1
|
Seydel B, Hegner P, Lauerer AM, Schildt S, Bayram F, Tafelmeier M, Wermers D, Rupprecht L, Schmid C, Wagner S, Maier LS, Arzt M, Lebek S. Increased Myocardial MARK4 Expression in Patients with Heart Failure and Sleep-Disordered Breathing. Int J Mol Sci 2025; 26:3614. [PMID: 40332117 PMCID: PMC12027440 DOI: 10.3390/ijms26083614] [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: 02/19/2025] [Revised: 04/05/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Cardiovascular diseases are the leading cause of morbidity and mortality worldwide, underscoring the urgent need for novel therapeutic targets and strategies. The kinase MARK4 (MAP (microtubule-associated proteins)/microtubule affinity-regulating kinase 4) regulates microtubule-associated proteins pivotal for cell polarity, protein stability, and intracellular signaling. Animal models of heart failure revealed elevated MARK4 levels, which correlated with impaired cardiac contractility. However, the involvement of MARK4 and its potential as a molecular drug target has not yet been explored in the myocardium of cardiovascular patients. We investigated the MARK4 mRNA expression in human myocardial biopsies of 152 high-risk cardiovascular patients undergoing cardiac surgery. Comprehensive echocardiography as well as testing for sleep-disordered breathing (SDB), a critical comorbidity in heart failure, were assessed preoperatively. We observed a substantial upregulation of myocardial MARK4 expression in patients with impaired cardiac contractility, resulting in an inverse correlation with the left ventricular ejection fraction. Myocardial MARK4 expression also correlated with echocardiographic E/e', a central parameter of diastolic dysfunction. Mechanistically, our analyses revealed that MARK4 expression increases in SDB and under hypoxic conditions, as evidenced by significant correlations between myocardial MARK4 expression and factors like mean oxygen saturation, time with oxygen saturation below 90%, and the oxygen desaturation index. Multivariable regression analysis revealed that both left ventricular ejection fraction and mean oxygen saturation were independently associated with dysregulated MARK4 levels, even when controlling for important clinical covariables as potential confounders. Taken together, our findings demonstrate that MARK4 expression is highly increased in the myocardium of cardiovascular high-risk patients, suggesting it is a potential molecular target against cardiovascular diseases.
Collapse
Affiliation(s)
- Bettina Seydel
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Philipp Hegner
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Anna-Maria Lauerer
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Sönke Schildt
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Fatma Bayram
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Maria Tafelmeier
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Dominik Wermers
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Leopold Rupprecht
- Department of Cardiothoracic Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Christof Schmid
- Department of Cardiothoracic Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Lars Siegfried Maier
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Michael Arzt
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Simon Lebek
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| |
Collapse
|
2
|
Tao Y, Zhang Q, Wang H, Yang X, Mu H. Alternative splicing and related RNA binding proteins in human health and disease. Signal Transduct Target Ther 2024; 9:26. [PMID: 38302461 PMCID: PMC10835012 DOI: 10.1038/s41392-024-01734-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 12/18/2023] [Accepted: 12/27/2023] [Indexed: 02/03/2024] Open
Abstract
Alternative splicing (AS) serves as a pivotal mechanism in transcriptional regulation, engendering transcript diversity, and modifications in protein structure and functionality. Across varying tissues, developmental stages, or under specific conditions, AS gives rise to distinct splice isoforms. This implies that these isoforms possess unique temporal and spatial roles, thereby associating AS with standard biological activities and diseases. Among these, AS-related RNA-binding proteins (RBPs) play an instrumental role in regulating alternative splicing events. Under physiological conditions, the diversity of proteins mediated by AS influences the structure, function, interaction, and localization of proteins, thereby participating in the differentiation and development of an array of tissues and organs. Under pathological conditions, alterations in AS are linked with various diseases, particularly cancer. These changes can lead to modifications in gene splicing patterns, culminating in changes or loss of protein functionality. For instance, in cancer, abnormalities in AS and RBPs may result in aberrant expression of cancer-associated genes, thereby promoting the onset and progression of tumors. AS and RBPs are also associated with numerous neurodegenerative diseases and autoimmune diseases. Consequently, the study of AS across different tissues holds significant value. This review provides a detailed account of the recent advancements in the study of alternative splicing and AS-related RNA-binding proteins in tissue development and diseases, which aids in deepening the understanding of gene expression complexity and offers new insights and methodologies for precision medicine.
Collapse
Affiliation(s)
- Yining Tao
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200000, Shanghai, China
- Shanghai Bone Tumor Institution, 200000, Shanghai, China
| | - Qi Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, 200000, Shanghai, China
| | - Haoyu Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200000, Shanghai, China
- Shanghai Bone Tumor Institution, 200000, Shanghai, China
| | - Xiyu Yang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200000, Shanghai, China
- Shanghai Bone Tumor Institution, 200000, Shanghai, China
| | - Haoran Mu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200000, Shanghai, China.
- Shanghai Bone Tumor Institution, 200000, Shanghai, China.
| |
Collapse
|
3
|
Li Y, Cheng M, Jin J, Zhang D, Zhang S, Bai Y, Xu J. Interaction of Sp1 and Setd8 promotes vascular smooth muscle cells apoptosis by activating Mark4 in vascular calcification. Aging (Albany NY) 2024; 16:2438-2456. [PMID: 38301049 PMCID: PMC10911351 DOI: 10.18632/aging.205492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/29/2023] [Indexed: 02/03/2024]
Abstract
Vascular calcification (VC) is directly related to high mortality in chronic kidney disease (CKD), and cellular apoptosis of vascular smooth muscle cells (VSMCs) is a crucial process in the initiation of VC. Microtubule affinity-regulating kinase 4 (Mark4), known as a serine/threonine protein kinase, can induce cell apoptosis and autophagy by modulating Akt phosphorylation. However, the potential functions and molecular mechanisms of Mark4 in VSMCs apoptosis and calcification need to be further explored. Initially, our data indicated that the mRNA expression of Mark4 was prominently elevated in high phosphorus-stimulated human VSMCs compared with the other members in Marks. Consistently, Mark4 expression was found to be significantly increased in the calcified arteries of both CKD patients and rats. In vitro, silencing Mark4 suppressed apoptosis-specific marker expression by promoting Akt phosphorylation, finally attenuating VSMCs calcification induced by high phosphate. Mechanically, the transcription factor Sp1 was enriched in the Mark4 promoter region and modulated Mark4 transcription. Moreover, SET domain-containing protein 8 (Setd8) was proved to interact with Sp1 and jointly participated in the transcriptional regulation of Mark4. Finally, rescue experiments revealed that Setd8 contributed to VSMCs apoptosis and calcification by modulating Mark4 expression. In conclusion, these findings reveal that Mark4 is transcriptionally activated by Sp1, which is interacted with Setd8, to promote VSMCs calcification through Akt-mediated antiapoptotic effects, suggesting that Mark4 represents a potent and promising therapeutic target for VC in CKD.
