understand the molecular basis for these dissimilarities, we report here the synthesis of chemical tools for three Hsp90 inhibitor classes. These agents will be useful for probing tumor-by-tumor the Hsp90 complexes isolated by specific inhibitors. Such information will lead to better understanding of tumor specific molecular markers to aid in their clinical development. It will also help to elucidate the molecular basis for the biological differences observed among Hsp90 inhibitors.
- Published
- 2010
42. Homodimerization is essential for the receptor for advanced glycation end products (RAGE)-mediated signal transduction
- Author
-
Angelina Madden, Hongliang Zong, Alan W. Stitt, Mark Mooney, Micheal S. Ward, and Christopher T. Elliott
- Subjects
MAPK/ERK pathway ,Cell signaling ,endocrine system diseases ,MAP Kinase Signaling System ,Receptor for Advanced Glycation End Products ,Ligands ,Biochemistry ,Protein–protein interaction ,RAGE (receptor) ,Cell Line ,Glycation ,Humans ,cardiovascular diseases ,Receptors, Immunologic ,Receptor ,Protein Structure, Quaternary ,Molecular Biology ,Chemistry ,Cell Membrane ,NF-kappa B ,nutritional and metabolic diseases ,Cell Biology ,Peptide Fragments ,Cell biology ,Protein Structure, Tertiary ,Solubility ,cardiovascular system ,Phosphorylation ,Signal transduction ,Protein Multimerization ,human activities ,Signal Transduction - Abstract
The receptor for advanced glycation end products (RAGE) is a pattern-recognition receptor that binds to diverse ligands and initiates a downstream proinflammatory signaling cascade. RAGE activation has been linked to diabetic complications, Alzheimer disease, infections, and cancers. RAGE is known to mediate cell signaling and downstream proinflammatory gene transcription activation, although the precise mechanism surrounding receptor-ligand interactions is still being elucidated. Recent fluorescence resonance energy transfer evidence indicates that RAGE may form oligomers on the cell surface and that this could be related to signal transduction. To investigate whether RAGE forms oligomers, protein-protein interaction assays were carried out. Here, we demonstrate the interaction between RAGE molecules via their N-terminal V domain, which is an important region involved in ligand recognition. By protein cross-linking using water-soluble and membrane-impermeable cross-linker bis(sulfosuccinimidyl) suberate and nondenaturing gels, we show that RAGE forms homodimers at the plasma membrane, a process potentiated by S100B and advanced glycation end products. Soluble RAGE, the RAGE inhibitor, is also capable of binding to RAGE, similar to V peptide, as shown by surface plasmon resonance. Incubation of cells with soluble RAGE or RAGE V domain peptide inhibits RAGE dimerization, subsequent phosphorylation of intracellular MAPK proteins, and activation of NF-kappaB pathways. Thus, the data indicate that dimerization of RAGE represents an important component of RAGE-mediated cell signaling.
- Published
- 2010
43. Trihydrophobin 1 attenuates androgen signal transduction through promoting androgen receptor degradation
- Author
-
Jianhui Xie, Yayun Chi, Xiangfei Kong, Weiying Zou, Yanlin Wang, Jianxin Gu, Hongliang Zong, Jianhai Jiang, Yi Hong, Hanzhou Wang, and Yanzhong Yang
- Subjects
Transcriptional Activation ,medicine.medical_specialty ,medicine.drug_class ,Regulator ,Down-Regulation ,Biology ,urologic and male genital diseases ,Biochemistry ,Prostate cancer ,Mice ,Prostate ,Internal medicine ,Cell Line, Tumor ,LNCaP ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,RNA, Messenger ,Receptor ,Promoter Regions, Genetic ,Molecular Biology ,Cell Nucleus ,Protein Stability ,Gene Expression Profiling ,Ubiquitination ,Cell Biology ,Prostate-Specific Antigen ,Androgen ,medicine.disease ,Cell biology ,Androgen receptor ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Endocrinology ,medicine.anatomical_structure ,Enhancer Elements, Genetic ,Receptors, Androgen ,COS Cells ,Androgens ,Signal transduction ,Carrier Proteins ,Protein Processing, Post-Translational ,Protein Binding ,Signal Transduction ,Transcription Factors - Abstract
The androgen-signaling pathway plays critical roles in normal prostate development, benign prostatic hyperplasia, established prostate cancer, and in prostate carcinogenesis. In this study, we report that trihydrophobin 1 (TH1) is a potent negative regulator to attenuate the androgen signal-transduction cascade through promoting androgen receptor (AR) degradation. TH1 interacts with AR both in vitro and in vivo, decreases the stability of AR, and promotes AR ubiquitination in a ligand-independent manner. TH1 also associates with AR at the active androgen-responsive prostate-specific antigen (PSA) promoter in the nucleus of LNCaP cells. Decrease of endogenous AR protein by TH1 interferes with androgen-induced luciferase reporter expression and reduces endogenous PSA expression. Taken together, these results indicate that TH1 is a novel regulator to control the duration and magnitude of androgen signal transduction and might be directly involved in androgen-related developmental, physiological, and pathological processes.
