1
|
Piazzi M, Bavelloni A, Cenni V, Faenza I, Blalock WL. Revisiting the Role of GSK3, A Modulator of Innate Immunity, in Idiopathic Inclusion Body Myositis. Cells 2021; 10:cells10113255. [PMID: 34831477 PMCID: PMC8625526 DOI: 10.3390/cells10113255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022] Open
Abstract
Idiopathic or sporadic inclusion body myositis (IBM) is the leading age-related (onset >50 years of age) autoimmune muscular pathology, resulting in significant debilitation in affected individuals. Once viewed as primarily a degenerative disorder, it is now evident that much like several other neuro-muscular degenerative disorders, IBM has a major autoinflammatory component resulting in chronic inflammation-induced muscle destruction. Thus, IBM is now considered primarily an inflammatory pathology. To date, there is no effective treatment for sporadic inclusion body myositis, and little is understood about the pathology at the molecular level, which would offer the best hopes of at least slowing down the degenerative process. Among the previously examined potential molecular players in IBM is glycogen synthase kinase (GSK)-3, whose role in promoting TAU phosphorylation and inclusion bodies in Alzheimer’s disease is well known. This review looks to re-examine the role of GSK3 in IBM, not strictly as a promoter of TAU and Abeta inclusions, but as a novel player in the innate immune system, discussing some of the recent roles discovered for this well-studied kinase in inflammatory-mediated pathology.
Collapse
Affiliation(s)
- Manuela Piazzi
- “Luigi Luca Cavalli-Sforza” Istituto di Genetica Molecolare-Consiglio Nazionale delle Ricerche (IGM-CNR), 40136 Bologna, Italy; (M.P.); (V.C.)
- IRCCS, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Alberto Bavelloni
- Laboratorio di Oncologia Sperimentale, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Vittoria Cenni
- “Luigi Luca Cavalli-Sforza” Istituto di Genetica Molecolare-Consiglio Nazionale delle Ricerche (IGM-CNR), 40136 Bologna, Italy; (M.P.); (V.C.)
- IRCCS, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Irene Faenza
- Dipartimento di Scienze Biomediche and Neuromotorie, Università di Bologna, 40136 Bologna, Italy;
| | - William L. Blalock
- “Luigi Luca Cavalli-Sforza” Istituto di Genetica Molecolare-Consiglio Nazionale delle Ricerche (IGM-CNR), 40136 Bologna, Italy; (M.P.); (V.C.)
- IRCCS, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Correspondence:
| |
Collapse
|
2
|
Wang H, Wang Z, Wei C, Wang J, Xu Y, Bai G, Yao Q, Zhang L, Chen Y. Anticancer potential of indirubins in medicinal chemistry: Biological activity, structural modification, and structure-activity relationship. Eur J Med Chem 2021; 223:113652. [PMID: 34161865 DOI: 10.1016/j.ejmech.2021.113652] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/13/2021] [Accepted: 06/13/2021] [Indexed: 10/21/2022]
Abstract
Indirubin is the crucial ingredient of Danggui Longhui Wan and Qing-Dai, traditional Chinese medicine herbal formulas used for the therapy of chronic myelocytic leukemia in China for hundreds of years. Although the monomeric indirubin has been used in China for the treatment human chronic myelocytic leukemia. However, due to low water solubility, poor pharmacokinetic properties and low therapeutic effects are the major obstacle, and had significantly limited its clinical application. Consequently, the attractive anticancer profile of indirubin has enthused numerous researchers to discover novel indirubin derivatives with improved pharmacodynamic activity as well as good pharmacokinetic property. In this paper, we comprehensively review the recent progress of anticancer potential of indirubins, structural modification and structure-activity relationship, which may provide useful direction for the further development of novel indirubins with improved pharmacological profiles for the treatment of various types of cancer.
Collapse
Affiliation(s)
- Hezhen Wang
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China
| | - Zhiyuan Wang
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China
| | - Chunyong Wei
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China
| | - Jing Wang
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China
| | - Yingshu Xu
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China
| | - Guohui Bai
- Key Laboratory of Oral Disease of Higher Schools in Guizhou Province, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China.
| | - Qizheng Yao
- School of Pharmacy, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing, 210009, PR China.
| | - Lei Zhang
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China.
| | - Yongzheng Chen
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China.