Collapse
Affiliation(s)
- Yun Li
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, People’s Republic of China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, People’s Republic of China
| | - Meijuan Cheng
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, People’s Republic of China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, People’s Republic of China
| | - Jingjing Jin
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, People’s Republic of China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, People’s Republic of China
| | - Dongxue Zhang
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, People’s Republic of China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, People’s Republic of China
| | - Shenglei Zhang
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, People’s Republic of China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, People’s Republic of China
| | - Yaling Bai
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, People’s Republic of China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, People’s Republic of China
| | - Jinsheng Xu
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, People’s Republic of China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, People’s Republic of China
| |
Collapse
|
4
|
Mumtaz, Ahmed F, Rabbani SA, El-Tanani M, Najmi AK, Ali J, Khan MA. Tauopathy in AD: Therapeutic Potential of MARK-4. Curr Alzheimer Res 2024; 21:779-790. [PMID: 39931856 DOI: 10.2174/0115672050358397250126151707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/26/2024] [Accepted: 12/05/2024] [Indexed: 04/30/2025]
Abstract
Alzheimer's disease (AD) is one of the leading causes of cognitive decline, which leads to dementia and poses significant challenges for its therapy. The reason is primarily the ineffective available treatments targeting the underlying pathology of AD. It is a neurodegenerative disease that is mainly characterised by the various molecular pathways contributing to its complex pathology, including extracellular amyloid beta (Aβ) plaques, intracellular neurofibrillary tangles (NFTs), oxidative stress, and neuroinflammation. One of the crucial features is the hyperphosphorylation of tau proteins, which is facilitated by microtubule affinity-regulating kinase-4 (MARK-4). The kinase plays a crucial role in the disease development by modifying microtubule integrity, leading to neuronal dysfunction and death. MARK-4 is thus a druggable target and has a pivotal role in AD. Amongst MARK-4 inhibitors, 16 compounds demonstrate significant capacity in molecular docking studies, showing high binding affinity to MARK-4 and promising potential for tau inhibition. Further, in-vitro investigations provide evidence of their neuroprotective properties. The present review mainly focuses on the role of MARK-4 and its potential inhibitors used in treating AD, which have been thoroughly investigated in silico and in vitro..
Collapse
Affiliation(s)
- Mumtaz
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Faraha Ahmed
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Syed Arman Rabbani
- RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Mohamed El-Tanani
- RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| |
Collapse
|
5
|
Alam M, Ahmed S, Abid M, Hasan GM, Islam A, Hassan MI. Therapeutic targeting of microtubule affinity-regulating kinase 4 in cancer and neurodegenerative diseases. J Cell Biochem 2023; 124:1223-1240. [PMID: 37661636 DOI: 10.1002/jcb.30468] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/12/2023] [Accepted: 08/21/2023] [Indexed: 09/05/2023]
Abstract
Microtubule affinity-regulating kinase 4 (MARK4) is a member of the Ser/Thr protein kinase family, phosphorylates the microtubule-connected proteins and plays a vital role in causing cancers and neurodegenerative diseases. This kinase modulates multiple signaling pathways, including mammalian target of rapamycin, nuclear factor-κB, and Hippo-signaling, presumably responsible for cancer and Alzheimer's. MARK4 acts as a negative controller of the Hippo-kinase cassette for promoting YAP/TAZ action, and the loss of MARK4 detains the tumorigenic properties of cancer cells. MARK4 is involved in tau hyperphosphorylation that consequently affects neurodegeneration. MARK4 is a promising drug target for cancer, diabetes, and Alzheimer's. Developing the potent and selective inhibitors of MAKR4 are promising in the therapeutic management of associated diseases. Despite its great significance, a few reviews are available to discuss its structure, function and clinical significance. In the current review, we aimed to provide detailed information on the structural features of MARK4 targeted in drug development and its role in various signaling pathways related to cancer and neurodegenerative diseases. We further described the therapeutic potential of MARK4 inhibitors in preventing numerous diseases. Finally, the updated information on MARK4 will be helpful in the further development of effective therapeutic molecules.
Collapse
Affiliation(s)
- Manzar Alam
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Sarfraz Ahmed
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Mohammad Abid
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Asimul Islam
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
6
|
Voura M, Anwar S, Sigala I, Parasidou E, Fragoulidou S, Hassan MI, Sarli V. Synthesis, Structural Modification, and Bioactivity Evaluation of Substituted Acridones as Potent Microtubule Affinity-Regulating Kinase 4 Inhibitors. ACS Pharmacol Transl Sci 2023; 6:1052-1074. [PMID: 37470016 PMCID: PMC10353068 DOI: 10.1021/acsptsci.3c00084] [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: 04/19/2023] [Indexed: 07/21/2023]
Abstract
Acridones present numerous pharmacological activities, including inhibition of microtubule affinity-regulating kinase 4 (MARK4) kinase activity. To investigate structure-activity relationships and develop potent MARK4 inhibitors, derivatives of 2-methylacridone were synthesized and tested for their activity against MARK4 kinase. Selective substitutions at the nitrogen atom were accomplished by treating 2-methylacridone with alkyl halides in the presence of K2CO3. In addition, amidation of acridone acetic acid 11 with piperazine or tryptophan methyl ester followed by derivatization with various amines gave a series of new acridone derivatives. Among the tested compounds, six were identified as possessing high inhibitory activity against MARK4. The molecular modeling studies showed that the derivatives bearing piperazine or tryptophan bind well to the ATP-binding site of MARK4. The antiproliferative activity of six active compounds was evaluated against HeLa and U87MG cancer cells. Tryptophan derivatives 23a, 23b, and 23c showed significant cytotoxicity against both cell lines with EC50 values ranging from 2.13 to 4.22 μM, while derivatives bearing piperazine were found to be not cytotoxic. Additionally, compound 23a decreased the proliferation of human MDA-MB-435 and U251 cancer cells in the low micromolar range; however, it also affects the non-cancerous HGF cells. Due to their high binding affinity against MARK4, the synthesized compounds could be potential agents to target MARK4 against cancer and tauopathies.
Collapse
Affiliation(s)
- Maria Voura
- Laboratory
of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Saleha Anwar
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ioanna Sigala
- Laboratory
of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Eleftheria Parasidou
- Laboratory
of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Souzanna Fragoulidou
- Laboratory
of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Md. Imtaiyaz Hassan
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Vasiliki Sarli
- Laboratory
of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| |
Collapse
|
7
|
Liu Y, Chen Y, Fukui K. Oxidative stress induces tau hyperphosphorylation via MARK activation in neuroblastoma N1E-115 cells. J Clin Biochem Nutr 2023; 73:24-33. [PMID: 37534088 PMCID: PMC10390814 DOI: 10.3164/jcbn.22-39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 12/28/2022] [Indexed: 08/04/2023] Open
Abstract
Reactive oxygen species are considered a cause of neuronal cell death in Alzheimer's disease (AD). Abnormal tau phosphorylation is a proven pathological hallmark of AD. Microtubule affinity-regulating kinases (MARKs) regulate tau-microtubule binding and play a crucial role in neuronal survival. In this study, we hypothesized that oxidative stress increases the phosphorylation of Ser262 of tau protein through activation of MARKs, which is the main reason for the development of AD. We investigated the relationship between tau hyperphosphorylation on Ser262 and MARKs in N1E-115 cells subjected to oxidative stress by exposure to a low concentration of hydrogen peroxide. This work builds on the observation that hyperphosphorylation of tau is significantly increased by oxidative stress. MARKs activation correlated with tau hyperphosphorylation at Ser262, a site that is essential to maintain microtubule stability and is the initial phosphorylation site in AD. These results indicated that MARKs inhibitors might serve a role as therapeutic tools for the treatment of AD.
Collapse
Affiliation(s)
- Yuhong Liu
- Molecular Cell Biology Laboratory, Department of Functional Control Systems, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan
| | - Yunxi Chen
- Molecular Cell Biology Laboratory, Department of Systems Engineering and Science, School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan
| | - Koji Fukui
- Molecular Cell Biology Laboratory, Department of Functional Control Systems, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan
- Molecular Cell Biology Laboratory, Department of Systems Engineering and Science, School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan
| |
Collapse
|
8
|
Al-Soud YA, Saber SOW, Shtaiwi A, Alsawakhneh SO, Alhelal KAS, Salman QFA, Abu-Qatouseh L, Khanfar MA, Al-Qawasmeh RA. Nitroimidazoles Part 10. Synthesis, crystal structure, molecular docking, and anticancer evaluation of 4-nitroimidazole derivatives combined with piperazine moiety. Z NATURFORSCH C 2023; 78:93-103. [PMID: 35589618 DOI: 10.1515/znc-2022-0023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/30/2022] [Indexed: 11/15/2022]
Abstract
Piperazine-tagged imidazole derivatives 3a (symmetrical di-substituted piperazine) and 5-11 were synthesized through the combination of 4-nitroimidazole derivatives with piperazine moiety. The structural characterization was done by different physical and spectral techniques like NMR (1H and 13C) and mass spectrometry. The constituency of compound 3a was confirmed by X-ray structural analyses. All compounds were assessed for their antiproliferative inhibition potency against five human cancer cell lines namely MCF-7, PC3, MDA-231, A549 and Fibro dental. Compound 5 was found to be the most potent anticancer agents against MCF-7 cell line with IC50 values of (1.0 ± 0 µm) and against PC3 with IC50 value of (9.00 ± 0.028 µm). The molecular docking of compound 5 had been studied, and the results revealed that the newly designed 4-nitroimidazole combined with piperazine moiety derivatives bond to the hydrophobic pocket and polar contacts with high affinity.
