1
|
Cassandri M, Porrazzo A, Pomella S, Noce B, Zwergel C, Aiello FA, Vulcano F, Milazzo L, Camero S, Pajalunga D, Spada M, Manzi V, Gravina GL, Codenotti S, Piccione M, Tomaciello M, Signore M, Barillari G, Marchese C, Fanzani A, De Angelis B, Quintarelli C, Vakoc CR, Chen EY, Megiorni F, Locatelli F, Valente S, Mai A, Rota R, Marampon F. HDAC3 genetic and pharmacologic inhibition radiosensitizes fusion positive rhabdomyosarcoma by promoting DNA double-strand breaks. Cell Death Discov 2024; 10:351. [PMID: 39107280 PMCID: PMC11303816 DOI: 10.1038/s41420-024-02115-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 08/09/2024] Open
Abstract
Radiotherapy (RT) plays a critical role in the management of rhabdomyosarcoma (RMS), the prevalent soft tissue sarcoma in childhood. The high risk PAX3-FOXO1 fusion-positive subtype (FP-RMS) is often resistant to RT. We have recently demonstrated that inhibition of class-I histone deacetylases (HDACs) radiosensitizes FP-RMS both in vitro and in vivo. However, HDAC inhibitors exhibited limited success on solid tumors in human clinical trials, at least in part due to the presence of off-target effects. Hence, identifying specific HDAC isoforms that can be targeted to radiosensitize FP-RMS is imperative. We, here, found that only HDAC3 silencing, among all class-I HDACs screened by siRNA, radiosensitizes FP-RMS cells by inhibiting colony formation. Thus, we dissected the effects of HDAC3 depletion using CRISPR/Cas9-dependent HDAC3 knock-out (KO) in FP-RMS cells, which resulted in Endoplasmatic Reticulum Stress activation, ERK inactivation, PARP1- and caspase-dependent apoptosis and reduced stemness when combined with irradiation compared to single treatments. HDAC3 loss-of-function increased DNA damage in irradiated cells augmenting H2AX phosphorylation and DNA double-strand breaks (DSBs) and counteracting irradiation-dependent activation of ATM and DNA-Pkcs as well as Rad51 protein induction. Moreover, HDAC3 depletion hampers FP-RMS tumor growth in vivo and maximally inhibits the growth of irradiated tumors compared to single approaches. We, then, developed a new HDAC3 inhibitor, MC4448, which showed specific cell anti-tumor effects and mirrors the radiosensitizing effects of HDAC3 depletion in vitro synergizing with ERKs inhibition. Overall, our findings dissect the pro-survival role of HDAC3 in FP-RMS and suggest HDAC3 genetic or pharmacologic inhibition as a new promising strategy to overcome radioresistance in this tumor.
Collapse
Affiliation(s)
- Matteo Cassandri
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Antonella Porrazzo
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Silvia Pomella
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Beatrice Noce
- Department of Drug Chemistry and Technologies, "Sapienza" University of Rome, Rome, Italy
| | - Clemens Zwergel
- Department of Drug Chemistry and Technologies, "Sapienza" University of Rome, Rome, Italy
| | - Francesca Antonella Aiello
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Francesca Vulcano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Luisa Milazzo
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Camero
- Department of Life Sciences, Health and Health Professions, Link Campus University, Rome, Italy
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Deborah Pajalunga
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Massimo Spada
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Valeria Manzi
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Silvia Codenotti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Michela Piccione
- Confocal Microscopy Core Facility, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Miriam Tomaciello
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Michele Signore
- RPPA Unit, Proteomics Area, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanni Barillari
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Cinzia Marchese
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Alessandro Fanzani
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Biagio De Angelis
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Concetta Quintarelli
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Eleanor Y Chen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Francesca Megiorni
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Franco Locatelli
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Rome, Italy
| | - Sergio Valente
- Department of Drug Chemistry and Technologies, "Sapienza" University of Rome, Rome, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, "Sapienza" University of Rome, Rome, Italy
- Pasteur Institute, Cenci-Bolognetti Foundation, "Sapienza" University of Rome, Rome, Italy
| | - Rossella Rota
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Francesco Marampon
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy.
| |
Collapse
|
2
|
Alsharoh H, Chiroi P, Isachesku E, Tanasa RA, Pop OL, Pirlog R, Berindan-Neagoe I. Personalizing Therapy Outcomes through Mitogen-Activated Protein Kinase Pathway Inhibition in Non-Small Cell Lung Cancer. Biomedicines 2024; 12:1489. [PMID: 39062063 PMCID: PMC11275062 DOI: 10.3390/biomedicines12071489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/18/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Lung cancer (LC) is a highly invasive malignancy and the leading cause of cancer-related deaths, with non-small cell lung cancer (NSCLC) as its most prevalent histological subtype. Despite all breakthroughs achieved in drug development, the prognosis of NSCLC remains poor. The mitogen-activated protein kinase signaling cascade (MAPKC) is a complex network of interacting molecules that can drive oncogenesis, cancer progression, and drug resistance when dysregulated. Over the past decades, MAPKC components have been used to design MAPKC inhibitors (MAPKCIs), which have shown varying efficacy in treating NSCLC. Thus, recent studies support the potential clinical use of MAPKCIs, especially in combination with other therapeutic approaches. This article provides an overview of the MAPKC and its inhibitors in the clinical management of NSCLC. It addresses the gaps in the current literature on different combinations of selective inhibitors while suggesting two particular therapy approaches to be researched in NSCLC: parallel and aggregate targeting of the MAPKC. This work also provides suggestions that could serve as a potential guideline to aid future research in MAPKCIs to optimize clinical outcomes in NSCLC.
Collapse
Affiliation(s)
- Hasan Alsharoh
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (H.A.); (P.C.); (E.I.); (I.B.-N.)
| | - Paul Chiroi
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (H.A.); (P.C.); (E.I.); (I.B.-N.)
| | - Ekaterina Isachesku
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (H.A.); (P.C.); (E.I.); (I.B.-N.)
| | | | - Ovidiu-Laurean Pop
- Department of Morphology Sciences, University of Oradea, 410087 Oradea, Romania;
| | - Radu Pirlog
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (H.A.); (P.C.); (E.I.); (I.B.-N.)
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (H.A.); (P.C.); (E.I.); (I.B.-N.)
| |
Collapse
|
3
|
Rahbar Farzam O, Baradaran B, Akbari B, Najafi S, Amini M, Yari A, Dabbaghipour R, Pourabdollah Kaleybar V, Ahdi Khosroshahi S. Improvement of 5-fluorouracil chemosensitivity in colorectal cancer cells by siRNA-mediated silencing of STAT6 oncogene. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:475-484. [PMID: 38419894 PMCID: PMC10897558 DOI: 10.22038/ijbms.2023.74275.16136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/18/2023] [Indexed: 03/02/2024]
Abstract
Objectives Colorectal cancer (CRC) remains a major health concern worldwide due to its high incidence, mortality rate, and resistance to conventional treatments. The discovery of new targets for cancer therapy is essential to improve the survival of CRC patients. Here, this study aims to present a finding that identifies the STAT6 oncogene as a potent therapeutic target for CRC. Materials and Methods HT-29 CRC cells were transfected with STAT6 siRNA and treated with 5-fluorouracil (5-FU) alone and combined. Then, to evaluate cellular proliferation and apoptosis percentage, MTT assay and annexin V/PI staining were carried out, respectively. Moreover, the migration ability of HT-29 cells was followed using a wound-healing assay, and a colony formation assay was performed to explore cell stemness features. Gene expression was quantified via qRT-PCR. Afterward, functional enrichment analysis was used to learn in-depth about the STAT6 co-expressed genes and the pathways to which they belong. Results Our study shows that silencing STAT6 with small interfering RNA (siRNA) enhances the chemosensitivity of CRC cells to 5-FU, a commonly used chemotherapy drug, by inducing apoptosis, reducing proliferation, and inhibiting metastasis. These results suggest that combining 5-FU with STAT6-siRNA could provide a promising strategy for CRC treatment. Conclusion Our study sheds light on the potential of STAT6 as a druggable target for CRC cancers, the findings offer hope for more effective treatments for CRC patients, especially those with advanced stages that are resistant to conventional therapies.