| |
Collapse
|
3
|
Small Molecules in the Treatment of Squamous Cell Carcinomas: Focus on Indirubins. Cancers (Basel) 2021; 13:cancers13081770. [PMID: 33917267 PMCID: PMC8068014 DOI: 10.3390/cancers13081770] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/23/2021] [Accepted: 03/28/2021] [Indexed: 02/08/2023] Open
Abstract
Simple Summary In this review, the genetic landscape of squamous cell carcinoma is related to the potential targets of indirubin-based small molecules in cancer therapy. Being a component of traditional Chinese medicine, indirubins are used to treat chronic or inflammatory diseases, and have received increasing attention in cancer treatment due to their proapoptotic and antiproliferative activity. Frequent genetic alterations of squamous cell carcinomas are summarized, and it is discussed how these may render tumors susceptible to indirubin-based small molecule inhibitors. Abstract Skin cancers are the most common malignancies in the world. Among the most frequent skin cancer entities, squamous cell carcinoma (SCC) ranks second (~20%) after basal cell carcinoma (~77%). In early stages, a complete surgical removal of the affected tissue is carried out as standard therapy. To treat advanced and metastatic cancers, targeted therapies with small molecule inhibitors are gaining increasing attention. Small molecules are a heterogeneous group of protein regulators, which are produced by chemical synthesis or fermentation. The majority of them belong to the group of receptor tyrosine kinase inhibitors (RTKIs), which specifically bind to certain RTKs and directly influence the respective signaling pathway. Knowledge of characteristic molecular alterations in certain cancer entities, such as SCC, can help identify tumor-specific substances for targeted therapies. Most frequently, altered genes in SCC include TP53, NOTCH, EGFR, and CCND1. For example, the gene CCND1, which codes for cyclin D1 protein, is upregulated in nearly half of SCC cases and promotes proliferation of affected cells. A treatment with the small molecule 5′-nitroindirubin-monoxime (INO) leads to inhibition of cyclin D1 and thus inhibition of proliferation. As a component of Danggui Longhui Wan, a traditional Chinese medicine, indirubins are used to treat chronic diseases and have been shown to inhibit inflammatory reactions. Indirubins are pharmacologically relevant small molecules with proapoptotic and antiproliferative activity. In this review, we discuss the current literature on indirubin-based small molecules in cancer treatment. A special focus is on the molecular biology of squamous cell carcinomas, their alterations, and how these are rendered susceptible to indirubin-based small molecule inhibitors. The potential molecular mechanisms of the efficacy of indirubins in killing SCC cells will be discussed as well.
Collapse
|
4
|
Wei D, Zhu X, Li S, Liu G, Wang Y, Wang W, Zhang Q, Jiang S. Tideglusib suppresses stem-cell-like features and progression of osteosarcoma by inhibiting GSK-3β/NOTCH1 signaling. Biochem Biophys Res Commun 2021; 554:206-213. [PMID: 33813076 DOI: 10.1016/j.bbrc.2020.12.055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 12/15/2020] [Indexed: 12/14/2022]
Abstract
Osteosarcoma is the most common primary bone tumor in children, teenagers and adolescents. Cancer stem cells (CSCs) have the function to self-renew and keep the phenotype of tumor, causing clinical treatment failure. Therefore, developing effective therapies to inhibit osteosarcoma progression is urgently necessary. Glycogen synthase kinase 3β (GSK-3β)is highly expressed in osteosarcoma. In the present study, we made an exploration on the anti-tumor effect of tideglusib (TID), a small-molecule inhibitor of GSK-3β, and revealed the underlying mechanisms. Here, we found that TID markedly reduced the cell viability of different osteosarcoma cell lines. Cell cycle arrest distributed in G2/M was markedly up-regulated in TID-incubated osteosarcoma cells through enhancing p21 expression levels. Apoptosis was evidently induced in osteosarcoma cells via blocking Caspase-3 activation. Consistently, tumor growth was effectively suppressed in an established murine xenograft model with few toxicity and side effects in vivo. Furthermore, TID markedly repressed stem-cell-like activity in osteosarcoma cells through down-regulating NOTCH1 expression. Notably, rescuing NOTCH1 significantly abolished the role of TID in reducing cell proliferation and sarcosphere-formation. Mechanistically, we found that TID-inhibited NOTCH1 expression was associated with the blockage of AKT/GSK-3β signaling pathway. In summary, we for the first time provided evidence that TID could effectively inhibit osteosarcoma progression through repressing cell proliferation, inducing apoptosis, suppressing stem-cell-like properties via down-regulating AKT/GSK-3β/NOTCH1 signaling pathway. Thus, TID may be a promising therapeutic strategy for osteosarcoma treatment without side effects.
Collapse
Affiliation(s)
- Dandan Wei
- School of the First Clinical Medical, Henan University of Chinese Medicine, Longzihu University Park, Zhengdong New District, 156 Jinshui East Road, Zhengzhou, 450000, China
| | - Xinghao Zhu
- School of the First Clinical Medical, Henan University of Chinese Medicine, Longzihu University Park, Zhengdong New District, 156 Jinshui East Road, Zhengzhou, 450000, China
| | - Shanshan Li
- School of the First Clinical Medical, Henan University of Chinese Medicine, Longzihu University Park, Zhengdong New District, 156 Jinshui East Road, Zhengzhou, 450000, China
| | - Guangyao Liu
- Biomedical Research and Development Center, Jilin Institute of Biomedicine Ltd.Co, Changchun, 130033, China
| | - Yongkun Wang
- Biomedical Research and Development Center, Jilin Institute of Biomedicine Ltd.Co, Changchun, 130033, China
| | - Wei Wang
- Biomedical Research and Development Center, Jilin Institute of Biomedicine Ltd.Co, Changchun, 130033, China
| | - Qiao Zhang
- Biomedical Research and Development Center, Jilin Institute of Biomedicine Ltd.Co, Changchun, 130033, China
| | - Shiqing Jiang
- Department of Oncology, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, 450000, China.