Collapse
Affiliation(s)
- Yaseen A Al-Soud
- Department of Chemistry, Faculty of Science, Al al-Bayt University, Al-Mafraq, Jordan
| | | | - Amneh Shtaiwi
- School of Pharmacy, Middle East University, Queen Alia Airport Street, 11118 Amman, Jordan
| | - Sondos O Alsawakhneh
- Department of Chemistry, Faculty of Science, Al al-Bayt University, Al-Mafraq, Jordan
| | - Kafa' A S Alhelal
- Department of Chemistry, Faculty of Science, Al al-Bayt University, Al-Mafraq, Jordan
| | - Qusay F A Salman
- Department of Chemistry, Faculty of Science, Al al-Bayt University, Al-Mafraq, Jordan
| | | | - Monther A Khanfar
- Department of Chemistry, Pure and Applied Chemistry Group, College of Sciences, University of Sharjah, 27272 Sharjah, UAE.,Department of Chemistry, The University of Jordan, 11942 Amman, Jordan
| | - Raed A Al-Qawasmeh
- Department of Chemistry, Pure and Applied Chemistry Group, College of Sciences, University of Sharjah, 27272 Sharjah, UAE.,Department of Chemistry, The University of Jordan, 11942 Amman, Jordan
| |
Collapse
|
9
|
Tang EI, Cheng CY. MARK2 and MARK4 Regulate Sertoli Cell BTB Dynamics Through Microtubule and Actin Cytoskeletons. Endocrinology 2022; 163:6667645. [PMID: 35971301 PMCID: PMC10147390 DOI: 10.1210/endocr/bqac130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Indexed: 11/19/2022]
Abstract
Microtubule affinity-regulating kinases (MARKs) are nonreceptor Ser/Thr protein kinases known to regulate cell polarity and microtubule dynamics in Caenorhabditis elegans, Drosophila, invertebrates, vertebrates, and mammals. An earlier study has shown that MARK4 is present at the ectoplasmic specialization and blood-testis barrier (BTB) in the seminiferous epithelium of adult rat testes. Here, we report the function of MARK4 and another isoform MARK2 in Sertoli cells at the BTB. Knockdown of MARK2, MARK4, or MARK2 and MARK4 by RNAi using the corresponding siRNA duplexes without apparent off-target effects was shown to impair tight junction (TJ)-permeability barrier at the Sertoli cell BTB. It also disrupted microtubule (MT)- and actin-based cytoskeletal organization within Sertoli cells. Although MARK2 and MARK4 were shown to share sequence homology, they likely regulated the Sertoli cell BTB and MT cytoskeleton differently. Disruption of the TJ-permeability barrier following knockdown of MARK4 was considerably more severe than loss of MARK2, though both perturbed the barrier. Similarly, loss of MARK2 affected MT organization in a different manner than the loss of MARK4. Knockdown of MARK2 caused MT bundles to be arranged around the cell periphery, whereas knockdown of MARK4 caused MTs to retract from the cell edge. These differences in effects on the TJ-permeability barrier are likely from the unique roles of MARK2 and MARK4 in regulating the MT cytoskeleton of the Sertoli cell.
Collapse
Affiliation(s)
- Elizabeth I Tang
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, Rockefeller University, New York, NY 10065, USA
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, Rockefeller University, New York, NY 10065, USA
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| |
Collapse
|
10
|
Jenardhanan P, Panneerselvam M, Mathur PP. Use of Molecular Modeling to Study Spermatogenesis: An Overview Using Proteins in Sertoli Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1288:205-214. [PMID: 34453738 DOI: 10.1007/978-3-030-77779-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Computational structure prediction and analysis helps in understanding the structure and function of varied proteins, which otherwise becomes implausible to understand by experimental procedures. Computational techniques prove to be instrumental in understanding the molecular mechanisms that underlies physiological processes and thereby also assist in identification of potent inhibitors. Spermatogenesis, being an important cellular process that decides the fate of the progeny, holds numerous molecular interaction data, which when identified and visualized with computational structural insights, might yield a cohesive and clear-cut perception to the functionality of several proteins involved. The present chapter deals with a few selected applications of computational structure prediction towards understanding the structure of proteins and highlights how these insights are useful in providing a better understanding of different processes in spermatogenesis.
Collapse
Affiliation(s)
| | - Manivel Panneerselvam
- Department of Biotechnology, BJM School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Premendu P Mathur
- Department of Biochemistry & Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, India.
| |
Collapse
|
11
|
Qin YS, Li H, Wang SZ, Wang ZB, Tang CK. Microtubule affinity regulating kinase 4: A promising target in the pathogenesis of atherosclerosis. J Cell Physiol 2021; 237:86-97. [PMID: 34289095 DOI: 10.1002/jcp.30530] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 12/25/2022]
Abstract
Microtubule affinity regulating kinase 4 (MARK4), an important member of the serine/threonine kinase family, regulates the phosphorylation of microtubule-associated proteins and thus modulates microtubule dynamics. In human atherosclerotic lesions, the expression of MARK4 is significantly increased. Recently, accumulating evidence suggests that MARK4 exerts a proatherogenic effect via regulation of lipid metabolism (cholesterol, fatty acid, and triglyceride), inflammation, cell cycle progression and proliferation, insulin signaling, and glucose homeostasis, white adipocyte browning, and oxidative stress. In this review, we summarize the latest findings regarding the role of MARK4 in the pathogenesis of atherosclerosis to provide a rationale for future investigation and therapeutic intervention.
Collapse
Affiliation(s)
- Yu-Sheng Qin
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province,Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic disease, Medical Instrument and equipment technology laboratory of Hengyang medical college, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Heng Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province,Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic disease, Medical Instrument and equipment technology laboratory of Hengyang medical college, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Shu-Zhi Wang
- Institute of Pharmacy and Pharmacology, School of Pharmacy; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China
| | - Zong-Bao Wang
- Institute of Pharmacy and Pharmacology, School of Pharmacy; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province,Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic disease, Medical Instrument and equipment technology laboratory of Hengyang medical college, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| |
Collapse
|
12
|
Sun J, Liao W, Su K, Jia J, Qin L, Liu W, He Y, Zhang H, Ou F, Zhang Z, Sun Y. Suberoylanilide Hydroxamic Acid Attenuates Interleukin-1β-Induced Interleukin-6 Upregulation by Inhibiting the Microtubule Affinity-Regulating Kinase 4/Nuclear Factor-κB Pathway in Synovium-Derived Mesenchymal Stem Cells from the Temporomandibular Joint. Inflammation 2021; 43:1246-1258. [PMID: 32279160 DOI: 10.1007/s10753-020-01204-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Synovium-derived mesenchymal stem cells (SMSCs) can migrate to the site of destroyed condylar cartilage and differentiate into chondrocytes to repair temporomandibular joint (TMJ) damage. Interleukin (IL)-1β-induced IL-6 secretion has been shown to inhibit the chondrogenic potential of SMSCs. The histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA) has recently been shown to be closely related to the inflammation induced by IL-1β. However, the relationship between SAHA and IL-6 secretion induced by IL-1β in SMSCs remains unclear. In this study, we evaluated the relationships between IL-1β and IL-6 in synovial specimens from patients with TMD and in model rats with osteoarthritis (OA). We found that IL-1β and IL-6 were positively correlated and that IL-6 expression in SMSCs increased with IL-1β stimulation in vitro. Moreover, microtubule affinity-regulating kinase 4 (MARK4) was significantly upregulated in IL-1β-stimulated SMSCs and in the synovium of rats with OA. MARK4 knockdown inhibited IL-6 secretion and nuclear factor (NF)-κB pathway activation in IL-1β-stimulated SMSCs. SAHA attenuated IL-6 secretion in IL-1β-induced SMSCs through NF-κB pathway inhibition, and MARK4 was also downregulated in SAHA-treated SMSCs. However, inhibition of the NF-κB pathway did not suppress MARK4 expression. Thus, these results showed that SAHA attenuated IL-6 secretion in IL-1β-induced SMSCs through inhibition of the MARK4/NF-κB pathway.