Collapse
Affiliation(s)
- Omid Rahbar Farzam
- Department of Medical Biotechnology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Akbari
- Department of Medical Biotechnology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - AmirHossein Yari
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | |
Collapse
|
4
|
Yu J, Li X, Cao J, Zhu T, Liang S, Du L, Cao M, Wang H, Zhang Y, Zhou Y, Shen B, Feng J, Zhang J, Wang J, Jin J. Components of the JNK-MAPK pathway play distinct roles in hepatocellular carcinoma. J Cancer Res Clin Oncol 2023; 149:17495-17509. [PMID: 37902853 DOI: 10.1007/s00432-023-05473-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/10/2023] [Indexed: 11/01/2023]
Abstract
PURPOSE Mitogen-activated protein kinases (MAPK), specifically the c-Jun N-terminal kinase (JNK)-MAPK subfamily, play a crucial role in the development of various cancers, including hepatocellular carcinoma (HCC). However, the specific roles of JNK1/2 and their upstream regulators, MKK4/7, in HCC carcinogenesis remain unclear. METHODS In this study, we performed differential expression analysis of JNK-MAPK components at both the transcriptome and protein levels using TCGA and HPA databases. We utilized Kaplan-Meier survival plots and receiver operating characteristic (ROC) curve analysis to evaluate the prognostic performance of a risk scoring model based on these components in the TCGA-HCC cohort. Additionally, we conducted immunoblotting, apoptosis analysis with FACS and soft agar assays to investigate the response of JNK-MAPK pathway components to various death stimuli (TRAIL, TNF-α, anisomycin, and etoposide) in HCC cell lines. RESULTS JNK1/2 and MKK7 levels were significantly upregulated in HCC samples compared to paracarcinoma tissues, whereas MKK4 was downregulated. ROC analyses suggested that JNK2 and MKK7 may serve as suitable diagnostic genes for HCC, and high JNK2 expression correlated with significantly poorer overall survival. Knockdown of JNK1 enhanced TRAIL-induced apoptosis in hepatoma cells, while JNK2 knockdown reduced TNF-α/cycloheximide (CHX)-and anisomycin-induced apoptosis. Neither JNK1 nor JNK2 knockdown affected etoposide-induced apoptosis. Furthermore, MKK7 knockdown augmented TNF-α/CHX- and TRAIL-induced apoptosis and inhibited colony formation in hepatoma cells. CONCLUSION Targeting MKK7, rather than JNK1/2 or MKK4, may be a promising therapeutic strategy to inhibit the JNK-MAPK pathway in HCC therapy.
Collapse
Affiliation(s)
- Jijun Yu
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Xinying Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Junxia Cao
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Ting Zhu
- Beijing No. 80 High School, Beijing, 100102, China
| | - Shuifeng Liang
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China
| | - Le Du
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China
| | - Meng Cao
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China
| | - Haitao Wang
- Department of Hematology, The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100071, China
| | - Yaolin Zhang
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yinxi Zhou
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China
| | - Beifen Shen
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Jiannan Feng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Jiyan Zhang
- Department of Molecular Immunology, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
| | - Jing Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Jianfeng Jin
- School of Basic Medicine, Hainan Medical University, Haikou, 571199, China.
| |
Collapse
|
5
|
Dong Y, Chen Y, Ma G, Cao H. The role of E3 ubiquitin ligases in bone homeostasis and related diseases. Acta Pharm Sin B 2023; 13:3963-3987. [PMID: 37799379 PMCID: PMC10547920 DOI: 10.1016/j.apsb.2023.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 10/07/2023] Open
Abstract
The ubiquitin-proteasome system (UPS) dedicates to degrade intracellular proteins to modulate demic homeostasis and functions of organisms. These enzymatic cascades mark and modifies target proteins diversly through covalently binding ubiquitin molecules. In the UPS, E3 ubiquitin ligases are the crucial constituents by the advantage of recognizing and presenting proteins to proteasomes for proteolysis. As the major regulators of protein homeostasis, E3 ligases are indispensable to proper cell manners in diverse systems, and they are well described in physiological bone growth and bone metabolism. Pathologically, classic bone-related diseases such as metabolic bone diseases, arthritis, bone neoplasms and bone metastasis of the tumor, etc., were also depicted in a UPS-dependent manner. Therefore, skeletal system is versatilely regulated by UPS and it is worthy to summarize the underlying mechanism. Furthermore, based on the current status of treatment, normal or pathological osteogenesis and tumorigenesis elaborated in this review highlight the clinical significance of UPS research. As a strategy possibly remedies the limitations of UPS treatment, emerging PROTAC was described comprehensively to illustrate its potential in clinical application. Altogether, the purpose of this review aims to provide more evidence for exploiting novel therapeutic strategies based on UPS for bone associated diseases.
Collapse
Affiliation(s)
| | | | - Guixing Ma
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Shenzhen 518055, China
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Shenzhen 518055, China
| |
Collapse
|
6
|
Zhang C, Lu LF, Li ZC, Han KJ, Wang XL, Chen DD, Xiong F, Li XY, Zhou L, Ge F, Li S. Zebrafish MAP2K7 Simultaneously Enhances Host IRF7 Stability and Degrades Spring Viremia of Carp Virus P Protein via Ubiquitination Pathway. J Virol 2023; 97:e0053223. [PMID: 37367226 PMCID: PMC10373533 DOI: 10.1128/jvi.00532-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/11/2023] [Indexed: 06/28/2023] Open
Abstract
During viral infection, host defensive proteins either enhance the host immune response or antagonize viral components directly. In this study, we report on the following two mechanisms employed by zebrafish mitogen-activated protein kinase kinase 7 (MAP2K7) to protect the host during spring viremia of carp virus (SVCV) infection: stabilization of host IRF7 and degradation of SVCV P protein. In vivo, map2k7+/- (map2k7-/- is a lethal mutation) zebrafish showed a higher lethality, more pronounced tissue damage, and more viral proteins in major immune organs than the controls. At the cellular level, overexpression of map2k7 significantly enhanced host cell antiviral capacity, and viral replication and proliferation were significantly suppressed. Additionally, MAP2K7 interacted with the C terminus of IRF7 and stabilized IRF7 by increasing K63-linked polyubiquitination. On the other hand, during MAP2K7 overexpression, SVCV P proteins were significantly decreased. Further analysis demonstrated that SVCV P protein was degraded by the ubiquitin-proteasome pathway, as the attenuation of K63-linked polyubiquitination was mediated by MAP2K7. Furthermore, the deubiquitinase USP7 was indispensable in P protein degradation. These results confirm the dual functions of MAP2K7 during viral infection. IMPORTANCE Normally, during viral infection, host antiviral factors individually modulate the host immune response or antagonize viral components to defense infection. In the present study, we report that zebrafish MAP2K7 plays a crucial positive role in the host antiviral process. According to the weaker antiviral capacity of map2k7+/- zebrafish than that of the control, we find that MAP2K7 reduces host lethality through two pathways, as follows: enhancing K63-linked polyubiquitination to promote host IRF7 stability and attenuating K63-mediated polyubiquitination to degrade the SVCV P protein. These two mechanisms of MAP2K7 reveal a special antiviral response in lower vertebrates.
Collapse
Affiliation(s)
- Can Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Long-Feng Lu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Zhuo-Cong Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ke-Jia Han
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Xue-Li Wang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Dan-Dan Chen
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Feng Xiong
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Xi-Yin Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Li Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Feng Ge
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Shun Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
7
|
Kim D, Orr MJ, Kwong AJ, Deibler KK, Munshi HH, Bridges CS, Chen TJ, Zhang X, Lacorazza HD, Scheidt KA. Rational Design of Highly Potent and Selective Covalent MAP2K7 Inhibitors. ACS Med Chem Lett 2023; 14:606-613. [PMID: 37197477 PMCID: PMC10184151 DOI: 10.1021/acsmedchemlett.3c00029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 04/10/2023] [Indexed: 05/19/2023] Open
Abstract
The mitogen-activated protein kinase signaling cascade is conserved across eukaryotes, where it plays a critical role in the regulation of activities including proliferation, differentiation, and stress responses. This pathway propagates external stimuli through a series of phosphorylation events, which allows external signals to influence metabolic and transcriptional activities. Within the cascade, MEK, or MAP2K, enzymes occupy a molecular crossroads immediately upstream to significant signal divergence and cross-talk. One such kinase, MAP2K7, also known as MEK7 and MKK7, is a protein of great interest in the molecular pathophysiology underlying pediatric T cell acute lymphoblastic leukemia (T-ALL). Herein, we describe the rational design, synthesis, evaluation, and optimization of a novel class of irreversible MAP2K7 inhibitors. With a streamlined one-pot synthesis, favorable in vitro potency and selectivity, and promising cellular activity, this novel class of compounds wields promise as a powerful tool in the study of pediatric T-ALL.