| |
Collapse
|
5
|
Arshad S, Naveed M, Ullia M, Javed K, Butt A, Khawar M, Amjad F. Targeting STAT-3 signaling pathway in cancer for development of novel drugs: Advancements and challenges. Genet Mol Biol 2020; 43:e20180160. [PMID: 32167126 PMCID: PMC7198026 DOI: 10.1590/1678-4685-gmb-2018-0160] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 10/20/2018] [Indexed: 12/25/2022] Open
Abstract
Signal transducers and activators of transcription 3 (STAT-3) is a transcription
factor that regulates the gene expression of several target genes. These factors
are activated by the binding of cytokines and growth factors with STAT-3
specific receptors on cell membrane. Few years ago, STAT-3 was considered an
acute phase response element having several cellular functions such as
inflammation, cell survival, invasion, metastasis and proliferation, genetic
alteration, and angiogenesis. STAT-3 is activated by several types of
inflammatory cytokines, carcinogens, viruses, growth factors, and oncogenes.
Thus, the STAT3 pathway is a potential target for cancer therapeutics. Abnormal
STAT-3 activity in tumor development and cellular transformation can be targeted
by several genomic and pharmacological methodologies. An extensive review of the
literature has been conducted to emphasize the role of STAT-3 as a unique cancer
drug target. This review article discusses in detail the wide range of STAT-3
inhibitors that show antitumor effects both in vitro and
in vivo. Thus, targeting constitutive STAT-3 signaling is a
remarkable therapeutic methodology for tumor progression. Finally, current
limitations, trials and future perspectives of STAT-3 inhibitors are also
critically discussed.
Collapse
Affiliation(s)
- Sundas Arshad
- University of Lahore, Department of Allied Health Sciences, Gujrat Campus, Pakistan
| | - Muhammad Naveed
- University of Central Punjab, Faculty of life sciences, Department of Biotechnology, Lahore, Pakistan
| | - Mahad Ullia
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Khadija Javed
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Ayesha Butt
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Masooma Khawar
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Fazeeha Amjad
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| |
Collapse
|
6
|
Zhang Y, Du H, Liu H, He Q, Xu Z. Isatin dimers and their biological activities. Arch Pharm (Weinheim) 2020; 353:e1900299. [DOI: 10.1002/ardp.201900299] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/20/2019] [Accepted: 12/28/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Ya‐Zhou Zhang
- Teaching and Research Office of Analytical Chemistry, School of Pharmaceutical SciencesGuizhou University of Traditional Chinese Medicine Guizhou China
| | - Hong‐Zhi Du
- Teaching and Research Office of Analytical Chemistry, School of Pharmaceutical SciencesGuizhou University of Traditional Chinese Medicine Guizhou China
| | - Hai‐Lin Liu
- Teaching and Research Office of Analytical Chemistry, School of Pharmaceutical SciencesGuizhou University of Traditional Chinese Medicine Guizhou China
| | - Qian‐Song He
- Teaching and Research Office of Analytical Chemistry, School of Pharmaceutical SciencesGuizhou University of Traditional Chinese Medicine Guizhou China
| | - Zhi Xu
- Teaching and Research Office of Analytical Chemistry, School of Pharmaceutical SciencesGuizhou University of Traditional Chinese Medicine Guizhou China
| |
Collapse
|
7
|
Otten C, Knox J, Boulday G, Eymery M, Haniszewski M, Neuenschwander M, Radetzki S, Vogt I, Hähn K, De Luca C, Cardoso C, Hamad S, Igual Gil C, Roy P, Albiges-Rizo C, Faurobert E, von Kries JP, Campillos M, Tournier-Lasserve E, Derry WB, Abdelilah-Seyfried S. Systematic pharmacological screens uncover novel pathways involved in cerebral cavernous malformations. EMBO Mol Med 2019; 10:emmm.201809155. [PMID: 30181117 PMCID: PMC6180302 DOI: 10.15252/emmm.201809155] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cerebral cavernous malformations (CCMs) are vascular lesions in the central nervous system causing strokes and seizures which currently can only be treated through neurosurgery. The disease arises through changes in the regulatory networks of endothelial cells that must be comprehensively understood to develop alternative, non-invasive pharmacological therapies. Here, we present the results of several unbiased small-molecule suppression screens in which we applied a total of 5,268 unique substances to CCM mutant worm, zebrafish, mouse, or human endothelial cells. We used a systems biology-based target prediction tool to integrate the results with the whole-transcriptome profile of zebrafish CCM2 mutants, revealing signaling pathways relevant to the disease and potential targets for small-molecule-based therapies. We found indirubin-3-monoxime to alleviate the lesion burden in murine preclinical models of CCM2 and CCM3 and suppress the loss-of-CCM phenotypes in human endothelial cells. Our multi-organism-based approach reveals new components of the CCM regulatory network and foreshadows novel small-molecule-based therapeutic applications for suppressing this devastating disease in patients.