Collapse
Affiliation(s)
- Jiadong Sun
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, No. 56 Lingyuan West Road, Guangzhou, Guangdong, People's Republic of China
| | - Wenting Liao
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, No. 56 Lingyuan West Road, Guangzhou, Guangdong, People's Republic of China
| | - Kai Su
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, No. 56 Lingyuan West Road, Guangzhou, Guangdong, People's Republic of China
| | - Jiaxin Jia
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, No. 56 Lingyuan West Road, Guangzhou, Guangdong, People's Republic of China
| | - Lingling Qin
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, No. 56 Lingyuan West Road, Guangzhou, Guangdong, People's Republic of China
| | - Wenjing Liu
- Stomatological Hospital of Guangdong Province, Guangzhou, Guangdong, People's Republic of China
| | - Yiqing He
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, No. 56 Lingyuan West Road, Guangzhou, Guangdong, People's Republic of China
| | - Hong Zhang
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, No. 56 Lingyuan West Road, Guangzhou, Guangdong, People's Republic of China
| | - Farong Ou
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, No. 56 Lingyuan West Road, Guangzhou, Guangdong, People's Republic of China
| | - Zhiguang Zhang
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, No. 56 Lingyuan West Road, Guangzhou, Guangdong, People's Republic of China.
| | - Yangpeng Sun
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, No. 56 Lingyuan West Road, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
13
|
Gupta P, Taiyab A, Hassan MI. Emerging role of protein kinases in diabetes mellitus: From mechanism to therapy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 124:47-85. [PMID: 33632470 DOI: 10.1016/bs.apcsb.2020.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Diabetes mellitus has emerged as a severe burden on the medical health system across the globe. Presently, around 422 million people are suffering from diabetes which is speculated to be expanded to about 600 million by 2035. Patients with type 2 diabetes are at increased risk of developing detrimental metabolic and cardiovascular complications. The scientific understanding of this chronic disease and its underlying root cause is not yet fully unraveled. Protein kinases are well known to regulate almost every cellular process through phosphorylation of target protein in diverse signaling pathways. The important role of several protein kinases including AMP-activated protein kinase, IκB kinase and protein kinase C have been well demonstrated in various animal models. They modulate glucose tolerance, inflammation and insulin resistance in the cells via acting on diverse downstream targets and signaling pathways. Thus, modulating the activity of potential human kinases which are significantly involved in diabetes by targeting with small molecule inhibitors could be an attractive therapeutic strategy to tackle diabetes. In this chapter, we have discussed the potential role of protein kinases in glucose metabolism and insulin sensitivity, and in the pathogenesis of diabetes mellitus. Furthermore, the small molecules reported in the literature that can be potentially used for the treatment of diabetes have been discussed in detail.
Collapse
Affiliation(s)
- Preeti Gupta
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Aaliya Taiyab
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| |
Collapse
|
14
|
Miller JB, Ward E, Staley LA, Stevens J, Teerlink CC, Tavana JP, Cloward M, Page M, Dayton L, Cannon-Albright LA, Kauwe JSK. Identification and genomic analysis of pedigrees with exceptional longevity identifies candidate rare variants. Neurobiol Dis 2020; 143:104972. [PMID: 32574725 PMCID: PMC7461696 DOI: 10.1016/j.nbd.2020.104972] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/05/2020] [Accepted: 06/12/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Longevity as a phenotype entails living longer than average and typically includes living without chronic age-related diseases. Recently, several common genetic components to longevity have been identified. This study aims to identify additional genetic variants associated with longevity using unique and powerful analyses of pedigrees with a statistical excess of healthy elderly individuals identified in the Utah Population Database (UPDB). METHODS From an existing biorepository of Utah pedigrees, six independent cousin pairs were selected from four extended pedigrees that exhibited an excess of healthy elderly individuals; whole exome sequencing (WES) was performed on two elderly individuals from each pedigree who were either first cousins or first cousins once removed. Rare (<.01 population frequency) variants shared by at least one elderly cousin pair in a region likely to be identical by descent were identified as candidates. Ingenuity Variant Analysis was used to prioritize putative causal variants based on quality control, frequency, and gain or loss of function. The variant frequency was compared in healthy cohorts and in an Alzheimer's disease cohort. Remaining variants were filtered based on their presence in genes reported to have an effect on the aging process, aging of cells, or the longevity process. Validation of these candidate variants included tests of segregation on other elderly relatives. RESULTS Fifteen rare candidate genetic variants spanning 17 genes shared within cousins were identified as having passed prioritization criteria. Of those variants, six were present in genes that are known or predicted to affect the aging process: rs78408340 (PAM), rs112892337 (ZFAT), rs61737629 (ESPL1), rs141903485 (CEBPE), rs144369314 (UTP4), and rs61753103 (NUP88 and RABEP1). ESPL1 rs61737629 and CEBPE rs141903485 show additional evidence of segregation with longevity in expanded pedigree analyses (p-values = .001 and .0001, respectively). DISCUSSION This unique pedigree analysis efficiently identified several novel rare candidate variants that may affect the aging process and added support to seven genes that likely contribute to longevity. Further analyses showed evidence for segregation for two rare variants, ESPL1 rs61737629 and CEBPE rs141903485, in the original longevity pedigrees in which they were initially observed. These candidate genes and variants warrant further investigation.
Collapse
Affiliation(s)
- Justin B Miller
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Elizabeth Ward
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Lyndsay A Staley
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Jeffrey Stevens
- Genetic Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Craig C Teerlink
- Genetic Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Justina P Tavana
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Matthew Cloward
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Madeline Page
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Louisa Dayton
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Lisa A Cannon-Albright
- Genetic Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - John S K Kauwe
- Department of Biology, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
15
|
Mark4 Inhibited the Browning of White Adipose Tissue by Promoting Adipocytes Autophagy in Mice. Int J Mol Sci 2020; 21:ijms21082752. [PMID: 32326642 PMCID: PMC7216233 DOI: 10.3390/ijms21082752] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/09/2020] [Accepted: 04/11/2020] [Indexed: 12/29/2022] Open
Abstract
Autophagy can remove excess or dysfunctional proteins and organelles to maintain cellular homeostasis. Browning of white adipose tissue increases the energy expenditure. Microtubules affinity regulated kinase 4 (Mark4) can regulate a variety of physiological processes. According to previous studies, we speculated that Mark4-autophagy-browning of white adipose tissue had certain linkages. Here, we established two autophagy models through serum starvation and rapamycin treatment and detected that the overexpression of Mark4 increased the expression of autophagy-related factors Beclin1, ATG7, and significantly decreased the autophagy substrate P62. Further tests showed that the overexpression of Mark4 promoted the conversion of autophagy marker protein LC3A to LC3B-II by activating the AMP-activated protein kinase (AMPK) pathway and inhibition of the AKT/mTOR signaling. Moreover, Mark4 decreased the expression of thermogenesis genes via promoting autophagy. These results indicated that Mark4 inhibited the browning of white adipose tissue via promoting autophagy.