Collapse
Affiliation(s)
- Dalton
R. Kim
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Meghan J. Orr
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Ada J. Kwong
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Kristine K. Deibler
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Hasan H. Munshi
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Cory Seth Bridges
- Department
of Pathology & Immunology, Baylor College
of Medicine, Houston, Texas 77030, United States
| | - Taylor Jie Chen
- Department
of Pathology & Immunology, Baylor College
of Medicine, Houston, Texas 77030, United States
| | - Xiaoyu Zhang
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Chemistry
of Life Processes Institute, Northwestern
University, Evanston, Illinois 60208, United
States
| | - H. Daniel Lacorazza
- Department
of Pathology & Immunology, Baylor College
of Medicine, Houston, Texas 77030, United States
| | - Karl A. Scheidt
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Chemistry
of Life Processes Institute, Northwestern
University, Evanston, Illinois 60208, United
States
- Department
of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| |
Collapse
|
8
|
Culhuac EB, Maggiolino A, Elghandour MMMY, De Palo P, Salem AZM. Antioxidant and Anti-Inflammatory Properties of Phytochemicals Found in the Yucca Genus. Antioxidants (Basel) 2023; 12:574. [PMID: 36978823 PMCID: PMC10044844 DOI: 10.3390/antiox12030574] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
The Yucca genus encompasses about 50 species native to North America. Species within the Yucca genus have been used in traditional medicine to treat pathologies related to inflammation. Despite its historical use and the popular notion of its antioxidant and anti-inflammatory properties, there is a limited amount of research on this genus. To better understand these properties, this work aimed to analyze phytochemical composition through documentary research. This will provide a better understanding of the molecules and the mechanisms of action that confer such antioxidant and anti-inflammatory properties. About 92 phytochemicals present within the genus have reported antioxidant or anti-inflammatory effects. It has been suggested that the antioxidant and anti-inflammatory properties are mainly generated through its free radical scavenging activity, the inhibition of arachidonic acid metabolism, the decrease in TNF-α (Tumor necrosis factor-α), IL-6 (Interleukin-6), iNOS (Inducible nitric oxide synthase), and IL-1β (Interleukin 1β) concentration, the increase of GPx (Glutathione peroxidase), CAT (Catalase), and SOD (Superoxide dismutase) concentration, and the inhibition of the MAPK (Mitogen-Activated Protein Kinase), and NF-κB (Nuclear factor kappa B), and the activation of the Nrf2 (Nuclear factor erythroid 2-related factor) signaling pathway. These studies provide evidence of its use in traditional medicine against pathologies related to inflammation. However, more models and studies are needed to properly understand the activity of most plants within the genus, its potency, and the feasibility of its use to help manage or treat chronic inflammation.
Collapse
Affiliation(s)
- Erick Bahena Culhuac
- Facultad de Ciencias, Universidad Autónoma del Estado de México, Toluca 50000, Estado de México, Mexico
| | - Aristide Maggiolino
- Department of Veterinary Medicine, University of Bari A. Moro, 70010 Valenzano, Italy
| | - Mona M. M. Y. Elghandour
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca 50000, Estado de México, Mexico
| | - Pasquale De Palo
- Department of Veterinary Medicine, University of Bari A. Moro, 70010 Valenzano, Italy
| | - Abdelfattah Z. M. Salem
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca 50000, Estado de México, Mexico
| |
Collapse
|
9
|
Prieto-Fernandez L, Villaronga MDLA, Hermida-Prado F, Hijazi M, Montoro-Jimenez I, Pevida M, Llames S, Rodrigo JP, Cutillas P, Calvo F, Garcia-Pedrero JM, Alvarez-Teijeiro S. Driving role of head and neck cancer cell secretome on the invasion of stromal fibroblasts: Mechanistic insights by phosphoproteomics. Biomed Pharmacother 2023; 158:114176. [PMID: 36916400 DOI: 10.1016/j.biopha.2022.114176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are major players in tumor-stroma communication, and participate in several cancer hallmarks to drive tumor progression and metastatic dissemination. This study investigates the driving effects of tumor-secreted factors on CAF biology, with the ultimate goal of identifying effective therapeutic targets/strategies for head and neck squamous cell carcinomas (HNSCC). METHODS Functionally, conditioned media (CM) from different HNSCC-derived cell lines and normal keratinocytes (Kc) were tested on the growth and invasion of populations of primary CAFs and normal fibroblasts (NFs) using 3D invasion assays in collagen matrices. The changes in MMPs expression were evaluated by RT-qPCR and kinase enrichment was analyzed using mass spectrometry phosphoproteomics. RESULTS Our results consistently demonstrate that HNSCC-secreted factors (but not Kc CM) specifically and robustly promoted pro-invasive properties in both CAFs and NFs, thereby reflecting the plasticity of fibroblast subtypes. Concomitantly, HNSCC-secreted factors massively increased metalloproteinases levels in CAFs and NFs. By contrast, HNSCC CM and Kc CM exhibited comparable growth-promoting effects on stromal fibroblasts. Mechanistically, phosphoproteomic analysis predominantly revealed phosphorylation changes in fibroblasts upon treatment with HNSCC CM, and various promising kinases were identified: MKK7, MKK4, ASK1, RAF1, BRAF, ARAF, COT, PDK1, RSK2 and AKT1. Interestingly, pharmacologic inhibition of RAF1/BRAF using sorafenib emerged as the most effective drug to block tumor-promoted fibroblast invasion without affecting fibroblast viability CONCLUSIONS: Our findings demonstrate that HNSCC-secreted factors specifically fine tune the invasive potential of stromal fibroblasts, thereby generating tumor-driven pro-invasive niches, which in turn to ultimately facilitate cancer cell dissemination. Furthermore, the RAF/BRAF inhibitor sorafenib was identified as a promising candidate to effectively target the onset of pro-invasive clusters of stromal fibroblasts in the HNSCC microenvironment.
Collapse
Affiliation(s)
- Llara Prieto-Fernandez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Maria de Los Angeles Villaronga
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco Hermida-Prado
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Maruan Hijazi
- Cell Signalling & Proteomics Group, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Irene Montoro-Jimenez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Pevida
- Tissue engineering unit, Centro Comunitario Sangre y Tejidos de Asturias (CCST), Oviedo, Spain; Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, Oviedo, Spain; Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Sara Llames
- Tissue engineering unit, Centro Comunitario Sangre y Tejidos de Asturias (CCST), Oviedo, Spain; Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, Oviedo, Spain; Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Juan Pablo Rodrigo
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Pedro Cutillas
- Cell Signalling & Proteomics Group, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Fernando Calvo
- Division of Cancer Biology, The Institute of Cancer Research, London, United Kingdom; Instituto de Biomedicina y Biotecnología de Cantabria (Consejo Superior de Investigaciones Científicas, Universidad de Cantabria), Santander, Spain
| | - Juana Maria Garcia-Pedrero
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| | - Saul Alvarez-Teijeiro
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
10
|
Zhang J, Sayakoummane S, Kim SA, Lee JS, Choung ES, Kim ES, Lee SG, Yum J, Lee BH, Lee S, Kim JH, Cho JY. Hymenocallis littoralis ameliorates inflammatory responses in LPS-stimulated RAW264.7 cells and HCl/EtOH-induced gastric mucosal injury via targeting the MAPK pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 295:115400. [PMID: 35623503 DOI: 10.1016/j.jep.2022.115400] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hymenocallis littoralis (Jacq.) Salisb. Also known as Pancratium littorale Jacq. And Hymenocallis panamensis Lindl., is a medicinal plant from the family Amarylideceae used for emetic and wound healing and has manifested anti-neoplastic, anti-oxidant, and anti-viral properties. AIM OF THE STUDY The aim of this paper is to investigate the anti-inflammatory potential and molecular mechanism of H. littoralis against lipopolysaccharide (LPS)-induced macrophages and in vivo HCl/EtOH-induced gastritis mucosal injury models. MATERIALS AND METHODS The production of pro-inflammatory cytokines and mediators was evaluated by Griess assay, RT-PCR, and real-time PCR. Moreover, the relevant proteins of mitogen-activated protein kinases (MAPKs) including ERK, JNK, p38, c-Jun, and c-Fos were detected using immunoblotting. RESULTS We demonstrated that H. littoralis prominently dampened production of nitric oxide (NO) in LPS-, poly I:C-, or pam3CSK-stimulated RAW264.7 cells; down-regulated the expression levels of interleukin 6 (IL-6) and inducible nitric oxide synthase; and markedly attenuated the luciferase activities of AP-1 reporter promoters. Moreover, H. littoralis administration prominently downregulated c-Fos and c-Jun phosphorylation as well as JNK1, ERK2, and MKK7 overexpression in HEK 293T cells. Furthermore, H. littoralis displayed anti-inflammatory effects in the HCl/EtOH-induced gastritis mice model. CONCLUSIONS Cumulatively, these results demonstrated that H. littoralis exerts eminently anti-inflammatory activities in LPS-stimulated RAW264.7 cells in vitro and in HCl/EtOH-induced gastritis mice models in vivo. These activities could be attributed to its modulatory effects on the MAPK signaling pathway.