Collapse
Affiliation(s)
- Cécile Otten
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany
| | - Jessica Knox
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Gwénola Boulday
- INSERM UMR-1161, Génétique et physiopathologie des maladies cérébro-vasculaires, Université Paris Diderot, Paris, France
| | - Mathias Eymery
- INSERM U1209, Grenoble, France.,Institute for Advanced Biosciences, Université Grenoble Alpes, Grenoble, France.,CNRS UMR 5309, Grenoble, France
| | - Marta Haniszewski
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Developmental and Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | | | - Silke Radetzki
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Ingo Vogt
- German Center for Diabetes Research, Neuherberg, Germany.,Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Kristina Hähn
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany
| | - Coralie De Luca
- INSERM UMR-1161, Génétique et physiopathologie des maladies cérébro-vasculaires, Université Paris Diderot, Paris, France
| | - Cécile Cardoso
- INSERM UMR-1161, Génétique et physiopathologie des maladies cérébro-vasculaires, Université Paris Diderot, Paris, France
| | - Sabri Hamad
- German Center for Diabetes Research, Neuherberg, Germany.,Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Carla Igual Gil
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany
| | - Peter Roy
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Corinne Albiges-Rizo
- INSERM U1209, Grenoble, France.,Institute for Advanced Biosciences, Université Grenoble Alpes, Grenoble, France.,CNRS UMR 5309, Grenoble, France
| | - Eva Faurobert
- INSERM U1209, Grenoble, France.,Institute for Advanced Biosciences, Université Grenoble Alpes, Grenoble, France.,CNRS UMR 5309, Grenoble, France
| | - Jens P von Kries
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Mónica Campillos
- German Center for Diabetes Research, Neuherberg, Germany.,Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Elisabeth Tournier-Lasserve
- INSERM UMR-1161, Génétique et physiopathologie des maladies cérébro-vasculaires, Université Paris Diderot, Paris, France.,AP-HP, Groupe hospitalier Saint-Louis, Lariboisière, Fernand-Widal, Service de génétique moléculaire neuro-vasculaire, Paris, France
| | - W Brent Derry
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Developmental and Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany .,Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
8
|
Zhang Y, Song L, Li J, Zhang Y, Lu X, Zhang B. Inhibitory effects of indirubin-3'-monoxime against human osteosarcoma. IUBMB Life 2019; 71:1465-1474. [PMID: 31050877 DOI: 10.1002/iub.2058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/17/2019] [Accepted: 04/17/2019] [Indexed: 11/08/2022]
Abstract
Indirubin is widely used as the active component of "Dangui Luhui Wan" in ancient China. However, its effects against the osteosarcoma (OS), the most common primary malignancy, are still unknown. In our present study, we investigated the effects of the Indirubin-3'-monoxime (I3M), a derivative of indirubin with better water solubility, against the OS cells. We found I3M inhibited OS cell proliferation in a dose-dependent manner. Flow cytometry assays showed that I3M could not only induce OS cell apoptosis in a time- and dose-dependent manner but also regulate the cell cycle distribution. Additionally, we demonstrated that several Bcl-2 family members, cyclin-dependent kinases (CDKs) and cyclins contributed to this process. Furthermore, out data verified that I3M suppressed OS cell migration and invasion by decreasing MMP-2 and MMP-9 levels. Moreover, survivin and focal adhesion kinase (FAK) might play important roles in the anti-OS effects of I3M. The administration of I3M also inhibited the OS cell growth in mice. Taken together, our results indicated the inhibitory effects of I3M against human OS and thus might be an efficient candidate for OS chemotherapy.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lu Song
- Department of Gerontology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiazhen Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Zhang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinchang Lu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bin Zhang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
9
|
Nutritional Preconditioning of Apigenin Alleviates Myocardial Ischemia/Reperfusion Injury via the Mitochondrial Pathway Mediated by Notch1/Hes1. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7973098. [PMID: 31015891 PMCID: PMC6446095 DOI: 10.1155/2019/7973098] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/01/2018] [Accepted: 01/27/2019] [Indexed: 01/08/2023]
Abstract
Apigenin (Api), a natural flavone found in high amounts in several herbs, has shown potent cardioprotective effects in clinical studies, although the underlying mechanisms are not clear. We hypothesized that Api protects the myocardium from simulated ischemia/reperfusion (SI/R) injury via nutritional preconditioning (NPC). Rats fed with Api-containing food showed improvement in cardiac functions; lactate dehydrogenase (LDH) and creatine phosphokinase (CPK) activities; infarct size; apoptosis rates; malondialdehyde (MDA) levels; caspase-3, superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and catalase (CAT) activities; and ferric reducing antioxidant power (FRAP) compared to those fed standard chow following SI/R injury. In addition, Api pretreatment significantly improved the viability, decreased the LDH activity and intracellular reactive oxygen species (ROS) generation, alleviated the loss of mitochondrial membrane potential (MMP), prevented the opening of the mitochondrial permeability transition pore (mPTP), and decreased the caspase-3 activity, cytochrome c (Cyt C) release, and apoptosis induced by SI/R in primary cardiomyocytes. Mechanistically, Api upregulated Hes1 expression and was functionally neutralized by the Notch1 γ-secretase inhibitor GSI, as well as the mPTP opener atractyloside (Atr). Taken together, Api protected the myocardium against SI/R injury via the mitochondrial pathway mediated by the Notch1/Hes1 signaling pathway.