Collapse
|
16
|
Naqvi AAT, Jairajpuri DS, Hussain A, Hasan GM, Alajmi MF, Hassan MI. Impact of glioblastoma multiforme associated mutations on the structure and function of MAP/microtubule affinity regulating kinase 4. J Biomol Struct Dyn 2020; 39:1781-1794. [DOI: 10.1080/07391102.2020.1738959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Ahmad Abu Turab Naqvi
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Deeba Shamim Jairajpuri
- Department of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Afzal Hussain
- Department of Pharmacognosy College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
| | - Mohamed F. Alajmi
- Department of Pharmacognosy College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Md. Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
17
|
Li F, Liu Z, Sun H, Li C, Wang W, Ye L, Yan C, Tian J, Wang H. PCC0208017, a novel small-molecule inhibitor of MARK3/MARK4, suppresses glioma progression in vitro and in vivo. Acta Pharm Sin B 2020; 10:289-300. [PMID: 32082974 PMCID: PMC7016295 DOI: 10.1016/j.apsb.2019.09.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/01/2019] [Accepted: 08/06/2019] [Indexed: 01/06/2023] Open
Abstract
Gliomas are the most common primary intracranial neoplasms among all brain malignancies, and the microtubule affinity regulating kinases (MARKs) have become potential drug targets for glioma. Here, we report a novel dual small-molecule inhibitor of MARK3 and MARK4, designated as PCC0208017. In vitro, PCC0208017 strongly inhibited kinase activity against MARK3 and MARK4, and strongly reduced proliferation in three glioma cell lines. This compound attenuated glioma cell migration, glioma cell invasion, and angiogenesis. Molecular mechanism studies revealed that PCC0208017 decreased the phosphorylation of Tau, disrupted microtubule dynamics, and induced a G2/M phase cell cycle arrest. In an in vivo glioma model, PCC0208017 showed robust anti-tumor activity, blood–brain barrier permeability, and a good oral pharmacokinetic profile. Molecular docking studies showed that PCC0208017 exhibited high binding affinity to MARK3 and MARK4. Taken together, our study describes for the first time that PCC0208017, a novel MARK3/MARK4 inhibitor, might be a promising lead compound for treatment of glioma.
Collapse
Affiliation(s)
- Fangfang Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai 264005, China
| | - Zongliang Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai 264005, China
| | - Heyuan Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai 264005, China
| | - Chunmei Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai 264005, China
| | - Wenyan Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai 264005, China
| | - Liang Ye
- Department of Clinical Medicine, Binzhou Medical College, Yantai 256603, China
| | - Chunhong Yan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai 264005, China
- GRU Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Jingwei Tian
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai 264005, China
- Corresponding authors. Tel./fax: +86 535 6706060.
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai 264005, China
- Corresponding authors. Tel./fax: +86 535 6706060.
| |
Collapse
|
18
|
Khan NS, Khan P, Inam A, Ahmad K, Yousuf M, Islam A, Ali S, Azam A, Husain M, Hassan MI. Discovery of 4-(2-(dimethylamino)ethoxy)benzohydrazide derivatives as prospective microtubule affinity regulating kinase 4 inhibitors. RSC Adv 2020; 10:20129-20137. [PMID: 35520423 PMCID: PMC9054212 DOI: 10.1039/d0ra00453g] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/14/2020] [Indexed: 01/03/2023] Open
Abstract
Microtubule affinity regulating kinase 4 (MARK4) is a Ser/Thr kinase, considered as a potential drug target for cancer, diabetes and neurodegenerative diseases. Due to its significant role in the development and progression of cancer, different in-house libraries of synthesized small molecules were screened to identify potential MARK4 inhibitors. A small library of hydrazone compounds showed a considerable binding affinity to MARK4. The selected compounds were further scrutinized using an enzyme inhibition assay and finally two hydrazone derivatives (H4 and H19) were selected that show excellent inhibition (nM range). These compounds have a strong binding affinity for MARK4 and moderate binding with human serum albumin. Anticancer studies were performed on MCF-7 and A549 cells, suggesting H4 and H19 selectively inhibit the growth of cancer cells. The IC50 value of compound H4 and H19 was found to be 27.39 μM and 34.37 μM for MCF-7 cells, while for A549 cells it was 45.24 μM and 61.50 μM, respectively. These compounds inhibited the colonogenic potential of cancer cells and induced apoptosis. Overall findings reflect that hydrazones/hydrazone derivatives could be exploited as potential lead molecules for developing effective anticancer therapies via targeting MARK4. Inhibition studies of MARK4 with selected hydrazone derivatives.![]()
Collapse
Affiliation(s)
| | - Parvez Khan
- Centre for Interdisciplinary Research in Basic Sciences
- Jamia Millia Islamia
- New Delhi 110025
- India
| | - Afreen Inam
- Department of Chemistry
- Jamia Millia Islamia
- New Delhi 110025
- India
| | - Kamal Ahmad
- Centre for Interdisciplinary Research in Basic Sciences
- Jamia Millia Islamia
- New Delhi 110025
- India
| | - Mohd. Yousuf
- Centre for Interdisciplinary Research in Basic Sciences
- Jamia Millia Islamia
- New Delhi 110025
- India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences
- Jamia Millia Islamia
- New Delhi 110025
- India
| | - Sher Ali
- Centre for Interdisciplinary Research in Basic Sciences
- Jamia Millia Islamia
- New Delhi 110025
- India
| | - Amir Azam
- Department of Chemistry
- Jamia Millia Islamia
- New Delhi 110025
- India
| | - Mohammad Husain
- Department of Biotechnology
- Jamia Millia Islamia
- New Delhi 110025
- India
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences
- Jamia Millia Islamia
- New Delhi 110025
- India
| |
Collapse
|
19
|
Hou Y, Shang C, Wang H, Yun J. Isatin-azole hybrids and their anticancer activities. Arch Pharm (Weinheim) 2019; 353:e1900272. [PMID: 31691360 DOI: 10.1002/ardp.201900272] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/14/2019] [Accepted: 10/16/2019] [Indexed: 12/15/2022]
Abstract
Isatin and azole moieties, which have the ability to form various noncovalent interactions with different therapeutic targets, are common pharmacophores in drug development. Isatin and azole derivatives possess promising in vitro and in vivo anticancer activity, and many of them, such as semaxanib, sunitinib, and carboxyamidotriazole, could be used to treat various cancers. Thus, it is conceivable that hybridization of the isatin moiety with azole may provide a valuable therapeutic intervention for the treatment of cancer. Substantial efforts have been made to develop isatin-azole hybrids as novel anticancer agents, and some of the isatin-azole hybrids exhibited considerable activity. This review emphasizes isatin-azole hybrids with potential anticancer activity, covering articles published between 2010 and 2019. The structure-activity relationships as well as the mechanisms of action are also discussed to provide insights for the rational design of more effective candidates.
Collapse
Affiliation(s)
- Yani Hou
- School of Medicine, Xi'an Peihua University, Xi'an, China
| | - Congshan Shang
- School of Medicine, Xi'an Peihua University, Xi'an, China
| | - Hui Wang
- School of Medicine, Xi'an Peihua University, Xi'an, China
| | - Jie Yun
- School of Medicine, Xi'an Peihua University, Xi'an, China
| |
Collapse
|
20
|
Fazilat A, Rashid N, Nigam A, Anjum S, Gupta N, Wajid S. Differential Expression of MARK4 Protein and Related Perturbations in Females with Ovulatory PCOS. Endocr Metab Immune Disord Drug Targets 2019; 19:1064-1074. [DOI: 10.2174/1871530319666190719145823] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 04/22/2019] [Accepted: 05/16/2019] [Indexed: 12/25/2022]
Abstract
Background:
Ovulatory PCOS (OPCOS) is the mildest form of the polycystic ovarian syndrome
among all four determined phenotypes. Though the females with OPCOS are ovulating, hyperandrogenism
and polycystic ovarian morphology increase the susceptibility of cardiovascular diseases,
insulin resistance, hyperlipidemia and metabolic syndrome in these females.
