35 results on '"Zhuoxiao Cao"'
Search Results
2. Abstract 4077: Dual-targeted CAR-NK cell therapy: optimized CAR design to prevent antigen escape and elicit a deep and durable response in multiple myeloma
- Author
-
Cuiqing Yang, Yifang Wang, Tingting Liu, Chao Wang, Huanyu Wang, Qingyang Wang, Qin Wang, Gang Ye, Renhong Tang, and Zhuoxiao Cao
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Multiple myeloma (MM) is an incurable hematologic malignancy and new strategies that offer a chance of obtaining long-term progression free survival are urgently needed. MM is associated with profound immune alterations and dysfunction of natural killer (NK) cells have been demonstrated to be crucial factors in MM progression. Myeloma cells are susceptible to killing by natural killer (NK) cells but acquire the ability to elude NK cell surveillance by avoiding recognition and suppressing NK cell function. Given the role of NK cells in the pathogenesis of MM, interest in harnessing NK cells to treat the cancer has been energized by the remarkable success of other adoptive cell therapies such as chimeric antigen receptor (CAR)-T cells. However, relapses associated with residual low-to-negative BCMA-expressing MM cells have been reported, necessitating the identification of additional targets. GPRC5D, expressed on MM cells from primary marrow samples with a distribution independent of BCMA. Methods: To prevent BCMA-antigen escape and elicit a deeper and more durable response in MM, we developed a new multiplexed edited NK cell configuration to restore anti-myeloma NK cell immunity, consisting of anti-BCMA VHH and anti-GPRC5D VHH antibodies, NKG2D and 2B4 co-stimulation signaling domains, and IL-15. Results: Dual targeting BCMA/GPRC5D CAR-NK showed potent in vitro killing of both BCMA+ and GPRC5D+ myeloma cells. Utilizing a repeated rounds of cancer cell clearance assay, BCMA/GPRC5D CAR NK cells showed remarkable persistence and antigen-mediated expansion of CAR-NK cells after more than 4 rounds of tumor cell re-challenges. Moreover, in comparison with single targeted BCMA CAR-NK cells, Dual targeting BCMA/GPRC5D CAR-NK cells effectively lysed BCMA negative target cells. In addition, in BCMA-antigenic escape model, it achieved more sustained tumor control than single targeting BCMA CAR-NK cells. PD-1/PD-L1 axis inhibition has been reported to enhance NK cell activity against MM cells by augmenting NK cell trafficking, immune complex formation, and cytotoxicity against PD-L1-expressing MM cells. We also demonstrated that combination of BCMA/GPRC5D CAR-NK with anti-PDL1-IL15 also showed more persistent tumor control. Conclusions: Our studies demonstrate the development of dual targeting BCMA/GPRC5D CAR NK cells may represent a highly effective off-the-shelf therapeutic product as a monotherapy or in combinations with other immune-regulating agents. Citation Format: Cuiqing Yang, Yifang Wang, Tingting Liu, Chao Wang, Huanyu Wang, Qingyang Wang, Qin Wang, Gang Ye, Renhong Tang, Zhuoxiao Cao. Dual-targeted CAR-NK cell therapy: optimized CAR design to prevent antigen escape and elicit a deep and durable response in multiple myeloma. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4077.
- Published
- 2023
- Full Text
- View/download PDF
3. Abstract 4087: Co-expression of membrane bound IL-15 enhanced anti-tumor response of CAR-T
- Author
-
Cuiqing Yang, Fuwei Jiang, Yifang Wang, Tingting Liu, Chao Wang, Qingyang Wang, Qin Wang, Gang Ye, Renhong Tang, and Zhuoxiao Cao
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Chimeric antigen receptors (CARs) T cells have been used successfully to treat patients with hematologic malignancies, but showed less effective in solid tumors. We investigated multiple approaches to engineer and enhance CAR-T activity in solid tumors. It has been previously reported that improvement in the quality of CAR-T cells, through CAR design or manufacturing optimization, could enhance the therapeutic potential of CAR-T cells. One parameter influencing the effectiveness of CAR-T cell therapy is the differentiation status of the final product: CAR-T cells that are less-differentiated and less exhausted are more therapeutically potent. It is well known that IL-2 is the main cytokine used to culture cells for adoptive cell therapy, as it plays an important role in the proliferation and functional effect of T cells. However, it has been noted T cells cultured with IL-2 are phenotypically heterogeneous, being predominantly composed of effector memory cells. In comparison with IL2, IL-15 can induce a memory stem-like T cell phenotype, which is less differentiated and with a superior capacity for cell expansion and survival. Several groups reported that compared with cells cultured with IL-2, CAR-T cells expanded with IL-15 preserve a less-differentiated stem cell memory (Tscm) phenotype and exhibited reduced expression of exhaustion markers, higher antiapoptotic properties, increased proliferative capacity upon antigen challenge, and promoted superior anti-tumor responses in vivo. Method: To provide localized IL-15 mediated signaling to T cells, we constructed a version of IL-15 as a membrane-bound molecule (mIL-15) designed to stimulate T cells in cis and in trans, and mIL-15 is tethered to the cell surface and functions locally to enhance the functionality of the CAR-T cell without systemic delivery of IL-15. Result: Expression of mbIL-15 is shown to enhance T cell expansion, preserve a less-differentiated Tscm phenotype and prevent CAR-T cell exhaustion, leading to longer persistence and an enduring anti-tumor response over the conventional CAR-T cells. Conclusion: We demonstrate that CAR-T activity and persistence can be enhanced by simultaneous expression of mIL-15, which preserves the CAR-T cell Tscm phenotype and improves their metabolic fitness, resulting in superior antitumor activity. These preclinical data support the notion that CAR-T cells are designed and optimized to persist in the hostile tumor microenvironment and potentially improve efficacy against solid tumors. Citation Format: Cuiqing Yang, Fuwei Jiang, Yifang Wang, Tingting Liu, Chao Wang, Qingyang Wang, Qin Wang, Gang Ye, Renhong Tang, Zhuoxiao Cao. Co-expression of membrane bound IL-15 enhanced anti-tumor response of CAR-T. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4087.
- Published
- 2023
- Full Text
- View/download PDF
4. Allogeneic CAR-NK Cell Therapy Targeting Both BCMA and GPRC5D for the Treatment of Multiple Myeloma
- Author
-
Zhuoxiao Cao, Cuiqing Yang, Yifang Wang, Chao Wang, Qingyang Wang, Gang Ye, Tingting Liu, Qin Wang, Huanyu Wang, Yanxue Gong, Biye Wang, and Fuwei Jiang
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
5. Abstract 5203: Discovery and development of SLD2435, a novel humanized IgG1 bispecific antibody simultaneously targeting human TIGIT and PVRIG for cancer immunotherapy
- Author
-
Xiaofeng Zhao, Yayuan Fu, Xiaoqing Liu, Fengli Shan, Zhenna Gao, Mengyu Wang, Huiming Xu, Hai Huang, Yang Liu, Wenqing Yang, Zhuoxiao Cao, and Renhong Tang
- Subjects
Cancer Research ,Oncology - Abstract
Despite dramatic success for immune checkpoint inhibition in treating certain patient subsets, there remains high unmet medical need for the treatment of patients who are refractory to or relapse following treatment with current checkpoint inhibitors. TIGIT and PVRIG, are exciting novel targets for immune-oncology which function as immune inhibitory receptors expressed on both T and NK cells and have been reported to inhibit anti-tumor activity. SLD2435, a novel humanized IgG1 bispecific antibody simultaneously targeting human TIGIT and PVRIG that synergistically enhances both NK and T cell functions, demonstrates significant preclinical anti-tumor efficacy. Here, we reported the discovery, development and characterization of SLD2435 for potential use in cancer patients. Anti-human TIGIT mAb was generated from mouse hybridoma whereas anti-human PVRIG Ab was discovered from Alpaca as single-domain antibody (sdAb). Both Abs were identified based on the ability to block the interaction of TIGIT or PVRIG with their cognate ligands (PVR or PVRL2 respectively) and for the ability to enhance primary human NK cytotoxicity over tumor cell lines as well as to promote the function of antigen-specific CD8+ T cells. Lead anti-human PVRIG sdAb was then selected and fused to the N-terminal of lead anti-human TIGIT mAb with a peptide linker for SLD2435 construction followed by subsequent testing in multiple abovementioned assays. SLD2435 simultaneously bound to human TIGIT and PVRIG with sub-nanomolar affinity and showed cross-reactivity to respective cynomolgus targets together with strong avidity mediated receptor-ligand interaction blocking at both protein and cellular levels. In vitro functional studies revealed that SLD2435 induced potent primary human NK cell mediated cytotoxicity over WiDr tumor cells better than the combination of anti-PVRIG and anti-TIGIT mAbs. It could also trigger preferential depletion of TIGIT+/TIGIT+PVRIG+ Treg via ADCC. In addition, SLD2435 enhanced IFN-γ production of CMV antigen-specific CD8+ T cells culturing with PVR+PVRL2+HLA matched Colo205 tumor cells and had synergistic effect on T cell activation when further combined with an anti-PD-L1 mAb. In consistent with in vitro results, SLD2435 displayed significant anti-tumor efficacy in vivo in a HuPBMC A375 xenograft model. At last, SLD2435 demonstrated great developability profile under various stress tests. Taken together, these data support the further development of SLD2435 as a potential novel therapeutic agent for the treatment of cancer patients. Citation Format: Xiaofeng Zhao, Yayuan Fu, Xiaoqing Liu, Fengli Shan, Zhenna Gao, Mengyu Wang, Huiming Xu, Hai Huang, Yang Liu, Wenqing Yang, Zhuoxiao Cao, Renhong Tang. Discovery and development of SLD2435, a novel humanized IgG1 bispecific antibody simultaneously targeting human TIGIT and PVRIG for cancer immunotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5203.