Collapse
|
10
|
Fogaça MV, Cândido-Bacani PDM, Benicio LM, Zapata LM, Cardoso PDF, de Oliveira MT, Calvo TR, Varanda EA, Vilegas W, de Syllos Cólus IM. Effects of indirubin and isatin on cell viability, mutagenicity, genotoxicity and BAX/ERCC1 gene expression. PHARMACEUTICAL BIOLOGY 2017; 55:2005-2014. [PMID: 28738722 PMCID: PMC7011876 DOI: 10.1080/13880209.2017.1354387] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 07/03/2017] [Accepted: 07/06/2017] [Indexed: 06/07/2023]
Abstract
CONTEXT Indigofera suffruticosa Miller (Fabaceae) and I. truxillensis Kunth produce compounds, such as isatin (ISA) and indirubin (IRN), which possess antitumour properties. Their effects in mammalian cells are still not very well understood. OBJECTIVE We evaluated the activities of ISA and/or IRN on cell viability and apoptosis in vitro, their genotoxic potentials in vitro and in vivo, and the IRN- and ISA-induced expression of ERCC1 or BAX genes. MATERIALS AND METHODS HeLa and/or CHO-K1 cell lines were tested (3 or 24 h) in the MTT, Trypan blue exclusion, acridine orange/ethidium bromide, cytokinesis-blocked micronucleus (CBMN) and comet (36, 24 and 72 h) tests after treatment with IRN (0.1 to 200 μM) or ISA (0.5 to 50 μM). Gene expression was measured by RT-qPCR in HeLa cells. Swiss albino mice received IRN (3, 4 or 24 h) by gavage (50, 100 and 150 mg/kg determined from the LD50 - 1 g/kg b.w.) and submitted to comet assay in vivo. RESULTS IRN reduced the viability of CHO-K1 (24 h; 5 to 200 μM) and HeLa cells (10 to 200 μM), and was antiproliferative in the CBMN test (CHO-K1: 0.5 to 10 μM; HeLa: 5 and 10 μM). The drug did not induce apoptosis, micronucleus neither altered gene expression. IRN and ISA were genotoxic for HeLa cells (3 and 24 h) at all doses tested. IRN (100 and 150 mg/kg) also induced genotoxicity in vivo (4 h). CONCLUSION IRN and ISA have properties that make them candidates as chemotherapeutics for further pharmacological investigations.
Collapse
Affiliation(s)
- Manoela Viar Fogaça
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | | | - Lucas Milanez Benicio
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Lara Martinelli Zapata
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | | | | | - Tamara Regina Calvo
- Araraquara Institute of Chemistry, São Paulo State University, Araraquara, Brazil
| | - Eliana Aparecida Varanda
- Araraquara Faculty of Pharmaceutical Sciences, Department of Biological Sciences, São Paulo State University, Araraquara, Brazil
| | - Wagner Vilegas
- Araraquara Institute of Chemistry, São Paulo State University, Araraquara, Brazil
- Experimental Campus of the Paulista Coast, São Paulo State University, São Vicente, Brazil
| | - Ilce Mara de Syllos Cólus
- Department of General Biology, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| |
Collapse
|
11
|
Zhou A, Yan L, Lai F, Chen X, Goto M, Lee KH, Xiao Z. Design, synthesis and biological evaluation of novel indolin-2-ones as potent anticancer compounds. Bioorg Med Chem Lett 2017. [PMID: 28625363 DOI: 10.1016/j.bmcl.2017.06.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The indolin-2-one core is a privileged structure for antitumor agents, especially kinase inhibitors. Twenty-three novel indolin-2-ones were designed by molecular dissection of the anticancer drug indirubin. Seventeen of them exhibited significant inhibition against the tested cell lines, and two of them (1c and 1h) showed IC50 values at the submicromolar level against HCT-116 cells. Compounds 1c and 2c were also potent inhibitors of the triple-negative breast cancer (TNBC) cell line MDA-MB-231. Flow cytometry was utilized to explore the antitumor mechanism of 1c and 2c with MDA-MB-231 cells, and distinct effects were observed on 2c. Furthermore, immunocytochemical examination of 1c suggested a destabilization of microtubules, which was significantly different from the effect of IM, an indirubin derivative.
Collapse
Affiliation(s)
- Andong Zhou
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lei Yan
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Fangfang Lai
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiaoguang Chen
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Masuo Goto
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599-7568, USA
| | - Kuo-Hsiung Lee
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599-7568, USA; Chinese Medicine Research and Development Center, China Medical University and Hospital, Taichung, Taiwan
| | - Zhiyan Xiao
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
12
|
Indirubin Treatment of Lipopolysaccharide-Induced Mastitis in a Mouse Model and Activity in Mouse Mammary Epithelial Cells. Mediators Inflamm 2017; 2017:3082805. [PMID: 28255203 PMCID: PMC5309412 DOI: 10.1155/2017/3082805] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/13/2016] [Accepted: 12/27/2016] [Indexed: 02/07/2023] Open
Abstract
Indirubin is a Chinese medicine extracted from indigo and known to be effective for treating chronic myelogenous leukemia, neoplasia, and inflammatory disease. This study evaluated the in vivo anti-inflammatory activity of indirubin in a lipopolysaccharide- (LPS-) induced mouse mastitis model. The indirubin mechanism and targets were evaluated in vitro in mouse mammary epithelial cells. In the mouse model, indirubin significantly attenuated the severity of inflammatory lesions, edema, inflammatory hyperemia, milk stasis and local tissue necrosis, and neutrophil infiltration. Indirubin significantly decreased myeloperoxidase activity and downregulated the production of tumor necrosis factor-α, interleukin-1β (IL-1β), and IL-6 caused by LPS. In vitro, indirubin inhibited LPS-stimulated expression of proinflammatory cytokines in a dose-dependent manner. It also downregulated LPS-induced toll-like receptor 4 (TLR4) expression and inhibited phosphorylation of LPS-induced nuclear transcription factor-kappa B (NF-κB) P65 protein and inhibitor of kappa B. In addition to its effect on the NF-κB signaling pathway, indirubin suppressed the mitogen-activated protein kinase (MAPK) signaling by inhibiting phosphorylation of extracellular signal-regulated kinase (ERK), P38, and c-jun NH2-terminal kinase (JNK). Indirubin improved LPS-induced mouse mastitis by suppressing TLR4 and downstream NF-κB and MAPK pathway inflammatory signals and might be a potential treatment of mastitis and other inflammatory diseases.