Objectives:
The aim of the study was to identify the significance associated with OPCOS phenotype
through serum proteomic profiling of OPCOS females and normal age-matched healthy ovulating females.
Methods:
One and two-dimensional gel-based proteomic approaches were adopted to fractionate the
complex serum proteome. Differential protein profiles generated were analyzed with PD-QUEST
Software. Protein spots differing in intensity by >2-fold were selected and identified further by
MALDI-TOF MS. Validation of identified protein was carried out by Biolayer Interferometry.
Results:
One and two-dimensional gel profiles revealed a differential expression pattern of proteins. 10
selected spots were identified as GMP synthase [glutamine hydrolyzing], zinc finger protein 518A,
pericentriolar material 1 protein, BCLAF1 and THRAP3 family member 3, MAP/microtubule affinityregulating
kinase 4, H/ACA ribonucleoprotein complex subunit 1, Melanoma-associated antigen B3
and Zinc finger protein 658B. Expression of MAP/microtubule affinity-regulating kinase 4 (MARK4)
was found to be downregulated in OPCOS females as compared to controls on validation.
Conclusion:
Reduced expression of MARK4 protein in OPCOS increases the associated risk of hyperlipidemia,
hyperandrogenism and metabolic syndrome, thus the protein holds strong candidature as a
drug target for the syndrome.
Collapse
Affiliation(s)
- Ahmad Fazilat
- Department of Biotechnology, School of Chemical and Life Sciences (SCLS), Jamia Hamdard, New Delhi, 110062, India
| | - Nadia Rashid
- Department of Biotechnology, School of Chemical and Life Sciences (SCLS), Jamia Hamdard, New Delhi, 110062, India
| | - Aruna Nigam
- Department of Gynaecology and Obstetrics, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Shadab Anjum
- Department of Biotechnology, School of Chemical and Life Sciences (SCLS), Jamia Hamdard, New Delhi, 110062, India
| | - Nimisha Gupta
- Department of Biotechnology, School of Chemical and Life Sciences (SCLS), Jamia Hamdard, New Delhi, 110062, India
| | - Saima Wajid
- Department of Biotechnology, School of Chemical and Life Sciences (SCLS), Jamia Hamdard, New Delhi, 110062, India
| |
Collapse
|
21
|
Naqvi AAT, Jairajpuri DS, Noman OMA, Hussain A, Islam A, Ahmad F, Alajmi MF, Hassan MI. Evaluation of pyrazolopyrimidine derivatives as microtubule affinity regulating kinase 4 inhibitors: Towards therapeutic management of Alzheimer’s disease. J Biomol Struct Dyn 2019; 38:3892-3907. [DOI: 10.1080/07391102.2019.1666745] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Ahmad Abu Turab Naqvi
- Center for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, New Delhi, India
| | - Deeba Shamim Jairajpuri
- Department of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Omar Mohammed Ali Noman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Asimul Islam
- Center for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, New Delhi, India
| | - Faizan Ahmad
- Center for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, New Delhi, India
| | - Mohammed F. Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Md. Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
22
|
Khan P, Queen A, Mohammad T, Khan NS, Hafeez ZB, Hassan MI, Ali S. Identification of α-Mangostin as a Potential Inhibitor of Microtubule Affinity Regulating Kinase 4. JOURNAL OF NATURAL PRODUCTS 2019; 82:2252-2261. [PMID: 31343173 DOI: 10.1021/acs.jnatprod.9b00372] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Microtubule affinity regulating kinase 4 (MARK4) is a potential drug target for neuronal disorders and several types of cancers. Filtration of naturally occurring compound libraries using high-throughput screening and enzyme assay suggest α-mangostin is a potential inhibitor of MARK4. Structure-based docking and 100 ns molecular dynamics simulation revealed that the binding of α-mangostin stabilizes the MARK4 structure. Enzyme inhibition and binding studies showed that α-mangostin inhibited MARK4 in the submicromolar range with IC50 = 1.47 μM and binding constant (Ka) 5.2 × 107 M-1. Cell-based studies suggested that α-mangostin inhibited the cell viability (MCF-7 and HepG2), induced apoptosis, arrested the cell cycle in the G0/G1 phase, and reduced tau-phosphorylation. This study implicates MARK4 as a new target of α-mangostin, adding an additional lead molecule to the anticancer repertoire.
Collapse
Affiliation(s)
- Parvez Khan
- Centre for Interdisciplinary Research in Basic Sciences , Jamia Millia Islamia , New Delhi 110025 , India
| | - Aarfa Queen
- Centre for Interdisciplinary Research in Basic Sciences , Jamia Millia Islamia , New Delhi 110025 , India
- Department of Chemistry , Jamia Millia Islamia , Jamia Nagar , New Delhi 110025 , India
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences , Jamia Millia Islamia , New Delhi 110025 , India
| | - Nashrah Sharif Khan
- Centre for Interdisciplinary Research in Basic Sciences , Jamia Millia Islamia , New Delhi 110025 , India
| | - Zubair Bin Hafeez
- Department of Biosciences , Jamia Millia Islamia , Jamia Nagar , New Delhi 110025 , India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences , Jamia Millia Islamia , New Delhi 110025 , India
| | - Sher Ali
- Centre for Interdisciplinary Research in Basic Sciences , Jamia Millia Islamia , New Delhi 110025 , India
| |
Collapse
|
23
|
Clement M, Chen X, Chenoweth HL, Teng Z, Thome S, Newland SA, Harrison J, Yu X, Finigan AJ, Mallat Z, Li X. MARK4 (Microtubule Affinity-Regulating Kinase 4)-Dependent Inflammasome Activation Promotes Atherosclerosis—Brief Report. Arterioscler Thromb Vasc Biol 2019; 39:1645-1651. [DOI: 10.1161/atvbaha.119.312478] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Objective:
MARK4 (microtubule affinity-regulating kinase 4) regulates NLRP3 (nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 3) inflammasome activation. The aim of the study is to examine the role of MARK4 in hematopoietic cells during atherosclerosis.
Methods and Results:
We show increased MARK4 expression in human atherosclerotic lesions compared with adjacent areas. MARK4 is coexpressed with NLRP3, and they colocalize in areas enriched in CD68-positive but α-SMA (α-smooth muscle actin)–negative cells. Expression of MARK4 and NLRP3 in the atherosclerotic lesions is associated with the production of active IL (interleukin)-1β and IL-18. To directly assess the role of hematopoietic MARK4 in NLRP3 inflammasome activation and atherosclerotic plaque formation,
Ldlr
(low-density lipoprotein receptor)-deficient mice were lethally irradiated and reconstituted with either wild-type or
Mark4
-deficient bone marrow cells, and were subsequently fed a high-fat diet and cholesterol diet for 9 weeks.
Mark4
deficiency in bone marrow cells led to a significant reduction of lesion size, together with decreased circulating levels of IL-18 and IFN-γ (interferon-γ). Furthermore,
Mark4
deficiency in primary murine bone marrow–derived macrophages prevented cholesterol crystal–induced NLRP3 inflammasome activation, as revealed by reduced caspase-1 activity together with reduced production of IL-1β and IL-18.
Conclusions:
MARK4-dependent NLRP3 inflammasome activation in the hematopoietic cells regulates the development of atherosclerosis.