Collapse
Affiliation(s)
- Jianmei Zhang
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Sousath Sayakoummane
- Department of Forestry, Ministry of Agriculture and Forestry, Vientiane Capital, P.O. Box 2932, Laos.
| | - Soo Ah Kim
- DanjoungBio Co. Ltd, Wonju, 26303, Republic of Korea.
| | - Jong Sub Lee
- DanjoungBio Co. Ltd, Wonju, 26303, Republic of Korea.
| | - Eui Su Choung
- DanjoungBio Co. Ltd, Wonju, 26303, Republic of Korea.
| | - Eun Sil Kim
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon, 22689, Republic of Korea.
| | - Seung-Gyu Lee
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon, 22689, Republic of Korea.
| | - Jinwhoa Yum
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon, 22689, Republic of Korea.
| | - Byoung-Hee Lee
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon, 22689, Republic of Korea.
| | - Sarah Lee
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon, 22689, Republic of Korea.
| | - Ji Hye Kim
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Jae Youl Cho
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
11
|
Alqahtani MJ, Mostafa SA, Hussein IA, Elhawary S, Mokhtar FA, Albogami S, Tomczyk M, Batiha GES, Negm WA. Metabolic Profiling of Jasminum grandiflorum L. Flowers and Protective Role against Cisplatin-Induced Nephrotoxicity: Network Pharmacology and In Vivo Validation. Metabolites 2022; 12:metabo12090792. [PMID: 36144196 PMCID: PMC9502427 DOI: 10.3390/metabo12090792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 12/02/2022] Open
Abstract
Cisplatin (CP) is a powerful chemotherapeutic agent; however, its therapeutic use is restricted due to its nephrotoxicity. In this work, we profiled the phytoconstituents of Jasminum grandiflorum flower extract (JGF) using LC-MS/MS and explored the possible molecular mechanisms against acute renal failure through pharmacological network analysis. Furthermore, the possible molecular mechanisms of JGF against acute renal failure were verified in an in vivo nephrotoxicity model caused by cisplatin. LC-MS analysis furnished 26 secondary metabolites. Altogether, there were 112 total hit targets for the identified metabolites, among which 55 were potential consensus targets related to nephrotoxicity based on the network pharmacology approach. Upon narrowing the scope to acute renal failure, using the DisGeNET database, only 30 potential targets were determined. The computational pathway analysis illustrated that JGF might inhibit renal failure through PI3K-Akt, MAPK signaling pathway, and EGFR tyrosine kinase inhibitor resistance. This study was confirmed by in vivo experiment in which kidneys were collected for histopathology and gene expression of mitogen-activated protein kinase 4 (MKK4), MKK7, I-CAM 1, IL-6, and TNF receptor-associated factor 2 (TRAF2). The animal-administered cisplatin exhibited a substantial rise in the expression levels of the MMK4, MKK7, I CAM 1, and TRFA2 genes compared to the control group. To summarize, J. grandiflorum could be a potential source for new reno-protective agents. Further experiments are needed to confirm the obtained activities and determine the therapeutic dose and time.
Collapse
Affiliation(s)
- Moneerah J. Alqahtani
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Sally A. Mostafa
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura 35511, Egypt
| | - Ismail A. Hussein
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| | - Seham Elhawary
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Fatma A. Mokhtar
- Department of Pharmacognosy, Faculty of Pharmacy, ALSalam University, Al Gharbiya, Kafr El Zayat 31616, Egypt
| | - Sarah Albogami
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Michał Tomczyk
- Department of Pharmacognosy, Medical University of Białystok, ul. Mickiewicza 2a, 15-230 Białystok, Poland
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt
| | - Walaa A. Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
- Correspondence:
| |
Collapse
|
12
|
Gehrtz P, Marom S, Bührmann M, Hardick J, Kleinbölting S, Shraga A, Dubiella C, Gabizon R, Wiese JN, Müller MP, Cohen G, Babaev I, Shurrush K, Avram L, Resnick E, Barr H, Rauh D, London N. Optimization of Covalent MKK7 Inhibitors via Crude Nanomole-Scale Libraries. J Med Chem 2022; 65:10341-10356. [PMID: 35912476 PMCID: PMC9376956 DOI: 10.1021/acs.jmedchem.1c02206] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
High-throughput nanomole-scale synthesis allows for late-stage functionalization (LSF) of compounds in an efficient and economical manner. Here, we demonstrated that copper-catalyzed azide-alkyne cycloaddition could be used for the LSF of covalent kinase inhibitors at the nanoscale, enabling the synthesis of hundreds of compounds that did not require purification for biological assay screening, thus reducing experimental time drastically. We generated crude libraries of inhibitors for the kinase MKK7, derived from two different parental precursors, and analyzed them via the high-throughput In-Cell Western assay. Select inhibitors were resynthesized, validated via conventional biological and biochemical methods such as western blots and liquid chromatography-mass spectrometry (LC-MS) labeling, and successfully co-crystallized. Two of these compounds showed over 20-fold increased inhibitory activity compared to the parental compound. This study demonstrates that high-throughput LSF of covalent inhibitors at the nanomole-scale level can be an auspicious approach in improving the properties of lead chemical matter.
Collapse
Affiliation(s)
- Paul Gehrtz
- Department of Chemical and Structural Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Shir Marom
- Department of Chemical and Structural Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Mike Bührmann
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, 44227 Dortmund, Germany.,Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), 44227 Dortmund, Germany
| | - Julia Hardick
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, 44227 Dortmund, Germany.,Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), 44227 Dortmund, Germany
| | - Silke Kleinbölting
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, 44227 Dortmund, Germany.,Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), 44227 Dortmund, Germany
| | - Amit Shraga
- Department of Chemical and Structural Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Christian Dubiella
- Department of Chemical and Structural Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Ronen Gabizon
- Department of Chemical and Structural Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Jan N Wiese
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, 44227 Dortmund, Germany.,Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), 44227 Dortmund, Germany
| | - Matthias P Müller
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, 44227 Dortmund, Germany.,Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), 44227 Dortmund, Germany
| | - Galit Cohen
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Ilana Babaev
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Khriesto Shurrush
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Liat Avram
- Department of Chemical Research Support, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Efrat Resnick
- Department of Chemical and Structural Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Haim Barr
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Daniel Rauh
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, 44227 Dortmund, Germany.,Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), 44227 Dortmund, Germany
| | - Nir London
- Department of Chemical and Structural Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| |
Collapse
|
13
|
Xiao J, Lu H, Ma T, Ni X, Chang T, Liu M, Li N, Lu P, Ke C, Tian Q, Zou L, Wang F, Wang W, Zhang L, Yuan P, Liu L, Zhang J, Shi F, Duan Q, Zhu F. Worenine Prevents Solar Ultraviolet–Induced Sunburn by Inhibiting JNK2. Front Pharmacol 2022; 13:881042. [PMID: 35979232 PMCID: PMC9377457 DOI: 10.3389/fphar.2022.881042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Excessive solar ultraviolet (SUV) radiation often causes dermatitis, photoaging, and even skin cancer. In the pathological processes of SUV-induced sunburn, JNK is activated by phosphorylation, and it in turn phosphorylates its downstream transcription factors, such as ATF2 and c-jun. The transcription factors further regulate the expression of pro-inflammatory genes, such as IL-6 and TNF-α, which ultimately leads to dermatitis. Therefore, inhibiting JNK may be a strategy to prevent dermatitis. In this study, we screened for worenine as a potential drug candidate for inhibiting sunburn. We determined that worenine inhibited the JNK-ATF2/c-jun signaling pathway and the secretion of IL-6 and TNF-α in cell culture and in vivo, confirming the role of worenine in inhibiting sunburn. Furthermore, we determined that worenine bound and inhibited JNK2 activity in vitro through the MST, kinase, and in vitro kinase assays. Therefore, worenine might be a promising drug candidate for the prevention and treatment of SUV-induced sunburn.
Collapse
Affiliation(s)
- Juanjuan Xiao
- Cancer Research Institute, The Affiliated Hospital of Guilin Medical University, Guilin, China
- Guangxi Health Commission Key Laboratory of Novel Onco-Kinases in Target Therapy, The Affiliated Hospital of Guilin Medical University, Guilin, China
- *Correspondence: Hui Lu, ; Juanjuan Xiao, ; Qiuhong Duan, ; Feng Zhu, , orcid.org/0000-0003-1172-0102
| | - Hui Lu
- Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Hui Lu, ; Juanjuan Xiao, ; Qiuhong Duan, ; Feng Zhu, , orcid.org/0000-0003-1172-0102
| | - Tengfei Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofang Ni
- Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Teding Chang
- Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Man Liu
- Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nijie Li
- Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peijiang Lu
- Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Changshu Ke
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Tian
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Zou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Yuan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianmin Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Shi
- Department of Dermatology, The General Hospital of Air Force, Beijing, China
| | - Qiuhong Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Hui Lu, ; Juanjuan Xiao, ; Qiuhong Duan, ; Feng Zhu, , orcid.org/0000-0003-1172-0102
| | - Feng Zhu
- Cancer Research Institute, The Affiliated Hospital of Guilin Medical University, Guilin, China
- Guangxi Health Commission Key Laboratory of Novel Onco-Kinases in Target Therapy, The Affiliated Hospital of Guilin Medical University, Guilin, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, The Affiliated Hospital of Guilin Medical University, Guilin, China
- *Correspondence: Hui Lu, ; Juanjuan Xiao, ; Qiuhong Duan, ; Feng Zhu, , orcid.org/0000-0003-1172-0102
| |
Collapse
|
14
|
Hong YH, Aziz N, Park JG, Lee D, Kim JK, Kim SA, Choi W, Lee CY, Lee HP, Huyen Trang HT, Kim HG, Jeon YJ, Kim B, Kim Y, Kim KH, Yoo BC, Han JW, Parameswaran N, Kim JH, Hur H, Cho JY. Running title: EEF1AKMT3/MAP2K7/TP53 axis in gastric cancerThe EEF1AKMT3/MAP2K7/TP53 axis suppresses tumor invasiveness and metastasis in gastric cancer. Cancer Lett 2022; 544:215803. [PMID: 35753528 DOI: 10.1016/j.canlet.2022.215803] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/15/2022] [Accepted: 06/18/2022] [Indexed: 11/29/2022]
Abstract
The importance of methylation in the tumorigenic responses of nonhistone proteins, such as TP53, PTEN, RB1, AKT, and STAT3, has been emphasized in numerous studies. In parallel, the corresponding nonhistone protein methyltransferases have been acknowledged in the pathophysiology of cancer. Thus, this study aimed to explore the pathological role of a nonhistone methyltransferase in gastric cancer (GC), identify nonhistone substrate protein, and understand the underlying mechanism. Interestingly, among the 24 methyltransferases and methyltransferase family 16 (MTF16) proteins, EEF1AKMT3 (METTL21B) expression was prominently lower in GC tissues than in normal adjacent tissues and was associated with a worse prognosis. In addition, EEF1AKMT3-knockdown induced gastric tumor invasiveness and migration. Through gain and loss-of-function studies, mass spectrometry analysis, RNA-seq, and phospho-antibody array, we identified EEF1AKMT3 as a novel tumor-suppressive methyltransferase that catalyzes the monomethylation of MAP2K7 (MKK7) at K296, thereby decreasing the phosphorylation, ubiquitination, and degradation of TP53. Furthermore, EEF1AKMT3, p-MAP2K7, and TP53 protein levels were positively correlated in GC tissues. Collectively, our results delineate the tumor-suppressive function of the EEF1AKMT3/MAP2K7/TP53 signaling axis and suggest the dysregulation of the signaling axis as potential targeted therapy in GC.