Collapse
|
13
|
Huang H, Lai S, Wan Q, Qi W, Liu J. Astragaloside IV protects cardiomyocytes from anoxia/reoxygenation injury by upregulating the expression of Hes1 protein. Can J Physiol Pharmacol 2015; 94:542-53. [PMID: 27070866 DOI: 10.1139/cjpp-2015-0457] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Astragaloside IV (ASI), a traditional Chinese medicine, is a main active ingredient of Astragalus membranaceus. Many clinical studies have found that ASI protects cardiomyocytes in cardiovascular diseases, but the underlying mechanisms remain obscure. The aim of this study was to investigate the molecular mechanisms responsible for the protective effects of ASI in cardiomyocytes from anoxia/reoxygenation (A/R) injury. According to the previous studies, we hypothesized that the cardioprotective effects of ASI against A/R injury might be associated with Notch1/Hes1 signaling pathway. In this study, neonatal rat primary cardiomyocytes were preconditioned with ASI prior to A/R injury. Our results showed that ASI effectively increased the cell viability, decreased the content of MDA, decreased the activities of CPK and LDH, increased the activities of GSH-Px and SOD, and reduced the reactive oxygen species (ROS) generation and the loss of mitochondrial membrane potential (Δψm). ASI inhibited the mitochondrial permeability transition pore (mPTP) opening and activation of caspase-3, and finally decreased the cell apoptosis in cardiomyocytes. Furthermore, ASI upregulated Hes1 protein expression. However, pretreatment with DAPT, a Notch1 inhibitor, effectively attenuated the cardioprotective effects of ASI against A/R injury, except MDA, SOD, GSH-Px, and the ROS generation. Taken together, we demonstrated that ASI could protect against A/R injury via the Notch1/Hes1 signaling pathway.
Collapse
Affiliation(s)
- Huang Huang
- a Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, No. 17, Yong Wai Zheng Street, Nanchang, Jiangxi 330006, P.R. China
| | - Songqing Lai
- a Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, No. 17, Yong Wai Zheng Street, Nanchang, Jiangxi 330006, P.R. China
| | - Qing Wan
- b Department of Pharmacy, The First Affiliated Hospital of Nanchang University, No. 17, Yong Wai Zheng Street, Nanchang, Jiangxi 330006, P.R. China
| | - Wanghong Qi
- a Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, No. 17, Yong Wai Zheng Street, Nanchang, Jiangxi 330006, P.R. China
| | - Jichun Liu
- a Department of Cardiac Surgery, The First Affiliated Hospital of Nanchang University, No. 17, Yong Wai Zheng Street, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
14
|
Indirubin and Indirubin Derivatives for Counteracting Proliferative Diseases. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:654098. [PMID: 26457112 PMCID: PMC4589628 DOI: 10.1155/2015/654098] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 08/23/2015] [Accepted: 08/24/2015] [Indexed: 02/08/2023]
Abstract
Indirubin is the active component of Danggui Longhui Wan, a traditional Chinese medicine formulation. The encouraging clinical results from the 1980s obtained in chronic myelocytic leukemia patients treated with indirubin stimulated numerous studies on this compound. These investigations explored the use of indirubin in different types of cancer and reported the synthesis of novel derivatives with improved chemical and pharmacokinetic properties. In this paper, we review the impressive progress that has been made in elucidating the mechanistic understanding of how indirubin and its derivatives affect physiological and pathophysiological processes, mainly by inhibition of cell proliferation and induction of cell death. Furthermore, we survey the therapeutic use of these compounds in combating proliferative diseases such as cancer, restenosis, and psoriasis.