- Published
- 2010
44. CDK11p58 represses vitamin D receptor-mediated transcriptional activation through promoting its ubiquitin-proteasome degradation
- Author
-
Jianxin Gu, Yi Hong, Junwu Yang, Hongliang Zong, Weiying Zou, Yanlin Wang, Xiaojing Yun, Yayun Chi, and Xiangfei Kong
- Subjects
musculoskeletal diseases ,Transcriptional Activation ,Proteasome Endopeptidase Complex ,Biophysics ,Transfection ,Biochemistry ,Calcitriol receptor ,Ubiquitin ,Transcription (biology) ,Cyclins ,Chlorocebus aethiops ,polycyclic compounds ,Animals ,Humans ,Cyclin D3 ,Receptor ,Molecular Biology ,Regulation of gene expression ,biology ,Kinase ,digestive, oral, and skin physiology ,Ubiquitination ,Cell Biology ,Cell cycle ,Molecular biology ,Cell biology ,Nuclear receptor ,COS Cells ,biology.protein ,Receptors, Calcitriol ,lipids (amino acids, peptides, and proteins) - Abstract
Vitamin D receptor (VDR) is a member of the nuclear receptor superfamily and regulates transcription of target genes. In this study, we identified CDK11p58 as a novel protein involved in the regulation of VDR. CDK11p58, a member of the large family of p34cdc2-related kinases, is associated with cell cycle progression, tumorigenesis, and apoptotic signaling. Our study demonstrated that CDK11p58 interacted with VDR and repressed VDR-dependent transcriptional activation. Furthermore, overexpression of CDK11p58 decreased the stability of VDR through promoting its ubiquitin–proteasome-mediated degradation. Taken together, these results suggest that CDK11p58 is involved in the negative regulation of VDR.
- Published
- 2009
45. Knockdown of BCL2L12 leads to cisplatin resistance in MDA-MB-231 breast cancer cells
- Author
-
Yuanyan Wei, Weibing Wu, Junwu Yang, Hongliang Zong, Xiangfei Kong, Jianxing Gu, Wenzong Wang, Yanlin Wang, Yi Hong, Si Zhang, and Xiaojing Yun
- Subjects
β-Catenin ,MDA-MB-231 ,Muscle Proteins ,Antineoplastic Agents ,Apoptosis ,Breast Neoplasms ,Breast cancer ,Downregulation and upregulation ,Cell Line, Tumor ,medicine ,Humans ,skin and connective tissue diseases ,Molecular Biology ,Cell Proliferation ,Cisplatin ,Gene knockdown ,Cell growth ,Chemistry ,Cell Cycle ,medicine.disease ,MCF-7 ,Proto-Oncogene Proteins c-bcl-2 ,Cell culture ,Drug Resistance, Neoplasm ,BCL2L12 ,Immunology ,Cancer research ,Molecular Medicine ,Female ,medicine.drug - Abstract
BCL2L12, a newly identified member of Bcl-2 family, contains a BH2 domain and a putative BH3 domain. It was found to be highly expressed in normal breast tissues, and was associated with favorable prognosis in breast cancer patients. Here, we reported that the mRNA levels of BCL2L12 and its transcript variant BCL2L12A could be upregulated upon cisplatin treatment in MDA-MB-231 breast cancer cells. Knockdown of BCL2L12 and BCL2L12A dramatically inhibited cisplatin-induced apoptosis. In contrast, ectopic expressions of each of the proteins promoted cisplatin-induced apoptosis. These results indicated that decreased expressions or loss of BCL2L12 and BCL2L12A may contribute to the cisplatin resistance in breast cancer patients. Furthermore, we found that cisplatin-induced downregulation of β-catenin was partially suppressed in BCL2L12- and BCL2L12A-knocked down MDA-MB-231 cells, which indicated that knockdown of these two proteins may stabilize β-catenin in cisplatin-induced apoptosis. In short, we proposed that BCL2L12 and BCL2L12A may play an important role in cisplatin-induced apoptosis in MDA-MB-231 breast cancer cells.