Collapse
Affiliation(s)
- Yo Han Hong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nur Aziz
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jae Gwang Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Division of Translational Science, Research Institute, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Dagyeong Lee
- Department of Surgery, Ajou University School of Medicine, And Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, Republic of Korea
| | - Jin Kyeong Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Seung A Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Wooram Choi
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Chae Young Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hwa Pyoung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Ha Thi Huyen Trang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Han Gyung Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Young-Jun Jeon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | | | | | - Kyung-Hee Kim
- Proteomic Analysis Team, Research Institute, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Byong Chul Yoo
- Division of Translational Science, Research Institute, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Jeung-Whan Han
- Research Center for Epigenome Regulation, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Narayana Parameswaran
- Department of Physiology and Division of Pathology, Michigan State University, East Lansing, MI, 48824, USA
| | - Ji Hye Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Hoon Hur
- Department of Surgery, Ajou University School of Medicine, And Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, Republic of Korea.
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
15
|
Yan J, Lai H, Zeng B, Wang L, Xing F, Xiao P. Photoinduced free radical-releasing systems and their anticancer properties. Photochem Photobiol Sci 2022; 21:1405-1417. [PMID: 35553411 DOI: 10.1007/s43630-022-00231-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/11/2022] [Indexed: 11/26/2022]
Abstract
Cancer has been a serious threat and impact on the health and life of human. Phototherapy is considered as a promising therapeutic method to replace the traditional treatment in clinic owing to its noninvasive nature and high efficiency. Photoinitiators have long been used in the field of photopolymerization; however, few studies have been carried out on their potential as anticancer agents under light irradiation. In this study, the effect of a photoinitiator, diphenyl (2, 4, 6-trimethylbenzoyl) phosphine oxide (TPO), on breast cancer is investigated and the related mechanism is elucidated. It is found that TPO has low dark toxicity and significant phototoxicity. TPO can inhibit cell growth and development and promote cell apoptosis through a mitochondrial pathway under light irradiation. Further studies show that cell apoptosis is induced by free radicals produced from the photolysis of TPO to activate JNK phosphorylation. Overall, we identify the antitumor effects of TPO in vitro for the first time, and provides a proof of concept for its application as a novel photolatent therapeutic drug.
Collapse
Affiliation(s)
- Jieyu Yan
- Department of Immunobiology, College of Life Science and Technology, Jinan University, #601 Huangpu West Avenue, Guangzhou, 510632, China
| | - Haiwang Lai
- Department of Immunobiology, College of Life Science and Technology, Jinan University, #601 Huangpu West Avenue, Guangzhou, 510632, China.
| | - Boning Zeng
- Department of Immunobiology, College of Life Science and Technology, Jinan University, #601 Huangpu West Avenue, Guangzhou, 510632, China
| | - Liqing Wang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, #601 Huangpu West Avenue, Guangzhou, 510632, China
| | - Feiyue Xing
- Department of Immunobiology, College of Life Science and Technology, Jinan University, #601 Huangpu West Avenue, Guangzhou, 510632, China.
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China.
| | - Pu Xiao
- Research School of Chemistry, The Australian National University, Canberra, ACT, 2601, Australia.
| |
Collapse
|
16
|
Park HB, Baek KH. E3 ligases and deubiquitinating enzymes regulating the MAPK signaling pathway in cancers. Biochim Biophys Acta Rev Cancer 2022; 1877:188736. [DOI: 10.1016/j.bbcan.2022.188736] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/30/2022] [Accepted: 05/11/2022] [Indexed: 12/13/2022]
|
17
|
Gehi BR, Gadhave K, Uversky VN, Giri R. Intrinsic disorder in proteins associated with oxidative stress-induced JNK signaling. Cell Mol Life Sci 2022; 79:202. [PMID: 35325330 PMCID: PMC11073203 DOI: 10.1007/s00018-022-04230-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 01/02/2023]
Abstract
The c-Jun N-terminal kinase (JNK) signaling cascade is a mitogen-activated protein kinase (MAPK) signaling pathway that can be activated in response to a wide range of environmental stimuli. Based on the type, degree, and duration of the stimulus, the JNK signaling cascade dictates the fate of the cell by influencing gene expression through its substrate transcription factors. Oxidative stress is a result of a disturbance in the pro-oxidant/antioxidant homeostasis of the cell and is associated with a large number of diseases, such as neurodegenerative disorders, cancer, diabetes, cardiovascular diseases, and disorders of the immune system, where it activates the JNK signaling pathway. Among different biological roles ascribed to the intrinsically disordered proteins (IDPs) and hybrid proteins containing ordered domains and intrinsically disordered protein regions (IDPRs) are signaling hub functions, as intrinsic disorder allows proteins to undertake multiple interactions, each with a different consequence. In order to ensure precise signaling, the cellular abundance of IDPs is highly regulated, and mutations or changes in abundance of IDPs/IDPRs are often associated with disease. In this study, we have used a combination of six disorder predictors to evaluate the presence of intrinsic disorder in proteins of the oxidative stress-induced JNK signaling cascade, and as per our findings, none of the 18 proteins involved in this pathway are ordered. The highest level of intrinsic disorder was observed in the scaffold proteins, JIP1, JIP2, JIP3; dual specificity phosphatases, MKP5, MKP7; 14-3-3ζ and transcription factor c-Jun. The MAP3Ks, MAP2Ks, MAPKs, TRAFs, and thioredoxin were the proteins that were predicted to be moderately disordered. Furthermore, to characterize the predicted IDPs/IDPRs in the proteins of the JNK signaling cascade, we identified the molecular recognition features (MoRFs), posttranslational modification (PTM) sites, and short linear motifs (SLiMs) associated with the disordered regions. These findings will serve as a foundation for experimental characterization of disordered regions in these proteins, which represents a crucial step for a better understanding of the roles of IDPRs in diseases associated with this important pathway.
Collapse
Affiliation(s)
- Bhuvaneshwari R Gehi
- School of Basic Sciences, Indian Institute of Technology Mandi, VPO Kamand, Mandi, Himachal Pradesh, 175005, India
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, 560012, India
| | - Kundlik Gadhave
- School of Basic Sciences, Indian Institute of Technology Mandi, VPO Kamand, Mandi, Himachal Pradesh, 175005, India
| | - Vladimir N Uversky
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow region, 142290, Russia.
| | - Rajanish Giri
- School of Basic Sciences, Indian Institute of Technology Mandi, VPO Kamand, Mandi, Himachal Pradesh, 175005, India.
| |
Collapse
|
18
|
Miller KJ, Asim M. Unravelling the Role of Kinases That Underpin Androgen Signalling in Prostate Cancer. Cells 2022; 11:cells11060952. [PMID: 35326402 PMCID: PMC8946764 DOI: 10.3390/cells11060952] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023] Open
Abstract
The androgen receptor (AR) signalling pathway is the key driver in most prostate cancers (PCa), and is underpinned by several kinases both upstream and downstream of the AR. Many popular therapies for PCa that target the AR directly, however, have been circumvented by AR mutation, such as androgen receptor variants. Some upstream kinases promote AR signalling, including those which phosphorylate the AR and others that are AR-regulated, and androgen regulated kinase that can also form feed-forward activation circuits to promotes AR function. All of these kinases represent potentially druggable targets for PCa. There has generally been a divide in reviews reporting on pathways upstream of the AR and those reporting on AR-regulated genes despite the overlap that constitutes the promotion of AR signalling and PCa progression. In this review, we aim to elucidate which kinases—both upstream and AR-regulated—may be therapeutic targets and require future investigation and ongoing trials in developing kinase inhibitors for PCa.