Collapse
|
15
|
Chiou CT, Lee WC, Liao JH, Cheng JJ, Lin LC, Chen CY, Song JS, Wu MH, Shia KS, Li WT. Synthesis and evaluation of 3-ylideneoxindole acetamides as potent anticancer agents. Eur J Med Chem 2015; 98:1-12. [PMID: 25988923 DOI: 10.1016/j.ejmech.2015.04.062] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/28/2015] [Accepted: 04/29/2015] [Indexed: 11/20/2022]
Abstract
Indirubin, an active component in the traditional Chinese medicine formula Danggui Longhui Wan, shows promising anticancer effects. Meisoindigo is an analog derived from indirubin, which is less toxic and appears to be even more potent against cancer. In considering meisoindigo as a structural template for the development of new drugs, we designed and synthesized a series of 3-ylideneoxindole acetamides as novel anticancer agents. The acetamides were then evaluated for in vitro and in vivo anticancer activities. The 3-ylideneoxindole acetamides were found to have better anticancer activity than was indirubin-3'-oxime in several cancer cell lines and also displayed a spectrum of activity similar to that of the drug candidate roscovitine, a CDK inhibitor. Among the 3-ylideneoxindole acetamides, compound 10 showed particularly good efficacy. Cell cycle analysis further revealed that compound 10 arrested cells in the G1 phase and caused an increase in the sub-G1 population, indicating that the apoptosis pathway had been induced. In addition, exposure of cells to compound 10 led to the upregulation of the cell-cycle regulator cyclin D1, which was sustained at a high level. In contrast, the same compound induced a short-term elevation in the level of cyclin E, which was followed by a rapid decrease and the attenuation of Rb phosphorylation. Furthermore, a docking model suggests that compound 10 binds to the active site of CDK4. In testing the therapeutic potency of compound 10 on CT26-xenografted BALB/c mice, a significant reduction in tumor size comparable to that of cisplatin was found when administrated via the i.p. route. The mice presented no loss of body weight, indicating that this compound possesses low toxicity. In the future, we are planning in vivo investigations of these new active anticancer agents to better elucidate active mechanisms at the cellular level and thus benefit the development of anticancer therapies.
Collapse
Affiliation(s)
- Chun-Tang Chiou
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11221, Taiwan
| | - Wei-Chun Lee
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11221, Taiwan
| | - Jiahn-Haur Liao
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Jing-Jy Cheng
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11221, Taiwan
| | - Lie-Chwen Lin
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11221, Taiwan
| | - Chih-Yu Chen
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11221, Taiwan
| | - Jen-Shin Song
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Ming-Hsien Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Kak-Shan Shia
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Wen-Tai Li
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11221, Taiwan.
| |
Collapse
|
16
|
Yan L, Lai F, Chen X, Xiao Z. Discovery of novel indirubin-3'-monoxime derivatives as potent inhibitors against CDK2 and CDK9. Bioorg Med Chem Lett 2015; 25:2447-51. [PMID: 25908517 DOI: 10.1016/j.bmcl.2015.03.066] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/06/2015] [Accepted: 03/24/2015] [Indexed: 10/23/2022]
Abstract
Indirubin-3'-monoxime (IM) is a potent cyclin-dependent kinase (CDK) inhibitor. Twenty novel IM derivatives were prepared to investigate the structure-activity relationships (SAR) of this compound class. Six compounds showed significant inhibition against both CDK2/cyclin E1 and CDK9/cyclin T1. The most potent compound 7t exhibited IC50 values at submicromolar level. Preliminary SAR trends were suggested and cytotoxicity of these compounds was investigated. Molecular docking studies on compounds 7l and 7t provided conducive clues for further structural optimization.
Collapse
Affiliation(s)
- Lei Yan
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Fangfang Lai
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Xiaoguang Chen
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Zhiyan Xiao
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
17
|
Ginzinger W, Mühlgassner G, Arion VB, Jakupec MA, Roller A, Galanski M, Reithofer M, Berger W, Keppler BK. A SAR study of novel antiproliferative ruthenium and osmium complexes with quinoxalinone ligands in human cancer cell lines. J Med Chem 2012; 55:3398-413. [PMID: 22417128 DOI: 10.1021/jm3000906] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A series of ruthenium(II) arene complexes with 3-(1H-benzimidazol-2-yl)-1H-quinoxalin-2-one, bearing pharmacophoric groups of known protein kinase inhibitors, and related benzoxazole and benzothiazole derivatives have been synthesized. In addition, the corresponding osmium complexes of the unsubstituted ligands have also been prepared. The compounds have been characterized by NMR, UV-vis, and IR spectroscopy, ESI mass spectrometry, elemental analysis, and by X-ray crystallography. Antiproliferative activity in three human cancer cell lines (A549, CH1, SW480) was determined by MTT assays, yielding IC(50) values of 6-60 μM for three unsubstituted metal-free ligands, whereas values for the metal complexes vary in a broad range from 0.3 to 140 μM. Complexation with osmium of quinoxalinone derivatives with benzimidazole or benzothiazole results in a more consistent increase in cytotoxicity than complexation with ruthenium. For selected compounds, the capacity to induce apoptosis was confirmed by fluorescence microscopy and flow-cytometric analysis, whereas cell cycle effects are only moderate.