- Published
- 2008
46. Functional interaction of E1AF and Sp1 in glioma invasion
- Author
-
Jin Zhou, Jianxin Gu, Jianhai Jiang, Yuanyan Wei, Xiaojing Yun, Hongliang Zong, Xiangfei Kong, Jialin Shen, Yanzhong Yang, Dan Liu, Si Zhang, and Xiaoning Chen
- Subjects
Transcriptional Activation ,Sp1 Transcription Factor ,Molecular Sequence Data ,Down-Regulation ,Biology ,Mediator ,Transcription (biology) ,Epidermal growth factor ,Cell Movement ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Humans ,Neoplasm Invasiveness ,Phosphorylation ,Promoter Regions, Genetic ,Molecular Biology ,Transcription factor ,Regulation of gene expression ,Sp1 transcription factor ,Base Sequence ,Epidermal Growth Factor ,Proto-Oncogene Proteins c-ets ,Promoter ,Cell Biology ,Glioma ,Articles ,Gene Expression Regulation, Neoplastic ,Cancer research ,Adenovirus E1A Proteins ,Protein Binding - Abstract
Transcription factor E1AF is widely known to play critical roles in tumor metastasis via directly binding to the promoters of genes involved in tumor migration and invasion. Here, we report for the first time E1AF as a novel binding partner for ubiquitously expressed Sp1 transcription factor. E1AF forms a complex with Sp1, contributes to Sp1 phosphorylation and transcriptional activity, and functions as a mediator between epidermal growth factor and Sp1 phosphorylation and activity. Sp1 functions as a carrier bringing E1AF to the promoter region, thus activating transcription of glioma-related gene for beta1,4-galactosyltransferase V (GalT V; EC 2.4.1.38). Biologically, E1AF functions as a positive invasion regulator in glioma in cooperation with Sp1 partly via up-regulation of GalT V. This report describes a new mechanism of glioma invasion involving a cooperative effort between E1AF and Sp1 transcription factors.
- Published
- 2007
47. Cyclin D3/CDK11p58 complex is involved in the repression of androgen receptor
- Author
-
Jianhai Jiang, Yanlin Wang, Xiaojing Yun, Zejuan Li, Hanzhou Wang, Yanzhong Yang, Li Zhang, Haijiao Chen, Hongliang Zong, Yi Hong, Jianxin Gu, and Yayun Chi
- Subjects
Male ,Transcription, Genetic ,Cyclin D ,Cyclin A ,Cyclin B ,Cell Line ,Mice ,Cyclin D1 ,Cyclin-dependent kinase ,Genes, Reporter ,Cyclins ,Testis ,Serine ,Animals ,Humans ,Protein Isoforms ,Cyclin D3 ,Phosphorylation ,Molecular Biology ,Cell Proliferation ,biology ,Cell Biology ,Articles ,Prostate-Specific Antigen ,Cyclin-Dependent Kinases ,Cell biology ,Protein Structure, Tertiary ,Gene Expression Regulation ,Receptors, Androgen ,Multiprotein Complexes ,biology.protein ,Cyclin-dependent kinase complex ,Cancer research ,RNA Interference ,Cyclin A2 ,Signal Transduction - Abstract
Androgen receptor (AR) is essential for the maintenance of the male reproductive systems and is critical for the carcinogenesis of human prostate cancers (PCas). D-type cyclins are closely related to the repression of AR function. It has been well documented that cyclin D1 inhibits AR function through multiple mechanisms, but the mechanism of how cyclin D3 exerts its repressive role in the AR signaling pathway remains to be identified. In the present investigation, we demonstrate that cyclin D3 and the 58-kDa isoform of cyclin-dependent kinase 11 (CDK11p58) repressed AR transcriptional activity as measured by reporter assays of transformed cells and prostate-specific antigen expression in PCa cells. AR, cyclin D3, and CDK11p58 formed a ternary complex in cells and were colocalized in the luminal epithelial layer of the prostate. AR activity is controlled by phosphorylation at specific sites. We found that AR was phosphorylated at Ser-308 by cyclin D3/CDK11p58 in vitro and in vivo, leading to the repressed activity of AR transcriptional activation unit 1 (TAU1). Furthermore, androgen-dependent proliferation of PCa cells was inhibited by cyclin D3/CDK11p58 through AR repression. These data suggest that cyclin D3/CDK11p58 signaling is involved in the negative regulation of AR function.