Collapse
|
19
|
Caliz AD, Vertii A, Fisch V, Yoon S, Yoo HJ, Keaney JF, Kant S. Mitogen-activated protein kinase kinase 7 in inflammatory, cancer, and neurological diseases. Front Cell Dev Biol 2022; 10:979673. [PMID: 36340039 PMCID: PMC9630596 DOI: 10.3389/fcell.2022.979673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
Stress-activated mitogen-activated protein kinase kinase 7 (MKK7) is a member of the dual-specificity mitogen-activated protein kinase family. In the human body, MKK7 controls essential physiological processes, including but not limited to proliferation and differentiation in multiple tissues and organs. MKK7, along with the MKK4 pathway, has been implicated in stress-activated activities and biological events that are mediated by c-Jun N-terminal kinase (JNK) signaling. Although numerous studies have been performed to identify the role of JNK in multiple biological processes, there are limited publications that focus on dissecting the independent role of MKK7. Recent research findings have spurred testing via in vivo genetically deficient models, uncovering previously undocumented JNK-independent functions of MKK7. Here we discuss both JNK-dependent and-independent functions of MKK7 in vivo. This review summarizes the role of MKK7 in inflammation, cytokine production, cancer, and neurological diseases.
Collapse
Affiliation(s)
- Amada D Caliz
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Anastassiia Vertii
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Vijay Fisch
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Soonsang Yoon
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Hyung-Jin Yoo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - John F Keaney
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Shashi Kant
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
20
|
Huang ZH, Huang C, Ji XR, Zhou WJ, Luo XF, Liu Q, Tang YL, Gong F, Zhu WB. MKK7-mediated phosphorylation of JNKs regulates the proliferation and apoptosis of human spermatogonial stem cells. World J Stem Cells 2021; 13:1797-1812. [PMID: 34909124 PMCID: PMC8641020 DOI: 10.4252/wjsc.v13.i11.1797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/28/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human spermatogonial stem cells (SSCs) are the basis of spermatogenesis. However, little is known about the developmental regulatory mechanisms of SSC due to sample origin and species differences.
AIM To investigates the mechanisms involved in the proliferation of human SSC.
METHODS The expression of mitogen-activated protein kinase kinase 7 (MKK7) in human testis was identified using immunohistochemistry and western blotting (WB). MKK7 was knocked down using small interfering RNA, and cell proliferation and apoptosis were detected by WB, EdU, cell counting kit-8 and fluorescence-activated cell sorting. After bioinformatic analysis, the interaction of MKK7 with c-Jun N-terminal kinases ( JNKs ) was verified by protein co-immunoprecipitation and WB. The phosphorylation of JNKs was inhibited by SP600125, and the phenotypic changes were detected by WB, cell counting kit-8 and fluorescence-activated cell sorting.
RESULTS MKK7 is mainly expressed in human SSCs, and MKK7 knockdown inhibits SSC proliferation and promotes their apoptosis. MKK7 mediated the phosphorylation of JNKs, and after inhibiting the phosphorylation of JNKs, the phenotypic changes of the cells were similar to those after MKK7 downregulation. The expression of MKK7 was significantly downregulated in patients with abnormal spermatogenesis, suggesting that abnormal MKK7 may be associated with spermatogenesis impairment.
CONCLUSION MKK7 regulates the proliferation and apoptosis of human SSC by mediating the phosphorylation of JNKs.
Collapse
Affiliation(s)
- Zeng-Hui Huang
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
- Department of Reproductive Center, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410008, Hunan Province, China
| | - Chuan Huang
- Department of Sperm Bank, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410008, Hunan Province, China
| | - Xi-Ren Ji
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Wen-Jun Zhou
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Xue-Feng Luo
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Qian Liu
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Yu-Lin Tang
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Fei Gong
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Wen-Bing Zhu
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
21
|
Protopanaxatriol-type saponin protects against acetaminophen-induced liver injury through ROS-mediated JNK pathway. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
22
|
Serafim RAM, Elkins JM, Zuercher WJ, Laufer SA, Gehringer M. Chemical Probes for Understudied Kinases: Challenges and Opportunities. J Med Chem 2021; 65:1132-1170. [PMID: 34477374 DOI: 10.1021/acs.jmedchem.1c00980] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Over 20 years after the approval of the first-in-class protein kinase inhibitor imatinib, the biological function of a significant fraction of the human kinome remains poorly understood while most research continues to be focused on few well-validated targets. Given the strong genetic evidence for involvement of many kinases in health and disease, the understudied fraction of the kinome holds a large and unexplored potential for future therapies. Specific chemical probes are indispensable tools to interrogate biology enabling proper preclinical validation of novel kinase targets. In this Perspective, we highlight recent case studies illustrating the development of high-quality chemical probes for less-studied kinases and their application in target validation. We spotlight emerging techniques and approaches employed in the generation of chemical probes for protein kinases and beyond and discuss the associated challenges and opportunities.
Collapse
Affiliation(s)
- Ricardo A M Serafim
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Jonathan M Elkins
- Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - William J Zuercher
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Stefan A Laufer
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided & Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany.,Tübingen Center for Academic Drug Discovery, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Matthias Gehringer
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided & Functionally Instructed Tumor Therapies", University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
23
|
Inhibition of the MAP2K7-JNK pathway with 5Z-7-oxozeaenol induces apoptosis in T-cell acute lymphoblastic leukemia. Oncotarget 2021; 12:1787-1801. [PMID: 34504651 PMCID: PMC8416565 DOI: 10.18632/oncotarget.28040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 07/28/2021] [Indexed: 01/10/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive pediatric leukemia with a worse prognosis than most frequent B-cell ALL due to a high incidence of treatment failures and relapse. Our previous work showed that loss of the pioneer factor KLF4 in a NOTCH1-induced T-ALL mouse model accelerated the development of leukemia through expansion of leukemia-initiating cells and activation of the MAP2K7 pathway. Similarly, epigenetic silencing of the KLF4 gene in children with T-ALL was associated with MAP2K7 activation. Here, we showed the small molecule 5Z-7-oxozeaenol (5Z7O) induces dose-dependent cytotoxicity in a panel of T-ALL cell lines mainly through inhibition of the MAP2K7-JNK pathway, which further validates MAP2K7 as a therapeutic target. Mechanistically, 5Z7O-mediated apoptosis was caused by the downregulation of regulators of the G2/M checkpoint and the inhibition of survival pathways. The anti-leukemic capacity of 5Z7O was evaluated using leukemic cells from two mouse models of T-ALL and patient-derived xenograft cells generated using lymphoblasts from pediatric T-ALL patients. Finally, a combination of 5Z7O with dexamethasone, a drug used in frontline therapy, showed synergistic induction of cytotoxicity. In sum, we report here that MAP2K7 inhibition thwarts survival mechanisms in T-ALL cells and warrants future pre-clinical studies for high-risk and relapsed patients.
Collapse
|
24
|
Ji C, Zhang Z, Chen J, Song D, Liu B, Li J, Liu R, Niu J, Wang D, Ling N, Qi Z, Li W. Immune-Enhancing Effects of a Novel Glucan from Purple Sweet Potato Ipomoea batatas (L.) Lam on RAW264.7 Macrophage Cells via TLR2- and TLR4-Mediated Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:9313-9325. [PMID: 34370469 DOI: 10.1021/acs.jafc.1c03850] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
PSPP-1 was obtained from purple sweet potato, and the effects of PSPP-1 on the immune modulation on macrophage cells were investigated for the first time. PSPP-1 promoted RAW264.7 proliferation and increased the total cell percentage in DNA synthesis and mitosis phases, and the cell morphology changed in volume and appearance. Additionally, the RAW264.7 immune functions of phagocytic activity and nitric oxide, reactive oxygen species, and cytokine production were improved by PSPP-1. The western blot experiment showed that PSPP-1 could activate toll-like receptor 2 and toll-like receptor 4-mediated pathways, and the expressions of proteins in MyD88-dependent, mitogen-activated protein kinase (MAPK)-signaling, NF-κB-signaling, AP-1 signaling, and TRIF-dependent pathways were improved markedly. Molecular docking and Biolayer Interferometry study further indicated that PSPP-1 could recognize and bind TLR2 and TLR4 by targeting the binding sites with a strong affinity. It suggested that PSPP-1 could enhance immunity via TLR2- and TLR4-mediated pathways, and it could be explored as an immunomodulatory agent.