Collapse
Affiliation(s)
- Marc Clement
- From the Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (M.C., H.L.C., S.T., S.A.N., J.H., X.Y., A.J.F., Z.M., X.L.)
| | - Xiao Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China (X.C.)
| | - Hannah L. Chenoweth
- From the Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (M.C., H.L.C., S.T., S.A.N., J.H., X.Y., A.J.F., Z.M., X.L.)
| | - Zhongzhao Teng
- Department of Radiology, University of Cambridge, United Kingdom (Z.T.)
| | - Sarah Thome
- From the Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (M.C., H.L.C., S.T., S.A.N., J.H., X.Y., A.J.F., Z.M., X.L.)
| | - Stephen A. Newland
- From the Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (M.C., H.L.C., S.T., S.A.N., J.H., X.Y., A.J.F., Z.M., X.L.)
| | - James Harrison
- From the Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (M.C., H.L.C., S.T., S.A.N., J.H., X.Y., A.J.F., Z.M., X.L.)
| | - Xian Yu
- From the Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (M.C., H.L.C., S.T., S.A.N., J.H., X.Y., A.J.F., Z.M., X.L.)
| | - Alison J. Finigan
- From the Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (M.C., H.L.C., S.T., S.A.N., J.H., X.Y., A.J.F., Z.M., X.L.)
| | - Ziad Mallat
- From the Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (M.C., H.L.C., S.T., S.A.N., J.H., X.Y., A.J.F., Z.M., X.L.)
- Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, France (Z.M.)
| | - Xuan Li
- From the Division of Cardiovascular Medicine, University of Cambridge, United Kingdom (M.C., H.L.C., S.T., S.A.N., J.H., X.Y., A.J.F., Z.M., X.L.)
| |
Collapse
|
24
|
Jenardhanan P, Panneerselvam M, Mathur PP. Targeting Kinase Interaction Networks: A New Paradigm in PPI Based Design of Kinase Inhibitors. Curr Top Med Chem 2019; 19:467-485. [PMID: 31184298 DOI: 10.2174/1568026619666190304155711] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 01/20/2019] [Accepted: 02/06/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Kinases are key modulators in regulating diverse range of cellular activities and are an essential part of the protein-protein interactome. Understanding the interaction of kinases with different substrates and other proteins is vital to decode the cell signaling machinery as well as causative mechanism for disease onset and progression. OBJECTIVE The objective of this review is to present all studies on the structure and function of few important kinases and highlight the protein-protein interaction (PPI) mechanism of kinases and the kinase specific interactome databases and how such studies could be utilized to develop anticancer drugs. METHODS The article is a review of the detailed description of the various domains in kinases that are involved in protein-protein interactions and specific inhibitors developed targeting these PPI domains. RESULTS The review has surfaced in depth the interacting domains in key kinases and their features and the roles of PPI in the human kinome and the various signaling cascades that are involved in certain types of cancer. CONCLUSION The insight availed into the mechanism of existing peptide inhibitors and peptidomimetics against kinases will pave way for the design and generation of domain specific peptide inhibitors with better productivity and efficiency and the various software and servers available can be of great use for the identification and analysis of protein-protein interactions.
Collapse
Affiliation(s)
| | - Manivel Panneerselvam
- Department of Biotechnology, BJM School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Premendu P Mathur
- Department of Biochemistry & Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, India
| |
Collapse
|
25
|
Shen X, Liu X, Wan S, Fan X, He H, Wei R, Pu W, Peng Y, Wang C. Discovery of Coumarin as Microtubule Affinity-Regulating Kinase 4 Inhibitor That Sensitize Hepatocellular Carcinoma to Paclitaxel. Front Chem 2019; 7:366. [PMID: 31179271 PMCID: PMC6543911 DOI: 10.3389/fchem.2019.00366] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 05/03/2019] [Indexed: 02/05/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent cancers worldwide. Nowadays, pharmacological therapy for HCC is in urgent needs. Paclitaxel is an effective drug against diverse solid tumors, but commonly resisted in HCC patients. We recently have disclosed that microtubule affinity-regulating kinase 4 (MARK4) increases the microtubule dynamics and confers paclitaxel resistance in HCC, suggesting MARK4 as an attractive target to overcome paclitaxel resistance. Herein, we synthesized and identified coumarin derivatives 50 as a novel MARK4 inhibitor. Biological evaluation indicated compound 50 directly interacted with MARK4 and inhibited its activity in vitro, suppressed cell viability and induced apoptosis of HCC cells in a MARK4-dependent manner. Importantly, compound 50 significantly increased the drug response of paclitaxel treatment to HCC cells, providing a promise strategy to HCC treatment and broadening the application of paclitaxel in cancer therapy.
Collapse
Affiliation(s)
- Xianyan Shen
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Xuesha Liu
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and College of Life Sciences, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Shunli Wan
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Xin Fan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and College of Life Sciences, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Huaiyu He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and College of Life Sciences, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Rong Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and College of Life Sciences, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Wenchen Pu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and College of Life Sciences, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and College of Life Sciences, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Chun Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| |
Collapse
|
26
|
Voura M, Khan P, Thysiadis S, Katsamakas S, Queen A, Hasan GM, Ali S, Sarli V, Hassan MI. Probing the Inhibition of Microtubule Affinity Regulating Kinase 4 by N-Substituted Acridones. Sci Rep 2019; 9:1676. [PMID: 30737440 PMCID: PMC6368574 DOI: 10.1038/s41598-018-38217-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 11/16/2018] [Indexed: 12/18/2022] Open
Abstract
Microtubule affinity regulating kinase 4 (MARK4) becomes a unique anti-cancer drug target as its overexpression is responsible for different types of cancers. In quest of novel, effective MARK4 inhibitors, some acridone derivatives were synthesized, characterized and evaluated for inhibitory activity against human MARK4. Among all the synthesized compounds, three (7b, 7d and 7f) were found to have better binding affinity and enzyme inhibition activity in µM range as shown by fluorescence binding, ITC and kinase assays. Here we used functional assays of selected potential lead molecules with commercially available panel of 26 kinases of same family. A distinctive kinase selectivity profile was observed for each compound. The selective compounds were identified with submicromolar cellular activity against MARK4. Furthermore, in vitro antitumor evaluation against cancerous cells (MCF-7 and HepG2) revealed that compounds 7b, 7d and 7f inhibit cell proliferation and predominantly induce apoptosis in MCF-7 cells, with IC50 values of 5.2 ± 1.2 μM, 6.3 ± 1.2 μM, and 5.8 ± 1.4 μM respectively. In addition, these compounds significantly upsurge the oxidative stress in cancerous cells. Our observations support our approach for the synthesis of effective inhibitors against MARK4 that can be taken forward for the development of novel anticancer molecules targeting MARK4.
Collapse
Affiliation(s)
- Maria Voura
- Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| | - Parvez Khan
- Centre for interdisciplinary research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Savvas Thysiadis
- Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| | - Sotiris Katsamakas
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| | - Aarfa Queen
- Centre for interdisciplinary research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
- Department of Chemistry, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj, 11942, Saudi Arabia
| | - Sher Ali
- Centre for interdisciplinary research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Vasiliki Sarli
- Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece.
| | - Md Imtaiyaz Hassan
- Centre for interdisciplinary research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India.
| |
Collapse
|
27
|
Design and development of Isatin-triazole hydrazones as potential inhibitors of microtubule affinity-regulating kinase 4 for the therapeutic management of cell proliferation and metastasis. Eur J Med Chem 2019; 163:840-852. [DOI: 10.1016/j.ejmech.2018.12.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/04/2018] [Accepted: 12/12/2018] [Indexed: 12/23/2022]
|
28
|
Annadurai N, Agrawal K, Džubák P, Hajdúch M, Das V. Microtubule affinity-regulating kinases are potential druggable targets for Alzheimer's disease. Cell Mol Life Sci 2017; 74:4159-4169. [PMID: 28634681 PMCID: PMC11107647 DOI: 10.1007/s00018-017-2574-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 06/13/2017] [Accepted: 06/15/2017] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects normal functions of the brain. Currently, AD is one of the leading causes of death in developed countries and the only one of the top ten diseases without a means to prevent, cure, or significantly slow down its progression. Therefore, newer therapeutic concepts are urgently needed to improve survival and the quality of life of AD patients. Microtubule affinity-regulating kinases (MARKs) regulate tau-microtubule binding and play a crucial role in neurons. However, their role in hyperphosphorylation of tau makes them potential druggable target for AD therapy. Despite the relevance of MARKs in AD pathogenesis, only a few small molecules are known to have anti-MARK activity and not much has been done to progress these compounds into therapeutic candidates. But given the diverse role of MARKs, the specificity of novel inhibitors is imperative for their successful translation from bench to bedside. In this regard, a recent co-crystal structure of MARK4 in association with a pyrazolopyrimidine-based inhibitor offers a potential scaffold for the development of more specific MARK inhibitors. In this manuscript, we review the biological role of MARKs in health and disease, and draw attention to the largely unexplored area of MARK inhibitors for AD.