- Published
- 2022
- Full Text
- View/download PDF
6. Abstract 6343: A multispecific T cell engager binding to both membrane proximal and membrane distal epitopes of MSLN with low affinity CD3 for the treatment of AML and solid tumors
- Author
-
Cuiqing Yang, Xiaohui Shao, Feng Zhou, Lei Zhou, Qi Deng, Yun Zhang, Guangcun Cheng, Yingying Hu, Huaiyuan Ma, Yadan Wu, Shuai Wang, Jie Zang, Lei Liu, Wenqing Yang, Yang Liu, Chunlei Xia, Jianzhong Hu, Ande Luo, Yayuan Fu, Zhuoxiao Cao, and Renhong Tang
- Subjects
Cancer Research ,Oncology - Abstract
Purpose: The effective treatment of AML and solid tumors remains an unmet medical need. Mesothelin (MSLN) is a glycophosphatidylinositol-linked tumor antigen overexpressed in a variety of malignancies, including AML and solid tumors, such as mesothelioma, cholangiocarcinoma, ovarian, pancreatic, lung, esophageal, gastric, colorectal, endometrial and triple-negative breast cancer. Early signs of clinical efficacy with MSLN-targeting agents have validated MSLN as a promising target for therapeutic intervention, but therapies with improved efficacy are still needed. CD3-based T cell engagers are highly potent therapeutic molecules with T cell cytotoxicity activities in the picomolar range. But high CRS risk and T cell exhaustion has been observed for high affinity CD3-based T cell engagers in clinicals, so we used high affinity MSLN and low affinity CD3 to improve efficacy of T cell engager. Experimental Design: We designed SMTE-001, a 78-kDa, tetra-specific, T-cell-activating protein molecule, which binds to two epitopes (membrane proximal and membrane distal) of MSLN, CD3ε on T cells, and to serum albumin. Experiments were conducted to assess the potency, activity, and half-life of HPN536 in in vitro assays, rodent models, and in nonhuman primates (NHP). This molecule was tested in a cytotoxicity assay using human PBMCs co-cultured with OVCAR3, Hela and NCI-H292 cells, which express different levels of MSLN. Soluble MSLN and CA125 (ligand of MSLN) was added to determine effects on cytotoxicity. In vivo xenograft mouse studies were conducted using a PBMC model. Results: Here we report the design and the promising preclinical activity of SMTE-001 molecule in vitro and in vivo. We demonstrate that the bivalent MSLN T cell engager has increased in vitro potency in T cell activation and tumor cell killing, as compared to a monovalent counterpart on high MSLN expressing cells. We also demonstrate that T cell exhaustion is reduced for the low-affinity CD3, compared to the low-affinity CD3 molecule. Because soluble MSLN and CA125 is shed from cancer cells into cancer patient serum, we also demonstrate that soluble MSLN and CA125 does not interfere with the cytotoxic activity of SMTE-001. Importantly, we demonstrate in vivo that SMTE-001 significantly inhibits tumor growth in a dose-dependent manner, while high-affinity CD3 molecule’ efficacy reduces when higher dosing. In cynomolgus monkeys, SMTE-001 shows pharmacokinetics in support of weekly dosing in humans. Conclusions: Collectively, these data demonstrate strong anti-tumor efficacy by this novel multispecific bivalent T cell engager. These data indicate the therapeutic potential of this molecule to activate T cells and improve the clinical efficacy in AML and MSLN-expressing solid tumors. Citation Format: Cuiqing Yang, Xiaohui Shao, Feng Zhou, Lei Zhou, Qi Deng, Yun Zhang, Guangcun Cheng, Yingying Hu, Huaiyuan Ma, Yadan Wu, Shuai Wang, Jie Zang, Lei Liu, Wenqing Yang, Yang Liu, Chunlei Xia, Jianzhong Hu, Ande Luo, Yayuan Fu, Zhuoxiao Cao, Renhong Tang. A multispecific T cell engager binding to both membrane proximal and membrane distal epitopes of MSLN with low affinity CD3 for the treatment of AML and solid tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 6343.
- Published
- 2022
- Full Text
- View/download PDF
7. Abstract 1029: Dissecting the mechanism of action targeting TNFR2 with a lignd blocking anti-TNFR2 antibody that elicits potent anti-tumor activity
- Author
-
Xiaoqing Liu, Wenqing Yang, Wenjing Li, Zhuoxiao Cao, Xiaofeng Zhao, Ran Pang, Zhenna Gao, Mengyu Wang, and Renhong Tang
- Subjects
Cancer Research ,education.field_of_study ,Tumor-infiltrating lymphocytes ,Chemistry ,T cell ,Population ,FOXP3 ,Immune system ,medicine.anatomical_structure ,Oncology ,Mechanism of action ,medicine ,Cancer research ,Tumor necrosis factor alpha ,medicine.symptom ,education ,CD8 - Abstract
Tumor necrosis factor receptor-2 (TNFR2) is a TNF receptor superfamily member and has been reported to have restricted expression in the immune system, including potent suppressive Tregs, myeloid suppressor cells and endothelial cells. The therapeutic potential of targeting TNFR2 for cancers has been previously demonstrated but with conflicting data reported by different groups. In order to gain further insight into the mechanism of action (MoA) of targeting TNFR2, we used an anti-murine TNFR2 surrogate antibody (suAb) and tested its functional activities both in vitro and in vivo. suAb was able to bind murine TNFR2 with high sub-nanomolar affinity, in the meantime, it can also block the interaction between murine TNFα and TNFR2 at both protein and cellular levels. CD4+ T cells were isolated from spleens of wildtype Balb/c mice, the co-incubation of which with murine TNFα and IL-2 rendering the significant proliferation of Treg within the CD4+ T cell population. Addition of suAb can potently reduce Treg proliferation while downregulating the expression of TNFR2 on these Tregs. Further detailed analysis by using isolated mouse FoxP3+ Treg confirmed that suAb could effectively suppress murine Treg proliferation induced by TNFα and IL-2. In vivo, suAb treatment resulted in significant tumor growth inhibition across various syngeneic tumor models (CT26, EMT6 and LL/2) as single agent or when combined with anti-mPD-1 (CT26). Tumor infiltrating lymphocytes (TILs) analysis revealed dramatic increase of the effective-memory pool of both CD4+ and CD8+ T cells together with an significantly improved CD8/Treg ratio. In addition, treatment with suAb led to decreased expression of PD-1 on tumor infiltrating CD8+ T cells suggesting a potential relieved exhaustion phenotype after the treatment. Based on this careful MoA characterization, corresponding human anti-TNFR2 lead antibodies have been identified and are currently under development as potential novel treatment for cancer patients. Citation Format: Xiaofeng Zhao, Xiaoqing Liu, Zhenna Gao, Mengyu Wang, Wenjing Li, Ran Pang, Wenqing Yang, Zhuoxiao Cao, Renhong Tang. Dissecting the mechanism of action targeting TNFR2 with a lignd blocking anti-TNFR2 antibody that elicits potent anti-tumor activity [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 1029.
- Published
- 2021
- Full Text
- View/download PDF
8. Construction of a versatile expression library for all human single-pass transmembrane proteins for receptor pairings by high throughput screening
- Author
-
Zhishuang Guan, Jinjing Xu, Dongxia Hao, Henrik Østergaard, Wei Yang, Lin Wang, Zhuoxiao Cao, Chih-Chuan Chang, Hong Cheng, Zibing Liu, Haibin Chen, Liu Wei, Chenxi Shen, Bing Shan, Fengting Xu, Di Liu, Bingke Yu, Wan Wang, Lujia Peng, Xun Li, Haisun Zhu, Jishu Wang, Haimei Xing, Zhiru Yang, Li Yang, Lin Li, Tengkun Li, Jing Su, Tzu-Yuan Chao, Zhe Sun, Yanping Du, Dapeng Li, Søren Berg Padkjær, Lishan Kang, Dennis Madsen, and Esper Boel
- Subjects
0301 basic medicine ,Recombinant Fusion Proteins ,High-throughput screening ,Receptors, Cell Surface ,Bioengineering ,Biology ,Transfection ,Applied Microbiology and Biotechnology ,Immunoglobulin Fab Fragments ,03 medical and health sciences ,Tissue factor ,Peptide Library ,Humans ,Cloning, Molecular ,Receptor ,Endothelial protein C receptor ,Membrane Proteins ,General Medicine ,Cell sorting ,Molecular biology ,Transmembrane protein ,High-Throughput Screening Assays ,Cell biology ,HEK293 Cells ,030104 developmental biology ,Immunoglobulin superfamily ,Biotechnology ,Protein ligand - Abstract
Interactions between protein ligands and receptors play crucial roles in cell-cell signalling. Most of the human cell surface receptors have been identified in the post-Human Genome Project era but many of their corresponding ligands remain unknown. To facilitate the pairing of orphan receptors, 2762 sequences encoding all human single-pass transmembrane proteins were selected for inclusion into a mammalian-cell expression library. This expression library, consisting of all the individual extracellular domains (ECDs), was constructed as a Fab fusion for each protein. In this format, individual ECD can be produced as a soluble protein or displayed on cell surface, depending on the applied heavy-chain Fab configuration. The unique design of the Fab fusion concept used in the library led to not only superior success rate of protein production, but also versatile applications in various high-throughput screening paradigms including protein-protein binding assays as well as cell binding assays, which were not possible for any other existing expression libraries. The protein library was screened against human coagulation factor VIIa (FVIIa), an approved therapeutic for the treatment of hemophilia, for binding partners by AlphaScreen and ForteBio assays. Two previously known physiological ligands of FVIIa, tissue factor (TF) and endothelial protein C receptor (EPCR) were identified by both assays. The cell surface displayed library was screened against V-domain Ig suppressor of T-cell activation (VISTA), an important immune-checkpoint regulator. Immunoglobulin superfamily member 11 (IgSF11), a potential target for cancer immunotherapy, was identified as a new and previously undescribed binding partner for VISTA. The specificity of the binding was confirmed and validated by both fluorescence-activated cell sorting (FACS) and surface plasmon resonance (SPR) assays in different experimental setups.
- Published
- 2017
- Full Text
- View/download PDF
9. Role of Krüppel-like factors in leukocyte development, function, and disease
- Author
-
Basak Icli, Akm Khyrul Wara, Mark W. Feinberg, Xinghui Sun, and Zhuoxiao Cao
- Subjects
Myeloid ,Cellular differentiation ,Immunology ,Kruppel-Like Transcription Factors ,Gene Expression ,Gene targeting ,Kruppel-like factors ,Review Article ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Hematopoiesis ,Leukemia ,Haematopoiesis ,medicine.anatomical_structure ,Leukocytes ,medicine ,Animals ,Humans ,Macrophage ,Transcription factor ,Signal Transduction - Abstract
The Krüppel-like transcription factor (KLF) family participates in diverse aspects of cellular growth, development, differentiation, and activation. Recently, several groups have identified new connections between the function of these factors and leukocyte responses in health and disease. Gene targeting of individual KLFs in mice has uncovered novel and unexpected physiologic roles among myeloid and lymphocyte cell lineage maturation, particularly in the bone marrow niche and blood. In addition, several KLF family members are downstream targets of stimuli and signaling pathways critical to T-cell trafficking, T regulatory cell differentiation or suppressor function, monocyte/macrophage activation or renewal, and B memory cell maturation or activation. Indeed, KLFs have been implicated in subtypes of leukemia, lymphoma, autoimmunity, and in acute and chronic inflammatory disease states, such as atherosclerosis, diabetes, and airway inflammation, raising the possibility that KLFs and their upstream signals are of therapeutic interest. This review focuses on the relevant literature of Krüppel-like factors in leukocyte biology and their implications in clinical settings.