Collapse
Affiliation(s)
- Chenfeng Ji
- Engineering Research Center for Medicine, College of Pharmacy, Harbin University of Commerce, Harbin 150076, China
- Engineering Research Center of Natural Anticancer Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Ziyi Zhang
- Engineering Research Center for Medicine, College of Pharmacy, Harbin University of Commerce, Harbin 150076, China
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Jinrui Chen
- Engineering Research Center for Medicine, College of Pharmacy, Harbin University of Commerce, Harbin 150076, China
| | - Dongxue Song
- Engineering Research Center for Medicine, College of Pharmacy, Harbin University of Commerce, Harbin 150076, China
- Engineering Research Center of Natural Anticancer Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Bing Liu
- Engineering Research Center for Medicine, College of Pharmacy, Harbin University of Commerce, Harbin 150076, China
- Engineering Research Center of Natural Anticancer Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Jun Li
- Engineering Research Center for Medicine, College of Pharmacy, Harbin University of Commerce, Harbin 150076, China
| | - Rongyu Liu
- Engineering Research Center for Medicine, College of Pharmacy, Harbin University of Commerce, Harbin 150076, China
| | - Junbo Niu
- Engineering Research Center for Medicine, College of Pharmacy, Harbin University of Commerce, Harbin 150076, China
| | - Di Wang
- Engineering Research Center for Medicine, College of Pharmacy, Harbin University of Commerce, Harbin 150076, China
| | - Na Ling
- Engineering Research Center for Medicine, College of Pharmacy, Harbin University of Commerce, Harbin 150076, China
- Engineering Research Center of Natural Anticancer Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Zheng Qi
- Engineering Research Center for Medicine, College of Pharmacy, Harbin University of Commerce, Harbin 150076, China
- Engineering Research Center of Natural Anticancer Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Wenlan Li
- Engineering Research Center for Medicine, College of Pharmacy, Harbin University of Commerce, Harbin 150076, China
- Engineering Research Center of Natural Anticancer Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| |
Collapse
|
25
|
Kasuya Y, Kim JD, Hatano M, Tatsumi K, Matsuda S. Pathophysiological Roles of Stress-Activated Protein Kinases in Pulmonary Fibrosis. Int J Mol Sci 2021; 22:ijms22116041. [PMID: 34204949 PMCID: PMC8199902 DOI: 10.3390/ijms22116041] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/22/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is one of the most symptomatic progressive fibrotic lung diseases, in which patients have an extremely poor prognosis. Therefore, understanding the precise molecular mechanisms underlying pulmonary fibrosis is necessary for the development of new therapeutic options. Stress-activated protein kinases (SAPKs), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (p38) are ubiquitously expressed in various types of cells and activated in response to cellular environmental stresses, including inflammatory and apoptotic stimuli. Type II alveolar epithelial cells, fibroblasts, and macrophages are known to participate in the progression of pulmonary fibrosis. SAPKs can control fibrogenesis by regulating the cellular processes and molecular functions in various types of lung cells (including cells of the epithelium, interstitial connective tissue, blood vessels, and hematopoietic and lymphoid tissue), all aspects of which remain to be elucidated. We recently reported that the stepwise elevation of intrinsic p38 signaling in the lungs is correlated with a worsening severity of bleomycin-induced fibrosis, indicating an importance of this pathway in the progression of pulmonary fibrosis. In addition, a transcriptome analysis of RNA-sequencing data from this unique model demonstrated that several lines of mechanisms are involved in the pathogenesis of pulmonary fibrosis, which provides a basis for further studies. Here, we review the accumulating evidence for the spatial and temporal roles of SAPKs in pulmonary fibrosis.
Collapse
Affiliation(s)
- Yoshitoshi Kasuya
- Department of Biomedical Science, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (M.H.); (S.M.)
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Correspondence: ; Tel.: +81-432-262-193; Fax: +81-432-262-196
| | - Jun-Dal Kim
- Department of Research and Development, Institute of Natural Medicine (INM), University of Toyama, Toyama 930-0194, Japan;
| | - Masahiko Hatano
- Department of Biomedical Science, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (M.H.); (S.M.)
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan;
| | - Shuichi Matsuda
- Department of Biomedical Science, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (M.H.); (S.M.)
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan;
| |
Collapse
|
26
|
Ha AT, Kim MY, Cho JY. TAK1/AP-1-Targeted Anti-Inflammatory Effects of Barringtonia augusta Methanol Extract. Molecules 2021; 26:molecules26103053. [PMID: 34065429 PMCID: PMC8160894 DOI: 10.3390/molecules26103053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
Barringtonia augusta methanol extract (Ba-ME) is a folk medicine found in the wetlands of Thailand that acts through an anti-inflammatory mechanism that is not understood fully. Here, we examine how the methanol extract of Barringtonia augusta (B. augusta) can suppress the activator protein 1 (AP-1) signaling pathway and study the activities of Ba-ME in the lipopolysaccharide (LPS)-treated RAW264.7 macrophage cell line and an LPS-induced peritonitis mouse model. Non-toxic concentrations of Ba-ME downregulated the mRNA expression of cytokines, such as cyclooxygenase and chemokine ligand 12, in LPS-stimulated RAW264.7 cells. Transfection experiments with the AP-1-Luc construct, HEK293T cells, and luciferase assays were used to assess whether Ba-ME suppressed the AP-1 functional activation. A Western blot assay confirmed that C-Jun N-terminal kinase is a direct pharmacological target of Ba-ME action. The anti-inflammatory effect of Ba-ME, which functions by β-activated kinase 1 (TAK1) inhibition, was confirmed by using an overexpression strategy and a cellular thermal shift assay. In vivo experiments in a mouse model of LPS-induced peritonitis showed the anti-inflammatory effect of Ba-ME on LPS-stimulated macrophages and acute inflammatory mouse models. We conclude that Ba-ME is a promising anti-inflammatory drug targeting TAK1 in the AP-1 pathway.
Collapse
Affiliation(s)
- Anh Thu Ha
- Department of Integrative Biotechnology, and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea;
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Korea
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.: +82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea;
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.: +82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| |
Collapse
|
27
|
Fang Z, Kim HG, Huang M, Chowdhury K, Li MO, Liangpunsakul S, Dong XC. Sestrin Proteins Protect Against Lipotoxicity-Induced Oxidative Stress in the Liver via Suppression of C-Jun N-Terminal Kinases. Cell Mol Gastroenterol Hepatol 2021; 12:921-942. [PMID: 33962074 PMCID: PMC8346671 DOI: 10.1016/j.jcmgh.2021.04.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND & AIMS Sestrin 1/2/3 (Sesn1/2/3) belong to a small family of proteins that have been implicated in the regulation of metabolic homeostasis and oxidative stress. However, the underlying mechanisms remain incompletely understood. The aim of this work was to illustrate the collective function of Sesn1/2/3 in the protection against hepatic lipotoxicity. METHODS We used Sesn1/2/3 triple knockout (TKO) mouse and cell models to characterize oxidative stress and signal transduction under lipotoxic conditions. Biochemical, histologic, and physiological approaches were applied to illustrate the related processes. RESULTS After feeding with a Western diet for 8 weeks, TKO mice developed remarkable metabolic associated fatty liver disease that was manifested by exacerbated hepatic steatosis, inflammation, and fibrosis compared with wild-type counterparts. Moreover, TKO mice exhibited higher levels of hepatic lipotoxicity and oxidative stress. Our biochemical data revealed a critical signaling node from sestrins to c-Jun N-terminal kinases (JNKs) in that sestrins interact with JNKs and mitogen-activated protein kinase kinase 7 and suppress the JNK phosphorylation and activity. In doing so, sestrins markedly reduced palmitate-induced lipotoxicity and oxidative stress in both mouse and human hepatocytes. CONCLUSIONS The data from this study suggest that Sesn1/2/3 play an important role in the protection against lipotoxicity-associated oxidative stress and related pathology in the liver.
Collapse
Affiliation(s)
- Zhigang Fang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana,Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China,The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hyeong-Geug Kim
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Menghao Huang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kushan Chowdhury
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ming O. Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Suthat Liangpunsakul
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana,Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana,Roudebush Veterans Administration Medical Center, Indianapolis, Indiana
| | - X. Charlie Dong
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana,Correspondence Address correspondence to: X. Charlie Dong, PhD, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS 1021D, Indianapolis, Indiana 46202. fax: (317) 274-4686.
| |
Collapse
|
28
|
Du X, Liu H, Liu X, Chen X, Yuan L, Ma Y, Huang H, Wang Y, Wang R, Zhang S, Tian Z, Shi L, Zhang H. Microcystin-LR induces ovarian injury and apoptosis in mice via activating apoptosis signal-regulating kinase 1-mediated P38/JNK pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 213:112066. [PMID: 33610944 DOI: 10.1016/j.ecoenv.2021.112066] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/06/2021] [Accepted: 02/12/2021] [Indexed: 06/12/2023]
Abstract
As an emerging pollutant in the aquatic environment, microcystin-LR (MC-LR) can enter the body through multiple pathways, and then induce apoptosis and gonadal damage, affecting reproductive function. Previous studies focused on male reproductive toxicity induced by MC-LR neglecting its effects on females. The apoptotic signal-regulated kinase 1 (ASK1) is an upstream protein of P38/JNK pathway, closely associated with apoptosis and organ damage. However, the role of ASK1 in MC-LR-induced reproductive toxicity is unclear. Therefore, this study investigated the role of ASK1 in mouse ovarian injury and apoptosis induced by MC-LR. After MC-LR exposure, ASK1 expression in mouse ovarian granulosa cells was increased at the protein and mRNA levels, and decreased following pretreatment by antioxidant N-acetylcysteine, suggesting that MC-LR-induced oxidative stress has a regulatory role in ASK1 expression. Inhibition of ASK1 expression with siASK1 and NQDI-1 could effectively alleviate MC-LR-induced mitochondrial membrane potential damage and apoptosis in ovarian granulosa cells, as well as pathological damage, apoptosis and the decreased gonadal index in ovaries of C57BL/6 mice. Moreover, the P38/JNK pathway and downstream apoptosis-related proteins (P-P38, P-JNK, P-P53, Fas) and genes (MKK4, MKK3, Ddit3, Mef2c) were activated in vivo and vitro, but their activation was restrained after ASK1 inhibition. Data presented herein suggest that the ASK1-mediated P38/JNK pathway is involved in ovarian injury and apoptosis induced by MC-LR in mice. It is confirmed that ASK1 has an important role in MC-LR-induced ovarian injury, which provides new insights for preventing MCs-induced reproductive toxicity in females.