Collapse
Affiliation(s)
- Werner Ginzinger
- University of Vienna, Institute of Inorganic Chemistry, Währinger Strasse 42, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Kim MY, Mo JS, Ann EJ, Yoon JH, Jung J, Choi YH, Kim SM, Kim HY, Ahn JS, Kim H, Kim K, Hoe HS, Park HS. Regulation of Notch1 signaling by the APP intracellular domain facilitates degradation of the Notch1 intracellular domain and RBP-Jk. J Cell Sci 2011; 124:1831-43. [PMID: 21558417 DOI: 10.1242/jcs.076117] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The Notch1 receptor is a crucial controller of cell fate decisions, and is also a key regulator of cell growth and differentiation in a variety of contexts. In this study, we have demonstrated that the APP intracellular domain (AICD) attenuates Notch1 signaling by accelerated degradation of the Notch1 intracellular domain (Notch1-IC) and RBP-Jk, through different degradation pathways. AICD suppresses Notch1 transcriptional activity by the dissociation of the Notch1-IC-RBP-Jk complex after processing by γ-secretase. Notch1-IC is capable of forming a trimeric complex with Fbw7 and AICD, and AICD enhances the protein degradation of Notch1-IC through an Fbw7-dependent proteasomal pathway. AICD downregulates the levels of RBP-Jk protein through the lysosomal pathway. AICD-mediated degradation is involved in the preferential degradation of non-phosphorylated RBP-Jk. Collectively, our results demonstrate that AICD functions as a negative regulator in Notch1 signaling through the promotion of Notch1-IC and RBP-Jk protein degradation.
Collapse
Affiliation(s)
- Mi-Yeon Kim
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Choi SJ, Lee JE, Jeong SY, Im I, Lee SD, Lee EJ, Lee SK, Kwon SM, Ahn SG, Yoon JH, Han SY, Kim JI, Kim YC. 5,5'-substituted indirubin-3'-oxime derivatives as potent cyclin-dependent kinase inhibitors with anticancer activity. J Med Chem 2010; 53:3696-706. [PMID: 20361800 DOI: 10.1021/jm100080z] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
To enhance the ability of indirubin derivatives to inhibit CDK2/cyclin E, a target of anticancer agents, we designed and synthesized a new series of indirubin-3'-oxime derivatives with combined substitutions at the 5 and 5' positions. A molecular docking study predicted the binding of derivatives with OH or halogen substitutions at the 5' position to the ATP binding site of CDK2, revealing the critical interactions that may explain the improved CDK2 inhibitory activity of these derivatives. Among the synthesized derivatives, the 5-nitro-5'-hydroxy analogue 3a and the 5-nitro-5'-fluoro analogue 5a displayed potent inhibitory activity against CDK2, with IC(50) values of 1.9 and 1.7 nM, respectively. These derivatives also showed antiproliferative activity against several human cancer cell lines, with IC(50) values of 0.2-3.3 microM. A representative analogue, 3a, showed greater than 500-fold selectivity for CDK relative to selected kinase panel and potent in vivo anticancer activity.
Collapse
Affiliation(s)
- Soo-Jeong Choi
- Department of Life Science, Gwangju Institute of Science and Technology, Gwangju 500-712, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Han SY, Ahn JH, Shin CY, Choi SU. Effects of indirubin derivatives on the FLT3 activity and growth of acute myeloid leukemia cell lines. Drug Dev Res 2010. [DOI: 10.1002/ddr.20363] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
21
|
Abstract
BACKGROUND Aberrant activation of the signal transducer and activator of transcription (STAT)3 occurs in many human tumors. Moreover, studies utilizing genetic and pharmacological approaches to modulate constitutive STAT3 activity have provided compelling evidence for the critical role of aberrant STAT3 activity in malignant transformation and tumor progression, and thereby validated STAT3 as a novel cancer drug target. OBJECTIVE This review is intended to be a full coverage of the efforts to develop direct STAT3 inhibitors and will provide a discussion on the inhibitory modalities developed to date. METHODS Review of the literature focused on the modalities and mechanisms that directly target and inhibit the STAT protein or its functions. RESULTS/CONCLUSION While a variety of STAT3 inhibitors have been identified that induce antitumor cell effects in vitro and in vivo, the landscape remains murky. With a few exceptions, most of the STAT3 inhibitors reported to date have not undergone an in vivo efficacy, pharmacology or toxicity testing. Also, there is no evidence, per the published literature of an impending clinical development for the few agents that were reported to exhibit in vivo efficacy. Overall, there is the need for a reassessment of the ongoing strategies to target STAT3 intended not only for refinement, but also for incorporating some new technologies to strengthen our efforts and ensure the success - sooner, rather than later - of identifying suitable anti-STAT3 agents for development into clinically useful anticancer therapeutics.
Collapse
Affiliation(s)
- Peibin Yue
- University of Central Florida College of Medicine, Burnett School of Biomedical Sciences, Biomolecular Science Center, Department of Molecular Biology and Microbiology, 12722 Research Parkway, Orlando, FL 32826, USA
| | | |
Collapse
|
22
|
Present and future drug treatments for chronic kidney diseases: evolving targets in renoprotection. Nat Rev Drug Discov 2008; 7:936-53. [PMID: 18846102 DOI: 10.1038/nrd2685] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
At present, there are no specific cures for most of the acquired chronic kidney diseases, and renal transplantation is limited by organ shortage, therefore present efforts are concentrated on the prevention of progression of renal diseases. There is robust experimental and clinical evidence that progression of chronic nephropathies is multifactorial; however, intraglomerular haemodynamic changes and proteinuria play a key role in this process. With a focus on renoprotection, we first examine more established therapies--such as those that modulate the renin-angiotensin-aldosterone system--that can be used for the treatment of proteinuric renal diseases. We then discuss examples of novel drugs and biologics that might be used to target the inflammatory and profibrotic process, and glomerular injury, highlighting results from recent clinical trials.
Collapse
|