- Published
- 2010
- Full Text
- View/download PDF
10. Antioxidants and Phase 2 Enzymes in Cardiomyocytes: Chemical Inducibility and Chemoprotection Against Oxidant and Simulated Ischemia-Reperfusion Injury
- Author
-
Jay L. Zweier, Hong Zhu, Li Zhang, Xue Zhao, Zhuoxiao Cao, and Yunbo Li
- Subjects
0301 basic medicine ,Blotting, Western ,Simulated ischemia ,Phase ii enzymes ,Oxidative phosphorylation ,medicine.disease_cause ,Antioxidants ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,NAD(P)H Dehydrogenase (Quinone) ,medicine ,Animals ,Myocytes, Cardiac ,RNA, Messenger ,Cardiac disorders ,Cytotoxicity ,Glutathione Transferase ,Glutathione Peroxidase ,Reverse Transcriptase Polymerase Chain Reaction ,Superoxide Dismutase ,Chemistry ,Chemoprotection ,Thiones ,Catalase ,Oxidants ,medicine.disease ,Glutathione ,Rats ,Oxidative Stress ,Glutathione Reductase ,030104 developmental biology ,Biochemistry ,Reperfusion Injury ,030220 oncology & carcinogenesis ,cardiovascular system ,Reperfusion injury ,Oxidative stress - Abstract
The increasing recognition of the role for oxidative stress in cardiac disorders has led to extensive investigation on the protection by exogenous antioxidants against oxidative cardiac injury. On the other hand, another strategy for protecting against oxidative cardiac injury may be through upregulation of the endogenous antioxidants and phase 2 enzymes in the myocardium by chemical inducers. However, our current understanding of the chemical inducibility of cardiac cellular antioxidants and phase 2 enzymes is very limited. In this study, using rat cardiac H9c2 cells we have characterized the concentration- and time-dependent induction of cellular antioxidants and phase 2 enzymes by 3H-1,2-dithiole-3-thione (D3T), and the resultant chemoprotective effects on oxidative cardiac cell injury. Incubation of H9c2 cells with D3T resulted in a marked concentration- and time-dependent induction of a number of cellular antioxidants and phase 2 enzymes, including catalase, reduced glutathione (GSH), GSH peroxidase, glutathione reductase (GR), GSH S-transferase (GST), and NAD(P)H:quinone oxidoreductase-1 (NQO1). D3T treatment of H9c2 cells also caused an increase in mRNA expression of catalase, γ-glutamylcysteine ligase catalytic subunit, GR, GSTA1, M1 and P1, and NQO1. Moreover, both mRNA and protein expression of Nrf2 were induced in D3T-treated cells. D3T pretreatment led to a marked protection against H9c2 cell injury elicited by various oxidants and simulated ischemia-reperfusion. D3T pretreatment also resulted in decreased intracellular accumulation of reactive oxygen in H9c2 cells after exposure to the oxidants as well as simulated ischemia-reperfusion. This study demonstrates that a series of endogenous antioxidants and phase 2 enzymes in H9c2 cells can be induced by D3T in a concentration- and time-dependent fashion, and that the D3T-upregulated cellular defenses are accompanied by a markedly increased resistance to oxidative cardiac cell injury.
- Published
- 2006
- Full Text
- View/download PDF
11. Upregulation of endogenous antioxidants and phase 2 enzymes by the red wine polyphenol, resveratrol in cultured aortic smooth muscle cells leads to cytoprotection against oxidative and electrophilic stress
- Author
-
Yunbo Li, Zhuoxiao Cao, and Hong Zhu
- Subjects
Xanthine Oxidase ,Myocytes, Smooth Muscle ,Glutathione reductase ,Wine ,Resveratrol ,medicine.disease_cause ,Antioxidants ,Muscle, Smooth, Vascular ,Superoxide dismutase ,chemistry.chemical_compound ,Stilbenes ,NAD(P)H Dehydrogenase (Quinone) ,medicine ,Animals ,Xanthine oxidase ,Aorta ,Cells, Cultured ,Glutathione Transferase ,Pharmacology ,chemistry.chemical_classification ,Aldehydes ,Glutathione Peroxidase ,Reactive oxygen species ,biology ,Superoxide Dismutase ,Glutathione peroxidase ,food and beverages ,Glutathione ,Catalase ,Rats ,Up-Regulation ,Isoenzymes ,Oxidative Stress ,Glutathione Reductase ,chemistry ,Biochemistry ,Cytoprotection ,biology.protein ,Reactive Oxygen Species ,Oxidative stress - Abstract
Resveratrol (3,4',5-trihydroxystilbene), a polyphenolic compound found in mulberries, grapes and red wine has been demonstrated to be capable of protecting against oxidative cardiovascular pathophysiology. However, the underlying cellular and biochemical mechanisms remain to be elucidated. This study was undertaken to determine if resveratrol could upregulate endogenous antioxidants and phase 2 enzymes in cultured aortic smooth muscle cells (ASMCs), and if such increased cellular defenses could provide protection against oxidative and electrophilic vascular cell injury. Incubation of rat ASMCs with resveratrol at low micromolar concentrations resulted in a significant induction of a scope of cellular antioxidants and phase 2 enzymes in a concentration- and/or time-dependent fashion. These cytoprotective factors include superoxide dismutase, catalase, glutathione, glutathione reductase, glutathione peroxidase, glutathione S-transferase (GST), and NAD(P)H:quinone oxidoreductase-1 (NOQ1). Notably, induction of catalase, GST, and NOQ1 was most remarkable among the above resveratrol-inducible antioxidants and phase 2 enzymes. Moreover, resveratrol treatment also significantly increased the mRNA expression of catalase, GSTA1, and NQO1 in a time-dependent manner. Pretreatment of ASMCs with resveratrol afforded a remarkable protection against xanthine oxidase (XO)/xanthine- or 4-hydroxy-2-nonenal-induced cytotoxicity, as assessed by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) reduction assay. Resveratrol pretreatment also led to a marked reduction in intracellular accumulation of reactive oxygen species in ASMCs after incubation with XO/xanthine. Taken together, this study demonstrates that a scope of key endogenous antioxidants and phase 2 enzymes in cultured ASMCs can be upregulated by resveratrol at low micromolar concentrations, and that such chemically-elevated cellular defenses rendered cells increased resistance to oxidative and electrophilic stress. The results of this study thus suggested a new mechanism, which might contribute to the cardiovascular protective effects of resveratrol.
- Published
- 2006
- Full Text
- View/download PDF
12. Differential roles of 3H-1,2-dithiole-3-thione-induced glutathione, glutathione S-transferase and aldose reductase in protecting against 4-hydroxy-2-nonenal toxicity in cultured cardiomyocytes
- Author
-
Hong Zhu, Michael A. Trush, Zhuoxiao Cao, and Yunbo Li
- Subjects
Cell Survival ,Biophysics ,Biochemistry ,Gene Expression Regulation, Enzymologic ,Cell Line ,chemistry.chemical_compound ,Aldehyde Reductase ,Animals ,Myocytes, Cardiac ,Buthionine sulfoximine ,Viability assay ,Molecular Biology ,Glutathione Transferase ,Aldehydes ,Aldose reductase ,biology ,Thiones ,Glutathione ,Cytoprotection ,Molecular biology ,Rats ,Up-Regulation ,Glutathione S-transferase ,chemistry ,Cell culture ,Inactivation, Metabolic ,biology.protein ,Sorbinil - Abstract
4-hydroxy-2-nonenal (HNE) plays an important role in the pathogenesis of cardiac disorders. While conjugation with glutathione (GSH) catalyzed by GSH S-transferase (GST) has been suggested to be a major detoxification mechanism for HNE in target cells, whether chemically upregulated cellular GSH and GST afford protection against HNE toxicity in cardiac cells has not been investigated. In addition, the differential roles of chemically induced GSH and GST as well as other cellular factors in detoxifying HNE in cardiomyocytes are unclear. In this study, we have characterized the induction of GSH and GST by 3H-1,2-dithiole-3-thione (D3T) and the protective effects of the D3T-elevated cellular defenses on HNE-mediated toxicity in rat H9C2 cardiomyocytes. Treatment of cardiomyocytes with D3T resulted in a significant induction of both GSH and GST as well as the mRNA expression of gamma-glutamylcysteine ligase catalytic subunit and GSTA. Both GSH and GST remained elevated for at least 72 h after removal of D3T from the culture media. Treatment of cells with HNE led to a significant decrease in cell viability and an increased formation of HNE-protein adducts. Pretreatment of cells with D3T dramatically protected against HNE-mediated cytotoxicity and protein-adduct formation. HNE treatment caused a significant decrease in cellular GSH level, which preceded the loss of cell viability. Either depletion of cellular GSH by buthionine sulfoximine (BSO) or inhibition of GST by sulfasalazine markedly sensitized the cells to HNE toxicity. Co-treatment of cardiomyocytes with BSO was found to completely block the D3T-mediated GSH elevation, which however failed to reverse the cytoprotective effects of D3T, suggesting that other cellular factor(s) might be involved in D3T cytotprotection. In this regard, D3T was shown to induce cellular aldose reductase (AR). Surprisingly, inhibition of AR by sorbinil failed to potentiate HNE toxicity in cardiomyocytes. In contrast, sorbinil dramatically augmented HNE cytotoxicity in cells with GSH depletion induced by BSO. Similarly, in BSO-treated cells, D3T cytoprotection was also largely reversed by sorbinil, indicating that AR played a significant role in detoxifying HNE only under the condition of GSH depletion in cardiomyocytes. Taken together, this study demonstrates that D3T can induce GSH, GST, and AR in cardiomyocytes, and that the above cellular factors appear to play differential roles in detoxification of HNE in cardiomyocytes.
- Published
- 2005
- Full Text
- View/download PDF
13. Induction of endogenous glutathione by the chemoprotective agent, 3H-1,2-dithiole-3-thione, in human neuroblastoma SH-SY5Y cells affords protection against peroxynitrite-induced cytotoxicity
- Author
-
Yunbo Li, Zhuoxiao Cao, Harry Z Qiu, Seema Hallur, and Xingxiang Peng
- Subjects
Chemoprotective agent ,SH-SY5Y ,Cell Survival ,Drug Resistance ,Biophysics ,Antineoplastic Agents ,Thiophenes ,Pharmacology ,Biochemistry ,Nitric oxide ,Neuroblastoma ,chemistry.chemical_compound ,Cell Line, Tumor ,Peroxynitrous Acid ,Humans ,Buthionine sulfoximine ,Cytotoxicity ,Molecular Biology ,Dose-Response Relationship, Drug ,Thiones ,Cell Biology ,Glutathione ,Cytoprotection ,chemistry ,Peroxynitrite - Abstract
Substantial evidence suggests that peroxynitrite generated from the bi-radical reaction of nitric oxide and superoxide is critically involved in the pathogenesis of neurodegenerative disorders, such as Parkinson's disease. Reaction with sulfhydryl (SH)-containing molecules has been proposed to be a major detoxification pathway of peroxynitrite in biological systems. This study was undertaken to determine if chemically elevated intracellular reduced glutathione (GSH), a major SH-containing biomolecule, affords protection against peroxynitrite-mediated toxicity in cultured neuronal cells. Incubation of human neuroblastoma SH-SY5Y cells with the unique chemoprotectant, 3H-1,2-dithiole-3-thione (D3T), led to a significant elevation of cellular GSH in a concentration-dependent fashion. To examine the protective effects of D3T-induced GSH on peroxynitrite-mediated toxicity, SH-SY5Y cells were pretreated with D3T and then exposed to either the peroxynitrite generator, 3-morpholinosydnonimine (SIN-1), or the authentic peroxynitrite. We observed that D3T-pretreated cells showed a markedly increased resistance to SIN-1- or authentic peroxynitrite-induced cytotoxicity, as assessed by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium reduction assay. Conversely, depletion of cellular GSH by buthionine sulfoximine (BSO) caused a marked potentiation of SIN-1- or authentic peroxynitrite-mediated cytotoxicity. To further demonstrate the causal role for GSH induction in D3T-mediated cytoprotection, SH-SY5Y cells were co-treated with BSO to abolish D3T-induced GSH elevation. Co-treatment of the cells with BSO was found to significantly reverse the protective effects of D3T on SIN-1- or authentic peroxynitrite-elicited cytotoxicity. Taken together, this study demonstrates for the first time that D3T can induce GSH in cultured SH-SY5Y cells, and that the D3T-augmented cellular GSH defense affords a marked protection against peroxynitrite-induced toxicity in cultured human neuronal cells.