- Published
- 2007
48. CDK11(p58) protein kinase activity is associated with Bcl-2 down-regulation in pro-apoptosis pathway
- Author
-
Jianhai Jiang, Xiaojing Yun, Yihong Wu, Jianxin Gu, Zhou Zhang, Luyang Yao, Yi Hong, Junwu Yang, and Hongliang Zong
- Subjects
Clinical Biochemistry ,Down-Regulation ,Apoptosis ,Cycloheximide ,Biology ,Transfection ,chemistry.chemical_compound ,Tumor Cells, Cultured ,Humans ,Kinase activity ,Protein kinase A ,Molecular Biology ,Regulation of gene expression ,Carcinoma ,Liver Neoplasms ,Cell Biology ,General Medicine ,Cyclin-Dependent Kinases ,Cell biology ,Genes, bcl-2 ,Gene Expression Regulation, Neoplastic ,chemistry ,Cancer research ,Phosphorylation ,Ectopic expression ,Signal transduction ,Signal Transduction - Abstract
CDK11(p58), a G2/M-specific protein kinase, has been shown to be associated with apoptosis in many cell lines, with largely unknown mechanisms. Our previous study proved that CDK11(p58)-enhanced cycloheximide (CHX)-induced apoptosis in SMMC-7721 hepatocarcinoma cells. Here we report for the first time that ectopic expression of CDK11(p58) down-regulates Bcl-2 expression and its Ser70, Ser87 phosphorylation in CHX-induced apoptosis in SMMC-7721 cells. Overexpression of Bcl-2 counteracts the pro-apoptotic activity of CDK11(p58). Furthermore, we confirm that the kinase activity of CDK11(p58) is essential to the down-regulation of Bcl-2 as well as apoptosis. Taken together, these results demonstrate that CDK11(p58) down-regulates Bcl-2 in pro-apoptosis pathway depending on its kinase activity, which elicits survival signal in hepatocarcinoma cells.
- Published
- 2007
49. A Hyperactive Signalosome Results in High Sensitivity to HSP90 Inhibitors in AML
- Author
-
Gail J. Roboz, Gabriela Chiosis, Hongliang Zong, Monica L. Guzman, and Tony Taldone
- Subjects
Immunology ,Cell Biology ,Hematology ,Biology ,medicine.disease_cause ,medicine.disease ,Biochemistry ,Hsp90 inhibitor ,Leukemia ,medicine.anatomical_structure ,In vivo ,Genetic model ,medicine ,Cancer research ,Bone marrow ,Signal transduction ,Stem cell ,Carcinogenesis - Abstract
HSP90 is well established in supporting tumorigenesis by stabilizing oncogenic client proteins. Given this crucial role, a number of HSP90 inhibitors have been tested in various types of cancer, including leukemia. However, clinical trials thus far revealed only a subset of AML patients benefited from the treatment. Therefore, precision medicine approaches to define parameters that predict the patients' response to HSP90 inhibitors are needed to select patients who are most likely to benefit. We have previously demonstrated that PU-H71, a novel purine scaffold HSP90 inhibitor with selectivity for a tumor-specific HSP90 and currently translating into Phase 2 clinical evaluation, is capable of ablating malignant blasts, progenitor and stem cells in AML patient samples using in vitro studies. We found that leukemia cell lines (n=18) and primary AML patient samples (n=26) with greater numbers of simultaneously activated signaling networks, including PI3K-AKT and JAK-STAT, were the most sensitive to HSP90 inhibition. Using different genetic models, our studies revealed that diverse oncogenic transformations that converge upon simultaneous hyperactivation of PI3K-AKT and JAK-STAT promote sensitivity to PU-H71. To validate the efficacy of PU-H71 in vivo, we generated AML-GFP-luciferase xenograft models using cell lines with hyperactive signalosome. Xenotransplanted mice were treated with PU-H71 one week post-engraftment. In vivo imaging indicated that MOLM-13 xenografted leukemia was rapidly and significantly reduced by PU-H71 treatment. Six doses of PU-H71 produced robust anti-leukemic activity as indicated by in vivo imaging and flow cytometric analysis of post-treatment bone marrow (no disease detected). In addition, we generated 7 AML patient-derived xenografts (PDX) cohorts with samples that displayed varied levels of activation of PI3K-AKT and JAK-STAT signaling pathways. After initial validation that status of the PI3K-AKT and JAK-STAT signaling pathways were preserved in the PDX, we initiated treatment with PU-H71 and found that, as predicted, the AML-PDX with the most hyperactive signalosome were the most sensitive to in vivo treatment to PU-H71. Importantly, samples with hyperactive PI3K-AKT and JAK-STAT signaling also demonstrated a significant reduction in LSC using secondary transplants. Taken together, we found that a hyperactive signalosome results in increased sensitivity to the HSP90 inhibitor PU-H71 in vitro and in vivo. Our study suggests that evaluation of PI3K-AKT and JAK-STAT signaling pathways may provide a means to select patients who are most likely to benefit from HSP90 inhibitory therapy. Disclosures No relevant conflicts of interest to declare.