Collapse
Affiliation(s)
- Xingde Du
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Haohao Liu
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaohui Liu
- School of Basic Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Xinghai Chen
- Department of Chemistry and Biochemistry, St Mary's University, San Antonio, TX 78228, USA
| | - Le Yuan
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Ya Ma
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Hui Huang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Yueqin Wang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Rui Wang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Shiyu Zhang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Zhihui Tian
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Linjia Shi
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Huizhen Zhang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
29
|
Tan XD, Liu B, Jiang Y, Yu HJ, Li CQ. Gadd45b mediates environmental enrichment-induced neurogenesis in the SVZ of rats following ischemia stroke via BDNF. Neurosci Lett 2021; 745:135616. [PMID: 33421485 DOI: 10.1016/j.neulet.2020.135616] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 12/15/2022]
Abstract
Ischemic stroke is a major cause of mortality and disability. Subventricular zone (SVZ) neurogenesis following an ischemic stroke may be beneficial for improving the outcomes. Environmental enrichment (EE) has been reported to increase neurogenesis following stroke. Growth arrest and DNA-damage-inducible protein 45 β (Gadd45b) is a crucial gene for activity-correlated neurogenesis in the adult hippocampus of mice. This study examined whether Gadd45b inhibition affects adult SVZ neurogenesis after an ischemic injury and explored the role of Gadd45b in EE-induced SVZ neurogenesis in adult male Sprague Dawley rats following middle cerebral artery occlusion (MCAO). Gadd45b expression was silenced by a lentivirus with RNA interference (RNAi). The 5-ethynyl-2-deoxyuridine (EdU) staining test was performed to detect cell proliferation. Gadd45b-RNAi after MCAO decreased SVZ proliferation and differentiation in the infarction boundary following ischemic injury, accompanied by the depressed expression of the brain-derived neurotrophic factor (BDNF). Treatment with EE following ischemic stroke upregulated Gadd45b and BDNF expressions and increased neurogenesis in the SVZ. Inhibition of Gadd45b markedly ameliorated the increased neurogenesis induced by EE. These data indicated that Gadd45b is related to SVZ neurogenesis following ischemic stroke, and Gadd45b mediates EE-induced neurogenesis via BDNF in the SVZ of rats following an ischemia stroke. These results implicate that Gadd45b can be a potential therapeutic target to enhance adult neurogenesis following cerebral ischemia.
Collapse
Affiliation(s)
- Xiao-Dan Tan
- Rehabilitation Department, The Third Affiliated Hospital of Chongqing Medical University, Yubei District, Chongqing, 401120, China
| | - Bin Liu
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014, China
| | - Ying Jiang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400010, China
| | - Hong-Jun Yu
- Rehabilitation Department, The Third Affiliated Hospital of Chongqing Medical University, Yubei District, Chongqing, 401120, China
| | - Chang-Qing Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
30
|
Insights into the Interaction Mechanism of DTP3 with MKK7 by Using STD-NMR and Computational Approaches. Biomedicines 2020; 9:biomedicines9010020. [PMID: 33396582 PMCID: PMC7824710 DOI: 10.3390/biomedicines9010020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 01/18/2023] Open
Abstract
GADD45β/MKK7 complex is a non-redundant, cancer cell-restricted survival module downstream of the NF-kB survival pathway, and it has a pathogenically critical role in multiple myeloma, an incurable malignancy of plasma cells. The first-in-class GADD45β/MKK7 inhibitor DTP3 effectively kills MM cells expressing its molecular target, both in vitro and in vivo, by inducing MKK7/JNK-dependent apoptosis with no apparent toxicity to normal cells. DTP3 combines favorable drug-like properties, with on-target-specific pharmacology, resulting in a safe and cancer-selective therapeutic effect; however, its mode of action is only partially understood. In this work, we have investigated the molecular determinants underlying the MKK7 interaction with DTP3 by combining computational, NMR, and spectroscopic methods. Data gathered by fluorescence quenching and computational approaches consistently indicate that the N-terminal region of MKK7 is the optimal binding site explored by DTP3. These findings further the understanding of the selective mode of action of GADD45β/MKK7 inhibitors and inform potential mechanisms of drug resistance. Notably, upon validation of the safety and efficacy of DTP3 in human trials, our results could also facilitate the development of novel DTP3-like therapeutics with improved bioavailability or the capacity to bypass drug resistance.
Collapse
|
31
|
Long X, Zhao B, Lu W, Chen X, Yang X, Huang J, Zhang Y, An S, Qin Y, Xing Z, Shen Y, Wu H, Qi Y. The Critical Roles of the SUMO-Specific Protease SENP3 in Human Diseases and Clinical Implications. Front Physiol 2020; 11:558220. [PMID: 33192553 PMCID: PMC7662461 DOI: 10.3389/fphys.2020.558220] [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: 05/01/2020] [Accepted: 10/07/2020] [Indexed: 11/23/2022] Open
Abstract
Post-translational modification by SUMO (small ubiquitin-like modifier) proteins has been shown to regulate a variety of functions of proteins, including protein stability, chromatin organization, transcription, DNA repair, subcellular localization, protein–protein interactions, and protein homeostasis. SENP (sentrin/SUMO-specific protease) regulates precursor processing and deconjugation of SUMO to control cellular mechanisms. SENP3, which is one of the SENP family members, deconjugates target proteins to alter protein modification. The effect of modification via SUMO and SENP3 is crucial to maintain the balance of SUMOylation and guarantee normal protein function and cellular activities. SENP3 acts as an oxidative stress-responsive molecule under physiological conditions. Under pathological conditions, if the SUMOylation process of proteins is affected by variations in SENP3 levels, it will cause a cellular reaction and ultimately lead to abnormal cellular activities and the occurrence and development of human diseases, including cardiovascular diseases, neurological diseases, and various cancers. In this review, we summarized the most recent advances concerning the critical roles of SENP3 in normal physiological and pathological conditions as well as the potential clinical implications in various diseases. Targeting SENP3 alone or in combination with current therapies might provide powerful targeted therapeutic strategies for the treatment of these diseases.
Collapse
Affiliation(s)
- Xiaojun Long
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Biying Zhao
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Wenbin Lu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xu Chen
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xinyi Yang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Jifang Huang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yuhong Zhang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Siming An
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yuanyuan Qin
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Zhengcao Xing
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yajie Shen
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Hongmei Wu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yitao Qi
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
32
|
Jiang J, Jiang B, He Z, Ficarro SB, Che J, Marto JA, Gao Y, Zhang T, Gray NS. Discovery of Covalent MKK4/7 Dual Inhibitor. Cell Chem Biol 2020; 27:1553-1560.e8. [PMID: 32916088 DOI: 10.1016/j.chembiol.2020.08.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/04/2020] [Accepted: 08/20/2020] [Indexed: 12/22/2022]
Abstract
MKK4/7 are kinases that phosphorylate JNKs and regulate the MAPK signaling pathway. Their overexpression has been associated with tumorigenesis and aggressiveness in cancers such as breast, prostate, non-small cell lung, and pediatric leukemia, making them a potential target for inhibitor development. Here, we report the discovery, development, and validation of a dual MKK4/7 inhibitor, BSJ-04-122, that covalently targets a conserved cysteine located before the DFG motif and displays excellent kinome selectivity. BSJ-04-122 exhibits potent cellular target engagement and induces robust target-specific downstream effects. The combination of the dual MKK4/7 inhibitor with a selective, covalent JNK inhibitor demonstrated an enhanced antiproliferative activity against triple-negative breast cancer cells. Taken together, the results show that BSJ-04-122 represents a pharmacological probe for MKK4/7 and credential covalent targeting as a way to explore the therapeutic potential of these kinases.
Collapse
Affiliation(s)
- Jie Jiang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Baishan Jiang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Zhixiang He
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Scott B Ficarro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Jianwei Che
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Jarrod A Marto
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA; Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Yang Gao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Tinghu Zhang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA.
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
33
|
The role of JNK in prostate cancer progression and therapeutic strategies. Biomed Pharmacother 2020; 121:109679. [DOI: 10.1016/j.biopha.2019.109679] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/10/2019] [Accepted: 11/16/2019] [Indexed: 12/31/2022] Open
|