- Published
- 2004
- Full Text
- View/download PDF
14. Potent induction of cellular antioxidants and phase 2 enzymes by resveratrol in cardiomyocytes: protection against oxidative and electrophilic injury
- Author
-
Yunbo Li and Zhuoxiao Cao
- Subjects
Xanthine Oxidase ,Antioxidant ,Cell Survival ,medicine.medical_treatment ,Oxidative phosphorylation ,Resveratrol ,medicine.disease_cause ,Antioxidants ,Superoxide dismutase ,chemistry.chemical_compound ,Stilbenes ,NAD(P)H Dehydrogenase (Quinone) ,medicine ,Animals ,Myocytes, Cardiac ,Xanthine oxidase ,Cells, Cultured ,Pharmacology ,chemistry.chemical_classification ,Aldehydes ,Glutathione Peroxidase ,Reactive oxygen species ,Antibiotics, Antineoplastic ,biology ,Superoxide Dismutase ,Glutathione ,Catalase ,Rats ,Oxidative Stress ,Glutathione Reductase ,chemistry ,Biochemistry ,Doxorubicin ,Enzyme Induction ,biology.protein ,Reactive Oxygen Species ,Oxidative stress - Abstract
Resveratrol is known to be protective against oxidative cardiovascular disorders. However, the underlying mechanisms remain unclear. This study was undertaken to determine if resveratrol could increase endogenous antioxidants and phase 2 enzymes in cardiomyocytes, and if such increased cellular defenses could provide protection against oxidative and electrophilic cell injury. Incubation of cardiac H9C2 cells with low micromolar resveratrol resulted in a significant induction of a scope of cellular antioxidants and phase 2 enzymes in a concentration- and/or time-dependent fashion. To investigate the protective effects of the resveratrol-induced cellular defenses on oxidative and electrophilic cell injury, H9C2 cells were first incubated with resveratrol, and then exposed to xanthine oxidase (XO)/xanthine, 4-hydroxy-2-nonenal or doxorubicin. We observed that resveratrol pretreatment afforded a marked protection against the above agent-mediated cytotoxicity in H9C2 cells. Moreover, the resveratrol pretreatment led to a great reduction in XO/xanthine-induced intracellular accumulation of ROS. Taken together, this study demonstrates that resveratrol induces antioxidants and phase 2 enzymes in cardiomyocytes, which is accompanied by increased resistance to oxidative and electrophilic cell injury.
- Published
- 2004
- Full Text
- View/download PDF
15. Protecting Against Peroxynitrite-Mediated Cytotoxicity in Vascular Smooth Muscle Cells Via Upregulating Endogenous Glutathione Biosynthesis by 3H-1,2-dithiole-3-thione
- Author
-
Yunbo Li and Zhuoxiao Cao
- Subjects
inorganic chemicals ,Vascular smooth muscle ,Cell Survival ,Antineoplastic Agents ,Thiophenes ,Pharmacology ,Biology ,Toxicology ,Muscle, Smooth, Vascular ,chemistry.chemical_compound ,Peroxynitrous Acid ,Animals ,Buthionine sulfoximine ,RNA, Messenger ,Cytotoxicity ,Buthionine Sulfoximine ,Molecular Biology ,Cells, Cultured ,Dose-Response Relationship, Drug ,Reverse Transcriptase Polymerase Chain Reaction ,Thiones ,Dipeptides ,Glutathione ,Molecular biology ,Cytoprotection ,Rats ,Up-Regulation ,Drug Combinations ,chemistry ,Molsidomine ,Toxicity ,cardiovascular system ,Cardiology and Cardiovascular Medicine ,Drug Antagonism ,Intracellular ,Peroxynitrite - Abstract
Peroxynitrite (ONOO(-)) is critically involved in the pathogenesis of cardiovascular diseases. Reaction with glutathione (GSH) was proposed to be a major detoxification pathway of ONOO(-) in the biological system. This study was undertaken to determine if chemically elevated intracellular GSH affords protection against ONOO(-)-mediated toxicity in vascular cells. Incubation of aortic smooth muscle A10 cells with 3H-1,2-dithiole-3-thione (D3T) led to a concentration- and time-dependent elevation of cellular GSH. Treatment of the cells with D3T also augmented protein and gene expression of gamma-glutamylcysteine ligase. To examine the effects of D3T-induced GSH on ONOO(-)-mediated toxicity, we pretreated A10 cells with D3T and then exposed them to either authentic ONOO(-) or the ONOO(-) generator, 3-morpholinosydnonimine. We observed that D3T pretreatment of A10 cells resulted in a significant protection against ONOO(-) cytotoxicity. Conversely, depletion of cellular GSH by buthionine sulfoximine (BSO) caused a marked potentiation of ONOO(-) cytotoxicity. To further demonstrate the causal involvement of GSH induction in D3T cytoprotection, we cotreated A10 cells with BSO to abolish D3T-induced GSH elevation. BSO cotreatment was found to greatly reverse the protective effects of D3T on ONOO(-)-elicited cytotoxicity. Taken together, our results demonstrate that upregulating GSH biosynthesis by D3T results in a marked protection against ONOO(-)-induced toxicity in vascular cells.
- Published
- 2004
- Full Text
- View/download PDF
16. Induction of cellular glutathione and glutathione S-transferase by 3H-1,2-dithiole-3-thione in rat aortic smooth muscle A10 cells: protection against acrolein-induced toxicity
- Author
-
Zhuoxiao Cao, Louis D. Trombetta, Yunbo Li, Diane Hardej, and Michael A. Trush
- Subjects
Chemoprotective agent ,Thiophenes ,Pharmacology ,Antioxidants ,Muscle, Smooth, Vascular ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Animals ,Buthionine sulfoximine ,Acrolein ,Aorta ,Cells, Cultured ,Glutathione Transferase ,Probability ,Analysis of Variance ,Dose-Response Relationship, Drug ,biology ,Thiones ,Glutathione ,Metabolism ,Rats ,Glutathione S-transferase ,Biochemistry ,chemistry ,Cell culture ,Inactivation, Metabolic ,Toxicity ,biology.protein ,Cardiology and Cardiovascular Medicine - Abstract
There is increasing evidence that aldehydes, including acrolein generated endogenously during the degradation process of biological molecules or the metabolism of foreign chemicals may be involved in the pathogenesis of cardiovascular diseases, such as atherosclerosis. Because glutathione (GSH) and GSH S-transferase (GST) are a major cellular defense against the toxic effects of reactive aldehydes, in this study we have characterized the inducibility of GSH and GST by the unique chemoprotective agent, 3H-1,2-dithiole-3-thione (D3T) and their protective effects against acrolein-induced toxicity in rat aortic smooth muscle A10 cells. Incubation of rat aortic A10 cells with micromolar concentrations of D3T resulted in a concentration- and time-dependent induction of both GSH and GST. Treatment of A10 cells with D3T also led to induction of gamma-glutamylcysteine synthetase, the key enzyme involved in GSH biosynthesis. Notably, the levels of GSH and GST remained higher than basal levels 72 h after removal of D3T from the culture media. To examine the protective effects of D3T-induced GSH and GST against reactive aldehyde-mediated toxicity, A10 cells were pretreated with D3T and then exposed to acrolein. Pretreatment of A10 cells with D3T resulted in a marked decrease of acrolein-induced toxicity as determined by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide reduction assay and morphological changes. To further demonstrate the involvement of GSH and GST in protecting against acrolein-induced toxicity, buthionine sulfoximine (BSO) and sulfasalazine were used to inhibit cellular GSH biosynthesis and GST activity, respectively. Either depletion of cellular GSH by BSO or inhibition of cellular GST by sulfasalazine led to a marked potentiation of acrolein-induced toxicity in A10 cells. Furthermore, co-treatment of cells with BSO was found to greatly abolish the protective effects of D3T on acrolein-induced toxicity. Taken together, our results demonstrate for the first time that both GSH and GST in aortic smooth muscle cells can be induced by D3T, and that this increased cellular defense affords great protection against reactive aldehyde-induced cardiovascular cell injury.
- Published
- 2003
- Full Text
- View/download PDF
17. The Role of Chemically Induced Glutathione and Glutathione S-Transferase in Protecting Against 4-Hydroxy-2-Nonenal- Mediated Cytotoxicity in Vascular Smooth Muscle Cells
- Author
-
Yunbo Li, Louis D. Trombetta, Zhuoxiao Cao, and Diane Hardej
- Subjects
Vascular smooth muscle ,Cell Survival ,Myocytes, Smooth Muscle ,Antineoplastic Agents ,Thiophenes ,Biology ,Toxicology ,Chemoprevention ,Muscle, Smooth, Vascular ,chemistry.chemical_compound ,Biosynthesis ,Animals ,Buthionine sulfoximine ,Viability assay ,Cytotoxicity ,Buthionine Sulfoximine ,Molecular Biology ,Aorta ,Cells, Cultured ,Glutathione Transferase ,Aldehydes ,Dose-Response Relationship, Drug ,Thiones ,Glutathione ,Molecular biology ,Rats ,Sulfasalazine ,Glutathione S-transferase ,chemistry ,Biochemistry ,Enzyme Induction ,Toxicity ,biology.protein ,Cardiology and Cardiovascular Medicine - Abstract
4-Hydroxy-2-nonenal (HNE) has been suggested to contribute to the pathogenesis of atherosclerosis. One of the major metabolic transformation pathways of HNE involves conjugation with glutathione (GSH) catalyzed by GSH S-transferase (GST). In this study, we have characterized the induction of GSH and GST by 3H-1,2-dithiole-3-thione (D3T) and the protective effects of the D3T-elevated cellular defenses on HNE-mediated toxicity in rat aortic smooth muscle A10 cells. Incubation of A10 cells with D3T resulted in a marked concentration- dependent induction of both GSH and GST. The induction of cellular GST by D3T also exhibited a time-dependent response. Pretreatment of A10 cells with D3T led to a dramatic decrease of HNE-induced cytotoxicity, as assessed by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) reduction assay and scanning electron microscopy. Incubation of A10 cells with HNE for 0.5 h and 1 h resulted in a significant depletion of cellular GSH, which preceded the decrease of cell viability. To further demonstrate the involvement of GSH and GST in protecting against HNE-induced cytotoxicity, buthionine sulfoximine (BSO) and sulfasalazine were used to inhibit cellular GSH biosynthesis and GST activity, respectively. Either depletion of GSH by BSO or inhibition of GST by sulfasalazine caused great potentiation of HNE-mediated cytotoxicity. Moreover, cotreatment of A10 cells with BSO was found to completely block the D3T-mediated GSH induction and to largely reverse the cytoprotective effects of D3T on HNE-induced toxicity. Taken together, this study demonstrates that D3T can induce both GSH and GST in aortic smooth muscle cells, and that the D3T-augmented cellular defenses afford a marked protection against HNE-induced vascular cell injury.