- Published
- 2015
- Full Text
- View/download PDF
50. Abstract 1667: Targeting acute myelogenous leukemia with novel combrestatin analogs and development of predictors of response
- Author
-
Paraskevi Giannakakou, Peter A. Crooks, Monica L. Guzman, Hongliang Zong, Vijayakumar N. Sonar, Narsimha Reddy Penthala, and Gail J. Roboz
- Subjects
Combretastatin ,Cancer Research ,Cell cycle checkpoint ,medicine.diagnostic_test ,Chemistry ,Cell cycle ,medicine.disease ,Flow cytometry ,Leukemia ,chemistry.chemical_compound ,Oncology ,Cell culture ,hemic and lymphatic diseases ,medicine ,Cancer research ,Viability assay ,Stem cell - Abstract
Introduction: Most adult acute myelogenous leukemia (AML) patients relapse and die of the disease. AML stem cells (AML-SCs) have been postulated to resist standard chemotherapeutic agents and thus give rise to relapse. Combretastatin A-4 (CA-4), a natural product isolated from the South African tree Combretum caffrum, has been reported to possess anti-leukemic activity in AML cell lines. To increase the repertoire of combretastatin analogs with potent anti-leukemic activity, about 100 CA-4 analogs were tested in AML cells, and studied the mechanism that AML cells resist to such microtubule (MT) inhibitors. Methods: Cell viability/apoptosis was determined by annexin V and 7-AAD staining and flow cytometry. Cellular ROS production was measured by CellROX. Cell cycle was determined by DAPI staining and flow cytometry. Stem cell function was evaluated by colony assay and mouse xenograft assays. Tubulin depolymerization was measured by immunofluorescence or immunoblotting for microtubule fraction. Interaction of tubulin with BTAN or p38 MAPK was determined by pull down assays. Results: One of the most potent analogs, BTAN, was further investigated in a panel of leukemic cell lines and primary AML samples. Similar to CA-4, BTAN inhibited tubulin polymerization in vitro and induced MT depolymerization and cell cycle arrest at G2/M in AML cells. BTAN induced cell death of AML blast, progenitor and stem cells via caspase activation regardless of their proliferating status and level of intracellular reactive oxygen species (ROS). In addition, BTAN impaired the physical contact of AML with stromal cells. To further identify the mechanism of resistance to BTAN, we investigated several signal transduction pathways including AKT, JAK-STAT, ERK1/2, JNK and p38. p38 MAPK inhibitor sensitized leukemia cells to BTAN treatment, and the sensitivity of AML cells to BTAN correlated with the basal level of active/phosphorylated p38 MAPK, suggesting p38 MAPK may interfere with the sensitivity to BTAN in AML. We demonstrated that tubulin interacted with p38 MAPK, especially its phosphorylated form, which was disrupted by treatment with a p38 MAPK inhibitor. In addition, in the presence of p38 inhibitor, the accumulation of BTAN was potentiated in AML cells, suggesting activated p38 may interfere with the interaction between BTAN and its tubulin targets. Conclusion: Our data demonstrated that BTAN is a newly identified anti-AML-SC agent that works by disrupting the MT cytoskeleton and inducing caspase activation. These data suggest that the MT network is involved in the regulation of pro-survival signaling pathways even in quiescent AML-SCs, and that p38 MAPK is a key modulator for the response of AML blasts to microtubule inhibitors and that co-treatment with p38 MAPK inhibitors could be beneficial to microtubule destabilizing agents in AML. Note: This abstract was not presented at the meeting. Citation Format: Hongliang Zong, Narsimha R. Penthala, Vijayakumar N. Sonar, Paraskevi Giannakakou, Gail J. Roboz, Peter A. Crooks, Monica L. Guzman. Targeting acute myelogenous leukemia with novel combrestatin analogs and development of predictors of response. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1667. doi:10.1158/1538-7445.AM2015-1667
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.