- Published
- 2003
- Full Text
- View/download PDF
18. Kruppel-like factor KLF10 targets transforming growth factor-beta1 to regulate CD4(+)CD25(-) T cells and T regulatory cells
- Author
-
Thomas C. Spelsberg, Harald von Boehmer, Zhuoxiao Cao, Göran K. Hansson, Irina Apostolou, Malayannan Subramaniam, Xinghui Sun, Basak Icli, Christopher J. Stapleton, Fehim Esen, Mark W. Feinberg, René R. Sevag Packard, Peter Libby, Karsten Kretschmer, and Akm Khyrul Wara
- Subjects
CD4-Positive T-Lymphocytes ,T cell ,chemical and pharmacologic phenomena ,Mice, Transgenic ,Cell Separation ,Biology ,Biochemistry ,Models, Biological ,T-Lymphocytes, Regulatory ,CCL5 ,Transforming Growth Factor beta1 ,Interleukin 21 ,Mice ,medicine ,Cytotoxic T cell ,Animals ,IL-2 receptor ,RNA, Small Interfering ,Molecular Biology ,ZAP70 ,Mechanisms of Signal Transduction ,Interleukin-2 Receptor alpha Subunit ,FOXP3 ,CD28 ,hemic and immune systems ,Cell Biology ,Atherosclerosis ,Flow Cytometry ,Molecular biology ,Cell biology ,DNA-Binding Proteins ,medicine.anatomical_structure ,Cytokines ,Transcription Factors - Abstract
CD4(+)CD25(+) regulatory T cells (T regs) play a major role in the maintenance of self-tolerance and immune suppression, although the mechanisms controlling T reg development and suppressor function remain incompletely understood. Herein, we provide evidence that Kruppel-like factor 10 (KLF10/TIEG1) constitutes an important regulator of T regulatory cell suppressor function and CD4(+)CD25(-) T cell activation through distinct mechanisms involving transforming growth factor (TGF)-beta1 and Foxp3. KLF10 overexpressing CD4(+)CD25(-) T cells induced both TGF-beta1 and Foxp3 expression, an effect associated with reduced T-Bet (Th1 marker) and Gata3 (Th2 marker) mRNA expression. Consistently, KLF10(-/-) CD4(+)CD25(-) T cells have enhanced differentiation along both Th1 and Th2 pathways and elaborate higher levels of Th1 and Th2 cytokines. Furthermore, KLF10(-/-) CD4(+)CD25(-) T cell effectors cannot be appropriately suppressed by wild-type T regs. Surprisingly, KLF10(-/-) T reg cells have reduced suppressor function, independent of Foxp3 expression, with decreased expression and elaboration of TGF-beta1, an effect completely rescued by exogenous treatment with TGF-beta1. Mechanistic studies demonstrate that in response to TGF-beta1, KLF10 can transactivate both TGF-beta1 and Foxp3 promoters, implicating KLF10 in a positive feedback loop that may promote cell-intrinsic control of T cell activation. Finally, KLF10(-/-) CD4(+)CD25(-) T cells promoted atherosclerosis by approximately 2-fold in ApoE(-/-)/scid/scid mice with increased leukocyte accumulation and peripheral pro-inflammatory cytokines. Thus, KLF10 is a critical regulator in the transcriptional network controlling TGF-beta1 in both CD4(+)CD25(-) T cells and T regs and plays an important role in regulating atherosclerotic lesion formation in mice.
- Published
- 2009
19. Nuclear factor E2-related factor 2-dependent myocardiac cytoprotection against oxidative and electrophilic stress
- Author
-
Li Zhang, Zhuoxiao Cao, Hara P. Misra, Yunbo Li, Masayuki Yamamoto, Michael A. Trush, Bhaba R. Misra, Hong Zhu, and Zhenquan Jia
- Subjects
Electrophoresis ,Time Factors ,Cell Survival ,NF-E2-Related Factor 2 ,Glutamate-Cysteine Ligase ,Glutathione reductase ,Oxidative phosphorylation ,Thiophenes ,Biology ,Toxicology ,medicine.disease_cause ,Antioxidants ,Gene Expression Regulation, Enzymologic ,chemistry.chemical_compound ,Mice ,Downregulation and upregulation ,medicine ,NAD(P)H Dehydrogenase (Quinone) ,Animals ,Myocytes, Cardiac ,Molecular Biology ,Cells, Cultured ,Glutathione Transferase ,chemistry.chemical_classification ,Mice, Knockout ,Glutathione Peroxidase ,Mice, Inbred ICR ,Dose-Response Relationship, Drug ,Superoxide Dismutase ,Glutathione peroxidase ,NADPH Dehydrogenase ,Thiones ,Glutathione ,Catalase ,Oxidants ,Molecular biology ,Cytoprotection ,Metabolic Detoxication, Phase II ,Oxidative Stress ,Glutathione Reductase ,chemistry ,Animals, Newborn ,Heme Oxygenase (Decyclizing) ,Cardiology and Cardiovascular Medicine ,Oxidative stress ,Peroxynitrite ,Signal Transduction - Abstract
Nuclear factor E2-related factor 2 (Nrf2) is a critical regulator of cytoprotective gene expression. However, the role of this transcription factor in myocardiac cytoprotection against oxidative and electrophilic stress remains unknown. This study was undertaken to investigate if Nrf2 signaling could control the constitutive and inducible expression of antioxidants and phase 2 enzymes in primary cardiomyocytes as well as the susceptibility of these cells to oxidative and electrophilic injury. The basal expression of a series of antioxidants and phase 2 enzymes was significantly lower in cardiomyocytes from Nrf2(-/-) mice than those from wild-type littermates. Incubation of wild-type cardiomyocytes with 3H-1,2-dithiole-3-thione (D3T) led to significant induction of various antioxidants and phase 2 enzymes, including catalase, glutathione, glutathione peroxidase (GPx), glutathione reductase, glutathione S-transferase, NAD(P)H:quinone oxidoreductase 1, and heme oxygenase-1. The inducibility of the above cellular defenses except GPx by D3T was abolished in Nrf2(-/-) cardiomyocytes. As compared to wild-type cells, Nrf2(-/-) cardiomyocytes were much more susceptible to cell injury induced by H(2)O(2), peroxynitrite, and 4-hydroxy-2-nonenal. Treatment of wild-type cardiomyocytes with D3T, which upregulated the cellular defenses, resulted in increased resistance to the above oxidant- and electrophile-induced cell injury, whereas D3T treatment of Nrf2(-/-) cardiomyocytes provided no cytoprotection. This study demonstrates that Nrf2 is an important factor in controlling both constitutive and inducible expression of a wide spectrum of antioxidants and phase 2 enzymes in cardiomyocytes and is responsible for protecting these cells against oxidative and electrophilic stress. These findings also implicate Nrf2 as an important signaling molecule for myocardiac cytoprotection.
- Published
- 2008
20. The Kruppel-like factor KLF4 is a critical regulator of monocyte differentiation
- Author
-
Mark W. Feinberg, Julie A. Segre, Frank Rosenbauer, Klaus H. Kaestner, Hiromi Iwasaki, Koichi Akashi, Jonathan P. Katz, Mukesh K. Jain, Zhuoxiao Cao, Hideyo Hirai, Maria A. Lebedeva, Richard L. Haspel, Susan Gray, Daniel G. Tenen, and Akm Khyrul Wara
- Subjects
Transcriptional Activation ,Cellular differentiation ,Hematopoietic System ,Kruppel-Like Transcription Factors ,Lipopolysaccharide Receptors ,Bone Marrow Cells ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Monocytes ,Kruppel-Like Factor 4 ,Mice ,stomatognathic system ,Cell Line, Tumor ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Cell Lineage ,RNA, Messenger ,Promoter Regions, Genetic ,Molecular Biology ,Transcription factor ,Myeloid Progenitor Cells ,General Immunology and Microbiology ,General Neuroscience ,Monocyte ,fungi ,Cell Differentiation ,DNA ,Rats ,Haematopoiesis ,medicine.anatomical_structure ,Phenotype ,KLF4 ,Monocyte differentiation ,embryonic structures ,Cancer research ,Trans-Activators ,Myelopoiesis ,sense organs ,Stem cell ,Protein Binding - Abstract
Monocyte differentiation involves the participation of lineage-restricted transcription factors, although the mechanisms by which this process occurs are incompletely defined. Within the hematopoietic system, members of the Kruppel-like family of factors (KLFs) play essential roles in erythrocyte and T lymphocyte development. Here we show that KLF4/GKLF is expressed in a monocyte-restricted and stage-specific pattern during myelopoiesis and functions to promote monocyte differentiation. Overexpression of KLF4 in HL-60 cells confers the characteristics of mature monocytes. Conversely, KLF4 knockdown blocked phorbol ester-induced monocyte differentiation. Forced expression of KLF4 in primary common myeloid progenitors (CMPs) or hematopoietic stem cells (HSCs) induced exclusive monocyte differentiation in clonogenic assays, whereas KLF4 deficiency inhibited monocyte but increased granulocyte differentiation. Mechanistic studies demonstrate that KLF4 is a target gene of PU.1. Consistently, KLF4 can rescue PU.1-/- fetal liver cells along the monocytic lineage and can activate the monocytic-specific CD14 promoter. Thus, KLF4 is a critical regulator in the transcriptional network controlling monocyte differentiation.
- Published
- 2007
21. Glutathione and glutathione-linked enzymes in normal human aortic smooth muscle cells: chemical inducibility and protection against reactive oxygen and nitrogen species-induced injury
- Author
-
Michael A. Trush, Hong Zhu, Yunbo Li, Li Zhang, and Zhuoxiao Cao
- Subjects
Xanthine Oxidase ,Vascular smooth muscle ,Antioxidant ,Glutamate-Cysteine Ligase ,medicine.medical_treatment ,Clinical Biochemistry ,Glutathione reductase ,Antineoplastic Agents ,Thiophenes ,Muscle, Smooth, Vascular ,chemistry.chemical_compound ,medicine ,Humans ,Buthionine sulfoximine ,RNA, Messenger ,Enzyme Inhibitors ,Buthionine Sulfoximine ,Molecular Biology ,Aorta ,Cells, Cultured ,Glutathione Transferase ,chemistry.chemical_classification ,Glutathione Peroxidase ,Reactive oxygen species ,Glutathione peroxidase ,Thiones ,Cell Biology ,General Medicine ,Glutathione ,Reactive Nitrogen Species ,Molecular biology ,Cytoprotection ,Mitochondria ,Glutathione Reductase ,Biochemistry ,chemistry ,Reactive Oxygen Species - Abstract
Substantial evidence suggests a crucial role for glutathione (GSH) and GSH-linked enzymes in protecting against oxidative vascular disorders. However, studies on the chemical inducibility of these antioxidant defenses and their protective effects on oxidant injury in normal human vascular cells are currently lacking. Accordingly, this study was undertaken to investigate the inducibility of GSH, glutathione reductase (GR), glutathione peroxidase (GPx), and glutathione S-transferase (GST) by the chemoprotective agent, 3H-1,2-dithiole-3-thione (D3T) in cultured normal human aortic smooth muscle cells (HASMCs). HASMCs expressed measurable levels/activities of GSH, GR, GPx, and GST. Incubation of HASMCs with low micromolar concentrations of D3T resulted in a marked elevation in total cellular GSH content and GR activity. The protein and mRNA expression of gamma-glutamylcysteine ligase (GCL) and GR were also upregulated by D3T. In addition, D3T caused significant increases in mitochondrial GSH content and GR activity. In contrast, neither cellular GPx nor GST activity was altered after D3T treatment. Pretreatment of HASMCs with D3T afforded remarkable protection against reactive oxygen and nitrogen species (ROS/RNS)-mediated cell injury. Depletion of cellular GSH by pretreatment with buthionine sulfoximine (BSO), an inhibitor of GSH biosynthesis led to marked potentiation of the ROS/RNS-induced cell injury. Moreover, co-treatment of HASMCs with BSO was found to completely abolish the D3T-mediated GSH elevation, and remarkably reverse D3T cytoprotection against the ROS/RNS-elicited injury. Taken together, this study demonstrates that both GSH/GCL and GR in normal HASMCs are inducible by D3T, and that upregulation of GSH biosynthesis appears to be the predominant mechanism underlying D3T-mediated cytoprotection against ROS/RNS-elicited injury to human vascular smooth muscle cells.
- Published
- 2007
- Full Text
- View/download PDF
22. Kruppel-like factor 4 is a mediator of proinflammatory signaling in macrophages
- Author
-
Maria A. Lebedeva, Akm Khyrul Wara, Sucharita SenBanerjee, Mark W. Feinberg, Zhuoxiao Cao, and Mukesh K. Jain
- Subjects
Lipopolysaccharides ,Transcription, Genetic ,Nitric Oxide Synthase Type II ,Kruppel-like factors ,Biochemistry ,Monocytes ,Mice ,Transforming Growth Factor beta ,p300-CBP Transcription Factors ,Phosphorylation ,Promoter Regions, Genetic ,KLF4 ,Cytokines ,Tumor necrosis factor alpha ,medicine.symptom ,Signal transduction ,Protein Binding ,Signal Transduction ,Blotting, Western ,Kruppel-Like Transcription Factors ,Down-Regulation ,Inflammation ,Enzyme-Linked Immunosorbent Assay ,Biology ,Transfection ,Models, Biological ,Proinflammatory cytokine ,Cell Line ,Transforming Growth Factor beta1 ,Interferon-gamma ,Kruppel-Like Factor 4 ,Mediator ,Coactivator ,Plasminogen Activator Inhibitor 1 ,medicine ,Animals ,RNA, Messenger ,Smad3 Protein ,Molecular Biology ,Nitrites ,Binding Sites ,Tumor Necrosis Factor-alpha ,Macrophages ,Transcription Factor RelA ,Cell Biology ,DNA ,Macrophage Activation ,Oligonucleotides, Antisense ,Blotting, Northern ,Protein Structure, Tertiary ,Mutation ,Cancer research ,Gene Deletion - Abstract
Activation of macrophages is important in chronic inflammatory disease states such as atherosclerosis. Proinflammatory cytokines such as interferon-gamma (IFN-gamma), lipopolysaccharide (LPS), or tumor necrosis factor-alpha can promote macrophage activation. Conversely, anti-inflammatory factors such as transforming growth factor-beta1 (TGF-beta1) can decrease proinflammatory activation. The molecular mediators regulating the balance of these opposing effectors remain incompletely understood. Herein, we identify Kruppel-like factor 4 (KLF4) as being markedly induced in response to IFN-gamma, LPS, or tumor necrosis factor-alpha and decreased by TGF-beta1 in macrophages. Overexpression of KLF4 in J774a macrophages induced the macrophage activation marker inducible nitric-oxide synthase and inhibited the TGF-beta1 and Smad3 target gene plasminogen activator inhibitor-1 (PAI-1). Conversely, KLF4 knockdown markedly attenuated the ability of IFN-gamma, LPS, or IFN-gamma plus LPS to induce the iNOS promoter, whereas it augmented macrophage responsiveness to TGF-beta1 and Smad3 signaling. The KLF4 induction of the iNOS promoter is mediated by two KLF DNA-binding sites at -95 and -212 bp, and mutation of these sites diminished induction by IFN-gamma and LPS. We further provide evidence that KLF4 interacts with the NF-kappaB family member p65 (RelA) to cooperatively induce the iNOS promoter. In contrast, KLF4 inhibited the TGF-beta1/Smad3 induction of the PAI-1 promoter independent of KLF4 DNA binding through a novel antagonistic competition with Smad3 for the C terminus of the coactivator p300/CBP. These findings support an important role for KLF4 as a regulator of key signaling pathways that control macrophage activation.
- Published
- 2005
23. The chemical inducibility of mouse cardiac antioxidants and phase 2 enzymes in vivo
- Author
-
Zhuoxiao Cao and Yunbo Li
- Subjects
Male ,Glutathione reductase ,Biophysics ,Endogeny ,Oxidative phosphorylation ,Thiophenes ,Pharmacology ,Biochemistry ,Antioxidants ,Superoxide dismutase ,chemistry.chemical_compound ,Mice ,In vivo ,Animals ,Quinone Reductases ,Molecular Biology ,Glutathione Transferase ,Cardioprotection ,Glutathione Peroxidase ,biology ,Superoxide Dismutase ,Myocardium ,Thiones ,Cell Biology ,Glutathione ,Catalase ,Mice, Inbred C57BL ,Glutathione Reductase ,chemistry ,Liver ,Enzyme Induction ,biology.protein - Abstract
The recognition of the critical involvement of oxidative and electrophilic stress in cardiac disorders has led to extensive investigation of the protective effects of exogenous antioxidants on cardiac injury. On the other hand, another strategy for protecting against oxidative/electrophilic cardiac injury may be through induction of the endogenous antioxidants and phase 2 enzymes in myocardium by chemical inducers. However, our understanding of the chemical inducibility of cardiac antioxidants/phase 2 enzymes in vivo is very limited. In addition, careful studies on the basal levels of a scope of endogenous antioxidants/phase 2 enzymes in myocardium as compared with other tissues, such as liver, are lacking. Accordingly, this study was undertaken to determine the basal levels of endogenous antioxidants/phase 2 enzymes, including superoxide dismutase (SOD), catalase, reduced glutathione (GSH), GSH peroxidase (GPx), glutathione reductase (GR), GSH S-transferase (GST), and NAD(P)H:quinone oxidoreductase 1 (NQO1), and investigate the inducibility of the above antioxidants/phase 2 enzymes by the chemoprotectant, 1,2-dithiole-3-thione (D3T), in cardiac as well as hepatic tissues in C57BL/6 mice. Our results demonstrated that in C57BL/6 mice, the levels of catalase, GSH, GPx, GR, and GST were significantly lower in cardiac tissue than in hepatic tissue. The level of total SOD did not differ significantly between mouse heart and liver. Notably, heart contained a much higher NQO1 activity than liver. Immunoblotting and RT-PCR analyses further demonstrated the high expression of NQO1 protein and mRNA in myocardium. Oral administration of D3T at 0.25 and 0.5 mmol/kg body weight for 3 consecutive days resulted in a significant induction of cardiac SOD, catalase, GR, GST, and NQO1. No significant induction of cardiac GSH and GPx was observed with the above D3T treatment. Only GR, GST, and NQO1 in mouse liver were induced by the D3T treatment. Unexpectedly, we observed a significant D3T dose-dependent decrease in hepatic GPx activity. Taken together, this study demonstrates for the first time that: (1) the expression of NQO1 is remarkably high in mouse myocardium though other cardiac antioxidants/phase 2 enzymes are relatively lower as compared with liver; (2) a number of endogenous antioxidants/phase 2 enzymes in mouse cardiac tissue can be significantly induced by D3T following oral administration; and (3) the inducibility of endogenous antioxidants/phase 2 enzymes by D3T differs between mouse cardiac and hepatic tissues. This study provides a basis for future investigation of the cardioprotection of chemically induced endogenous antioxidants and phase 2 enzymes in myocardium in animal models of oxidative/electrophilic cardiac disorders.
- Published
- 2004
24. Potent inhibition of peroxynitrite-induced DNA strand breakage by ethanol: possible implications for ethanol-mediated cardiovascular protection
- Author
-
Yunbo Li and Zhuoxiao Cao
- Subjects
Heart Diseases ,Bicarbonate ,Biology ,Pathogenesis ,chemistry.chemical_compound ,Oxygen Consumption ,Peroxynitrous Acid ,Cytotoxic T cell ,Cytotoxicity ,Incubation ,Pharmacology ,Ethanol ,Rhodamines ,Central Nervous System Depressants ,Molecular biology ,Bicarbonates ,chemistry ,Biochemistry ,Molsidomine ,DNA, Viral ,Oxidation-Reduction ,DNA ,Peroxynitrite ,Bacteriophage phi X 174 ,DNA Damage ,Plasmids - Abstract
Epidemiological studies have conclusively demonstrated that moderate consumption of ethanol is causally associated with a significant reduction in cardiovascular events. However, the exact mechanisms underlying the ethanol-mediated cardiovascular protection remain to be elucidated. Because peroxynitrite has been extensively implicated in the pathogenesis of various forms of cardiovascular disorders via its cytotoxic effects, this study was undertaken to investigate if ethanol could inhibit peroxynitrite-induced DNA strand breaks, a critical event leading to peroxynitrite-elicited cytotoxicity. Toward this goal, φX-174 RF I plasmid DNA was used as an in vitro model to determine the protective effects of ethanol on peroxynitrite-induced DNA strand breaks. Incubation of φX-174 plasmid DNA with the peroxynitrite generator, 3-morpholinosydnonimine (SIN-1) led to the formation of both single- and double-stranded DNA breaks in a concentration- and time-dependent fashion. The presence of ethanol at concentrations ranging from 0.01 to 1% (w/v) resulted in a significant inhibition of SIN-1-induced DNA strand breaks. Ethanol also showed inhibitory effects on SIN-1-induced DNA strand breakage in the presence of bicarbonate. The inhibition of SIN-1-induced DNA strand breaks by ethanol exhibited a concentration-dependent manner. Notably, a marked inhibition of SIN-1-elicited DNA strand breaks was observed with 0.01% ethanol. Ethanol at 0.01–1% was unable to affect SIN-1-mediated oxygen consumption, indicating that ethanol did not affect the auto-oxidation of SIN-1 to form peroxynitrite. Furthermore, incubation of the plasmid DNA with authentic peroxynitrite resulted in a significant formation of DNA strand breaks, which could be dramatically inhibited by the presence of 0.02–0.1% ethanol. Taken together, this study demonstrates for the first time that ethanol at physiologically relevant concentrations can potently inhibit peroxynitrite-induced DNA strand breakage. In view of the critical involvement of peroxynitrite in cardiovascular disorders, the results of this study might have implications for the cardiovascular protection associated with moderate consumption of ethanol in humans.
- Published
- 2003
25. Induction of endogenous antioxidants and phase 2 enzymes by alpha-lipoic acid in rat cardiac H9C2 cells: protection against oxidative injury
- Author
-
Maggie Tsang, Hai Zhao, Zhuoxiao Cao, and Yunbo Li
- Subjects
Time Factors ,Glutathione reductase ,Biophysics ,Tetrazolium Salts ,medicine.disease_cause ,Biochemistry ,Models, Biological ,Xanthine ,Antioxidants ,chemistry.chemical_compound ,medicine ,Animals ,Xanthine oxidase ,Coloring Agents ,Molecular Biology ,Cells, Cultured ,Glutathione Transferase ,chemistry.chemical_classification ,Reactive oxygen species ,Glutathione Peroxidase ,biology ,Dose-Response Relationship, Drug ,Thioctic Acid ,Myocardium ,Cell Biology ,Glutathione ,Rats ,Oxygen ,Lipoic acid ,Oxidative Stress ,Thiazoles ,Glutathione Reductase ,chemistry ,Models, Chemical ,Catalase ,biology.protein ,Reactive Oxygen Species ,Oxidative stress - Abstract
Alpha-lipoic acid (LA) has recently been reported to exert protective effects on various forms of oxidative cardiac disorders. However, the mechanisms underlying LA-mediated cardioprotection remain to be investigated. This study was undertaken to determine whether LA treatment could increase endogenous antioxidants and phase 2 enzymes in cultured cardiomyocytes, and whether such increased cellular defenses could afford protection against oxidative cardiac cell injury. Incubation of rat cardiac H9C2 cells with low micromolar concentrations of LA resulted in a significant induction of a scope of cellular antioxidants and phase 2 enzymes in a concentration- and/or time-dependent fashion. These include catalase, reduced glutathione, glutathione reductase, glutathione S-transferase, and NAD(P)H:quinone oxidoreductase-1 (NOQ1). Induction of catalase and NOQ1 was most dramatic among the above LA-inducible antioxidants and phase 2 enzymes. To further investigate the protective effects of the LA-induced cellular defenses on oxidative cardiac cell injury, H9C2 cells were pretreated with LA (25-100 microM) for 72h and then exposed to xanthine oxidase (XO)/xanthine, a system that generates reactive oxygen species (ROS), for another 24h. We observed that LA pretreatment of H9C2 cells led to a marked protection against XO/xanthine-mediated cytotoxicity, as detected by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium reduction assay. The cytoprotective effects also exhibited a LA concentration-dependent fashion. Moreover, the LA pretreatment resulted in a great inhibition of intracellular accumulation of ROS in H9C2 cells following incubation with XO/xanthine. Taken together, this study demonstrates for the first time that a number of endogenous antioxidants and phase 2 enzymes in cultured cardiomyocytes can be induced by LA at low micromolar concentrations, and that the LA-mediated elevation of cellular defenses is accompanied by a markedly increased resistance to ROS-elicited cardiac cell injury. The results of this study have important implications for the cardioprotective effects of LA.
- Published
- 2003
26. The neuroprotectant ebselen inhibits oxidative DNA damage induced by dopamine in the presence of copper ions
- Author
-
Yunbo Li and Zhuoxiao Cao
- Subjects
Azoles ,DNA damage ,Cations, Divalent ,Dopamine ,Isoindoles ,medicine.disease_cause ,Neuroprotection ,chemistry.chemical_compound ,Organoselenium Compounds ,medicine ,Humans ,chemistry.chemical_classification ,Reactive oxygen species ,Dose-Response Relationship, Drug ,Chemistry ,Ebselen ,General Neuroscience ,Glutathione ,Oxidative Stress ,Neuroprotective Agents ,Biochemistry ,Biophysics ,Catecholamine ,Oxidation-Reduction ,Oxidative stress ,Copper ,medicine.drug ,DNA Damage ,Plasmids - Abstract
Ebselen (2-phenyl-1,2-benzisoselenazol-3(2H)-one), a seleno-organic compound with glutathione peroxidase-like activity, has been shown to be protective against brain ischemic injury and Parkinson's disease. This study was undertaken to investigate the protective effects of ebselen on oxidative DNA damage induced by dopamine in the presence of copper ions. Incubation of phiX-174 plasmid DNA with micromolar dopamine in the presence of Cu(II) resulted in a concentration-dependent induction of DNA strand breaks. Both a Cu(II)/Cu(I) redox cycle and H(2)O(2) formation were critically involved in the induction of DNA strand breaks by the dopamine/Cu(II) system. The presence of ebselen at micromolar concentrations led to a marked concentration-dependent inhibition of DNA strand breaks induced by the dopamine/Cu(II) system. Further studies showed that ebselen did not affect either the Cu(II)-mediated oxidation of dopamine to dopamine quinone or the reduction of Cu(II) to Cu(I) by dopamine. Instead, the presence of ebselen resulted in a marked decrease in the levels of H(2)O(2) derived from the Cu(II)-mediated oxidation of dopamine. Taken together, our results demonstrate for the first time that ebselen is able to inhibit the dopamine/Cu(II)-induced oxidative DNA damage, which appears to be attributable to the ability of ebselen to decrease the levels of H(2)O(2) derived from the dopamine/Cu(II) system. Since oxidative DNA damage has been implicated in the pathogenesis of various neurodegenerative diseases, the inhibition of oxidative DNA damage by ebselen may be responsible, at least partially, for its neuroprotective activities observed in both humans and experimental animals.
- Published
- 2002
27. Chemical induction of cellular antioxidants affords marked protection against oxidative injury in vascular smooth muscle cells
- Author
-
Zhuoxiao Cao and Yunbo Li
- Subjects
Cytoplasm ,Xanthine Oxidase ,Vascular smooth muscle ,Cell Survival ,Biophysics ,Thiophenes ,Pharmacology ,medicine.disease_cause ,Biochemistry ,Antioxidants ,Muscle, Smooth, Vascular ,4-Hydroxynonenal ,Cell Line ,Superoxide dismutase ,chemistry.chemical_compound ,medicine ,Animals ,Xanthine oxidase ,Molecular Biology ,chemistry.chemical_classification ,Reactive oxygen species ,Aldehydes ,biology ,Dose-Response Relationship, Drug ,Superoxide Dismutase ,Thiones ,Cell Biology ,Glutathione ,Catalase ,Cytoprotection ,Rats ,Oxidative Stress ,chemistry ,biology.protein ,Oxidative stress ,Cadmium - Abstract
Extensive evidence suggests that reactive oxygen species are critically involved in the pathogenesis of cardiovascular diseases, such as atherosclerosis and myocardial ischemia-reperfusion injury. Consistent with this concept, administration of exogenous antioxidants has been shown to be protective against oxidative cardiovascular injury. However, whether induction of endogenous antioxidants by chemical inducers in vasculature also affords protection against oxidative vascular cell injury has not been extensively investigated. In this study, using rat aortic smooth muscle A10 cells as an in vitro system, we have studied the induction of cellular antioxidants by the unique chemoprotector, 3H-1,2-dithiole-3-thione [corrected] (D3T) and the protective effects of the D3T-induced cellular antioxidants against oxidative cell injury. Incubation of A10 cells with micromolar concentrations of D3T for 24 h resulted in a significant induction of a battery of cellular antioxidants in a concentration-dependent manner. These included reduced glutathione (GSH), GSH peroxidase, GSSG reductase, GSH S-transferase, superoxide dismutase, and catalase. To further examine the protective effects of the induced endogenous antioxidants against oxidative cell injury, A10 cells were pretreated with D3T and then exposed to either xanthine oxidase (XO)/xanthine, 4-hydroxynonenal, or cadmium. We observed that D3T pretreatment of A10 cells led to significant protection against the cytotoxicity induced by XO/xanthine, 4-hydroxynonenal or cadmium, as determined by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium reduction assay. Taken together, this study demonstrates for the first time that a number of endogenous antioxidants in vascular smooth muscle cells can be induced by exposure to D3T, and that this chemical induction of cellular antioxidants is accompanied by markedly increased resistance to oxidative vascular cell injury.
- Published
- 2002
28. Different effects of genistein and resveratrol on oxidative DNA damage in vitro
- Author
-
Zhuoxiao Cao, Yunbo Li, Michael A. Trush, William Win, and Xingxiang Peng
- Subjects
DNA damage ,Health, Toxicology and Mutagenesis ,Genistein ,Oxidative phosphorylation ,Resveratrol ,Antioxidants ,chemistry.chemical_compound ,Stilbenes ,Genetics ,Anticarcinogenic Agents ,chemistry.chemical_classification ,Reactive oxygen species ,Hydroquinone ,food and beverages ,DNA ,Hydrogen Peroxide ,In vitro ,Hydroquinones ,chemistry ,Biochemistry ,Oxidation-Reduction ,Copper ,DNA Damage - Abstract
Previous studies have demonstrated that phenolic compounds, including genistein (4',5,7-trihydroxyisoflavone) and resveratrol (3,4',5-trihydroxystilbene), are able to protect against carcinogenesis in animal models. This study was undertaken to examine the ability of genistein and resveratrol to inhibit reactive oxygen species (ROS)-mediated strand breaks in phi X-174 plasmid DNA. H(2)O(2)/Cu(II) and hydroquinone/Cu(II) were used to cause oxidative DNA strand breaks in the plasmid DNA. We demonstrated that the presence of genistein at micromolar concentrations resulted in a marked inhibition of DNA strand breaks induced by either H(2)O(2)/Cu(II) or hydroquinone/Cu(II). Genistein neither affected the Cu(II)/Cu(I) redox cycle nor reacted with H(2)O(2) suggest that genistein may directly scavenge the ROS that participate in the induction of DNA strand breaks. In contrast to the inhibitory effects of genistein, the presence of resveratrol at similar concentrations led to increased DNA strand breaks induced by H(2)O(2)/Cu(II). Further studies showed that in the presence of Cu(II), resveratrol, but not genistein was able to cause DNA strand breaks. Moreover, both Cu(II)/Cu(I) redox cycle and H(2)O(2) were shown to be critically involved in resveratrol/copper-mediated DNA strand breaks. The above results indicate that despite their similar in vivo anticarcinogenic effects, genistein and resveratrol appear to exert different effects on oxidative DNA damage in vitro.
- Published
- 2001
29. Kruppel-Like Factor 10 (KLF10)-Deficient Mice Have Marked Defects In EPC Differentiation, Function, and Angiogenesis
- Author
-
Thomas C. Spelsberg, Kevin Croce, Basak Icli, Jeffery L. Kutok, Shi-Yin Foo, Scott J. Rodig, Xinghui Sun, Fehim Esen, Zhuoxiao Cao, Mark W. Feinberg, Anthony Rosenzweig, Yevgenia Tesmenitsky, Malayannan Subramaniam, and Akm Khyrul Wara
- Subjects
Angiogenesis ,Immunology ,Ischemia ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Neovascularization ,Haematopoiesis ,medicine.anatomical_structure ,cardiovascular system ,medicine ,Cancer research ,Bone marrow ,Stem cell ,Progenitor cell ,medicine.symptom ,Signal transduction - Abstract
Abstract 4314 Background: Emerging evidence demonstrates that endothelial progenitor cells (EPCs) may originate from the bone marrow and are capable of being recruited to sites of ischemic injury and contribute to neovascularization. However, the identities of these bone marrow cells and the signaling pathways that regulate their differentiation into functional EPCs remain poorly understood. Methods and Results: We previously identified that among hematopoietic progenitor stem cells, common myeloid progenitors (CMPs) and granulocyte-macrophage progenitors (GMPs) can preferentially differentiate into EPCs and possess high angiogenic activity under ischemic conditions compared to megakaryocyte-erythrocyte progenitors (MEPs), hematopoietic stem cells (HSCs), and common lymphoid progenitors (CLPs). Herein, we identify that a TGF-β1-responsive Kruppel-like Factor, KLF10, is robustly expressed in EPCs derived from CMPs and GMPs, compared to progenitors lacking EPC markers. KLF10–/– mice have marked defects in circulating EPCs (–23.6% vs. WT, P Background: Collectively, these observations identify that TGF-β1 signaling and KLF10 are part of a key signaling pathway that regulates EPC differentiation from CMPs and GMPs and may provide a therapeutic target during cardiovascular ischemic states. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2010
- Full Text
- View/download PDF
30. Erratum to 'Chemical Induction of Cellular Antioxidants Affords Marked Protection against Oxidative Injury in Vascular Smooth Muscle Cells' [Biochem. Biophys. Res. Commun. 292 (2002) 50–57]
- Author
-
Yunbo Li and Zhuoxiao Cao
- Subjects
Vascular smooth muscle ,Chemistry ,Biophysics ,Oxidative injury ,Cell Biology ,Molecular Biology ,Biochemistry ,Cell biology - Published
- 2002
- Full Text
- View/download PDF
31. Kruppel-like Factor KLF10 Targets Transforming Growth Factor-β1 to Regulate CD4+CD25- T Cells and T Regulatory Cells.
- Author
-
Zhuoxiao Cao, Wara, Akm Khyrul, IcIi, Basak, Xinghui Sun, Packard, René R. S., Esen, Fehim, StapIeton, Christopher J., Subramaniam, Malayannan, Kretschmer, Karsten, Apostolou, Irina, Von Boehmer, Harald, Hansson, Göran K., SpeIsberg, Thomas C., Libby, Peter, and Feinberg, Mark W.
- Subjects
- *
TRANSFORMING growth factors , *T cells , *IMMUNOSUPPRESSION , *CELL growth , *ATHEROSCLEROSIS treatment - Abstract
CD4+CD25+ regulatory T cells (T regs) play a major role in the maintenance of self-tolerance and immune suppression, although the mechanisms controlling T reg development and suppressor function remain incompletely understood. Herein, we provide evidence that Kruppel-like factor 10 (KLF1O/TIEG1) constitutes an important regulator of T regulatory cell suppressor function and CD4+CD25 T cell activation through distinct mechanisms involving transforming growth factor (TGF)-β1 and Foxp3. KLF10 overexpressing CD4+CD25 T cells induced both TGF-β1 and Foxp3 expression, an effect associated with reduced T-Bet (Thi marker) and Gata3 (Th2 marker) mRNA expression. Consistently, KLF10-/- CD4+CD25- T cells have enhanced differentiation along both Th1 and Th2 pathways and elaborate higher levels of Th1 and Th2 cytokines. Furthermore, KLF10-/- CD4+CD25 T cell effectors cannot be appropriately suppressed by wild-type T regs. Surprisingly, KLF10-/- T reg cells have reduced suppressor function, independent of Foxp3 expression, with decreased expression and elaboration of TGF-β1, an effect completely rescued by exogenous treatment with TGF-β1. Mechanistic studies demonstrate that in response to TGF-β1, KLF10 can transactivate both TGF-β1 and Foxp3 promoters, implicating KLF10 in a positive feedback ioop that may promote cell-intrinsic control of T cell activation. Finally, KLF10-/- CD4+CD25 T cells promoted atherosclerosis by ∼2-fold in ApoE-/-/scid/scid mice with increased leukocyte accumulation and peripheral pro-inflammatory cytokines. Thus, KLF10 is a critical regulator in the transcriptional network controlling TGF-β1 in both CD4+CD25 T cells and T regs and plays an important role in regulating atherosclerotic lesion formation in mice. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
32. Nuclear Factor E2-Related Factor 2-Dependent Myocardiac Cytoprotection Against Oxidative and Electrophilic Stress.
- Author
-
Hong Zhu, Zhenquan Jia, Misra, Bhaba R., Li Zhang, Zhuoxiao Cao, Yamamoto, Masayuki, Trush, Michael A., Misra, Hara P., and Yunbo Li
- Subjects
GENE expression ,ANTIOXIDANTS ,ENZYMES ,HEMOGLOBINS ,TRANSCRIPTION factors - Abstract
Nuclear factor E2-related factor 2 (Nrf2) is a critical regulator of cytoprotective gene expression. How- ever, the role of this transcription factor in myocardiac cytoprotection against oxidative and electrophilic stress remains unknown. This study was undertaken to investigate if Nrf2 signaling could control the constitutive and inducible expression of antioxidants and phase 2 enzymes in primary cardiomyocytes as well as the susceptibility of these cells to oxidative and electrophilic injury. The basal expression of a series of antioxidants and phase 2 enzymes was significantly lower in cardiomyocytes from Nrf2
-/- mice than those from wild-type littermates. Incubation of wild-type cardiomyocytes with 3H-1,2-dithiole-3-thione (D3T) led to significant induction of various antioxidants and phase 2 enzymes, including catalase, glutathione, glutathione peroxidase (GPx), glutathione reductase, glutathione S-transferase, NAD(P)H:quinone oxidoreductase 1, and heme oxygenase-1. The inducibility of the above cellular defenses except GPx by D3T was abolished in Nr2-/- cardiomyocytes. As compared to wild-type cells, Nrf2-/- cardiomyocytes were much more susceptible to cell injury induced by H2 O2 , peroxynitrite, and 4-hydroxy-2-nonenal. Treatment of wild-type cardiomyocytes with D3T, which upregulated the cellular defenses, resulted in increased resistance to the above oxidant- and electrophile-induced cell injury, whereas D3T treatment of Nrf2-/- cardiomyocytes provided no cytoprotection. This study demonstrates that Nrf2 is an important factor in controlling both constitutive and inducible expression of a wide spectrum of antioxidants and phase 2 enzymes in cardiomyocytes and is responsible for protecting these cells against oxidative and electrophilic stress. These findings also implicate Nrf2 as an important signaling molecule for myocardiac cytoprotection. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
33. The Kruppel-like factor KLF4 is a critical regulator of monocyte differentiation.
- Author
-
Feinberg, Mark W., Wara, Akm Khyrul, Zhuoxiao Cao, Lebedeva, Maria A., Rosenbauer, Frank, Iwasaki, Hiromi, Hirai, Hideyo, Katz, Jonathan P., Haspel, Richard L., Gray, Susan, Akashi, Koichi, Segre, Julie, Kaestner, Klaus H., Tenen, Daniel G., and Jain, Mukesh K.
- Subjects
HEMATOPOIETIC system ,MONOCYTES ,CELL differentiation ,TRANSCRIPTION factors ,ERYTHROCYTES ,T cells ,HEMATOPOIETIC stem cells - Abstract
Monocyte differentiation involves the participation of lineage-restricted transcription factors, although the mechanisms by which this process occurs are incompletely defined. Within the hematopoietic system, members of the Kruppel-like family of factors (KLFs) play essential roles in erythrocyte and T lymphocyte development. Here we show that KLF4/GKLF is expressed in a monocyte-restricted and stage-specific pattern during myelopoiesis and functions to promote monocyte differentiation. Overexpression of KLF4 in HL-60 cells confers the characteristics of mature monocytes. Conversely, KLF4 knockdown blocked phorbol ester-induced monocyte differentiation. Forced expression of KLF4 in primary common myeloid progenitors (CMPs) or hematopoietic stem cells (HSCs) induced exclusive monocyte differentiation in clonogenic assays, whereas KLF4 deficiency inhibited monocyte but increased granulocyte differentiation. Mechanistic studies demonstrate that KLF4 is a target gene of PU.1. Consistently, KLF4 can rescue PU.1−/− fetal liver cells along the monocytic lineage and can activate the monocytic-specific CD14 promoter. Thus, KLF4 is a critical regulator in the transcriptional network controlling monocyte differentiation. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
34. Glutathione and glutathione-linked enzymes in normal human aortic smooth muscle cells: chemical inducibility and protection against reactive oxygen and nitrogen species-induced injury.
- Author
-
Hong Zhu, Zhuoxiao Cao, Li Zhang, and Michael Trush
- Abstract
Abstract Substantial evidence suggests a crucial role for glutathione (GSH) and GSH-linked enzymes in protecting against oxidative vascular disorders. However, studies on the chemical inducibility of these antioxidant defenses and their protective effects on oxidant injury in normal human vascular cells are currently lacking. Accordingly, this study was undertaken to investigate the inducibility of GSH, glutathione reductase (GR), glutathione peroxidase (GPx), and glutathione S-transferase (GST) by the chemoprotective agent, 3H-1,2-dithiole-3-thione (D3T) in cultured normal human aortic smooth muscle cells (HASMCs). HASMCs expressed measurable levels/activities of GSH, GR, GPx, and GST. Incubation of HASMCs with low micromolar concentrations of D3T resulted in a marked elevation in total cellular GSH content and GR activity. The protein and mRNA expression of γ-glutamylcysteine ligase (GCL) and GR were also upregulated by D3T. In addition, D3T caused significant increases in mitochondrial GSH content and GR activity. In contrast, neither cellular GPx nor GST activity was altered after D3T treatment. Pretreatment of HASMCs with D3T afforded remarkable protection against reactive oxygen and nitrogen species (ROS/RNS)-mediated cell injury. Depletion of cellular GSH by pretreatment with buthionine sulfoximine (BSO), an inhibitor of GSH biosynthesis led to marked potentiation of the ROS/RNS-induced cell injury. Moreover, co-treatment of HASMCs with BSO was found to completely abolish the D3T-mediated GSH elevation, and remarkably reverse D3T cytoprotection against the ROS/RNS-elicited injury. Taken together, this study demonstrates that both GSH/GCL and GR in normal HASMCs are inducible by D3T, and that upregulation of GSH biosynthesis appears to be the predominant mechanism underlying D3T-mediated cytoprotection against ROS/RNS-elicited injury to human vascular smooth muscle cells. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
35. Kruppel-like Factor 4 Is a Mediator of Proinflammatory Signaling in Macrophages.
- Author
-
Feinberg, Mark W., Zhuoxiao Cao, Wara, Akm Khyrul, Lebedeva, Maria A., Senbanerjee, Sucharita, and Jam, Mukesh K.
- Subjects
- *
MACROPHAGE activation , *INFLAMMATORY mediators , *ATHEROSCLEROSIS , *TUMOR necrosis factors , *IMMUNOREGULATION , *NITRIC-oxide synthases , *PROTEOLYTIC enzymes , *CELLULAR immunity - Abstract
Activation of macrophages is important in chronic inflammatory disease states such as atherosclerosis. Proinflammatory cytokines such as interferon-γ (IFN-γ), lipopolysaccharide (LPS), or tumor necrosis factor-α can promote macrophage activation. Conversely, anti-inflammatory factors such as transforming growth factor-β1 (TGF-β1) can decrease proinflammatory activation. The molecular mediators regulating the balance of these opposing effectors remain incompletely understood. Herein, we identify Kruppel-like factor 4 (KLF4) as being markedly induced in response to IFN-γ, LPS, or tumor necrosis factor-α and decreased by TGF-β1 in macrophages. Overexpression of KLF4 in J774a macrophages induced the macrophage activation marker inducible nitric-oxide synthase and inhibited the TGF-β1 and Smad3 target gene plasminogen activator inhibitor-1 (PAl-1). Conversely, KLF4 knockdown markedly attenuated the ability of IFN-γ, LPS, or IFN-γ plus LPS to induce the iNOS promoter, whereas it augmented macrophage responsiveness to TGF-β1 and Smad3 signaling. The KLF4 induction of the iNOS promoter is mediated by two KLF DNA-binding sites at -95 and -212 bp, and mutation of these sites diminished induction by IFN-γ and LPS. We further provide evidence that KLF4 interacts with the NF-κB family member p65 (RelA) to cooperatively induce the iNOS promoter. In contrast, KLF4 inhibited the TGF-β1/Smad3 induction of the PAl-1 promoter independent of KLF4 DNA binding through a novel antagonistic competition with Smad3 for the C terminus of the coactivator p300/CBP. These findings support an important role for KLF4 as a regulator of key signaling pathways that control macrophage activation. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.