Collapse
Affiliation(s)
- Narendran Annadurai
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 77900, Olomouc, Czech Republic
| | - Khushboo Agrawal
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 77900, Olomouc, Czech Republic
| | - Petr Džubák
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 77900, Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 77900, Olomouc, Czech Republic
| | - Viswanath Das
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 77900, Olomouc, Czech Republic.
| |
Collapse
|
29
|
Naz F, Khan FI, Mohammad T, Khan P, Manzoor S, Hasan GM, Lobb KA, Luqman S, Islam A, Ahmad F, Hassan MI. Investigation of molecular mechanism of recognition between citral and MARK4: A newer therapeutic approach to attenuate cancer cell progression. Int J Biol Macromol 2017; 107:2580-2589. [PMID: 29079437 DOI: 10.1016/j.ijbiomac.2017.10.143] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/22/2017] [Accepted: 10/23/2017] [Indexed: 10/18/2022]
Abstract
Microtubule affinity regulating kinase 4 (MARK4) is a member of AMP-activated protein kinase, found to be involved in apoptosis, inflammation and many other regulatory pathways. Since, its aberrant expression is directly associated with the cell cycle and thus cancer. Therefore, MARK4 is being considered as a potential drug target for cancer therapy. Here, we investigated the mechanism of inhibition of MARK4 activity by citral. Docking studies suggested that citral effectively binds to the active site cavity, and complex is stabilized by several interactions. We further performed molecular dynamics simulation of MARK4-citral complex under explicit water condition for 100ns and observed that binding of citral to MARK4 was quite stable. Fluorescence binding studies suggested that citral strongly binds to MARK4 and thereby inhibits its enzyme activity which was measured by the kinase inhibition assay. We further performed MTT assay and observed that citral inhibits proliferation of breast cancer cell line MCF-7. This work provides a newer insight into the use of citral as novel cancer therapeutics through the MARK4 inhibition. Results may be employed to design novel therapeutic molecule using citral as a scaffold for MARK4 inhibition to fight related diseases.
Collapse
Affiliation(s)
- Farha Naz
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India; Department of Molecular & Cellular Engineering, Sam Higginbottom Institute of Agriculture, Technology and Sciences, Allahabad, 211 007, Uttar Pradesh, India
| | - Faez Iqbal Khan
- Department of Chemistry, Rhodes University, Grahamstown 6139, South Africa
| | - Taj Mohammad
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Parvez Khan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Saaliqa Manzoor
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Kevin A Lobb
- Department of Chemistry, Rhodes University, Grahamstown 6139, South Africa
| | - Suaib Luqman
- CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, India
| | - Asimul Islam
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Faizan Ahmad
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India.
| |
Collapse
|
30
|
Khan P, Rahman S, Queen A, Manzoor S, Naz F, Hasan GM, Luqman S, Kim J, Islam A, Ahmad F, Hassan MI. Elucidation of Dietary Polyphenolics as Potential Inhibitor of Microtubule Affinity Regulating Kinase 4: In silico and In vitro Studies. Sci Rep 2017; 7:9470. [PMID: 28842631 PMCID: PMC5573368 DOI: 10.1038/s41598-017-09941-4] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 08/01/2017] [Indexed: 02/08/2023] Open
Abstract
Microtubule affinity regulating kinase 4 (MARK4) is a Ser/Thr kinase belonging to AMPK-like family, has recently become an important drug target against cancer and neurodegenerative disorders. In this study, we have evaluated different natural dietary polyphenolics including rutin, quercetin, ferulic acid, hesperidin, gallic acid and vanillin as MARK4 inhibitors. All compounds are primarily binds to the active site cavity of MARK4. In silico observations were further complemented by the fluorescence-binding studies and isothermal titration calorimetry (ITC) measurements. We found that rutin and vanillin bind to MARK4 with a reasonably high affinity. ATPase and tau-phosphorylation assay further suggesting that rutin and vanillin inhibit the enzyme activity of MARK4 to a great extent. Cell proliferation, ROS quantification and Annexin-V staining studies are clearly providing sufficient evidences for the apoptotic potential of rutin and vanillin. In conclusion, rutin and vanillin may be considered as potential inhibitors for MARK4 and further exploited to design novel therapeutic molecules against MARK4 associated diseases.
Collapse
Affiliation(s)
- Parvez Khan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Shafikur Rahman
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 712-749, South Korea
| | - Aarfa Queen
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Saaliqa Manzoor
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Farha Naz
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Suaib Luqman
- CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, India
| | - Jihoe Kim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 712-749, South Korea
| | - Asimul Islam
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Faizan Ahmad
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India.
| |
Collapse
|
31
|
MARK4 regulates NLRP3 positioning and inflammasome activation through a microtubule-dependent mechanism. Nat Commun 2017; 8:15986. [PMID: 28656979 PMCID: PMC5493753 DOI: 10.1038/ncomms15986] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/17/2017] [Indexed: 12/17/2022] Open
Abstract
Excessive activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome is involved in many chronic inflammatory diseases, including cardiovascular and Alzheimer's disease. Here we show that microtubule-affinity regulating kinase 4 (MARK4) binds to NLRP3 and drives it to the microtubule-organizing centre, enabling the formation of one large inflammasome speck complex within a single cell. MARK4 knockdown or knockout, or disruption of MARK4-NLRP3 interaction, impairs NLRP3 spatial arrangement and limits inflammasome activation. Our results demonstrate how an evolutionarily conserved protein involved in the regulation of microtubule dynamics orchestrates NLRP3 inflammasome activation by controlling its transport to optimal activation sites, and identify a targetable function for MARK4 in the control of innate immunity.
Collapse
|
32
|
Liu Z, Gan L, Chen Y, Luo D, Zhang Z, Cao W, Zhou Z, Lin X, Sun C. Mark4 promotes oxidative stress and inflammation via binding to PPARγ and activating NF-κB pathway in mice adipocytes. Sci Rep 2016; 6:21382. [PMID: 26888669 PMCID: PMC4766853 DOI: 10.1038/srep21382] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 01/22/2016] [Indexed: 12/25/2022] Open
Abstract
MAP/Microtubule affinity-regulating kinase 4 (Mark4) plays an important role in the regulation of microtubule organization, adipogenesis and apoptosis. However, the role of Mark4 plays in oxidative stress and inflammation are poorly understood. In this study, we found Mark4 was induced by high fat diet (HFD) while PPARγ was elevated significantly in mice adipocytes. Further analyses revealed Mark4 impaired mitochondrial oxidative respiration and increased reactive oxygen species (ROS) production. At same time, the activities of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) were greatly reduced. By treating cells with H2O2 and vitamin E (VE), Mark4 accentuated oxidative stress along with increased mRNA level of inflammatory factor interleukin-6 (IL-6) and decreased leptin mRNA. Furthermore, we found PPARγ bind to Mark4 promoter region and inhibited Mark4 expression. We showed PPARγ interacted with Mark4 and inhibited the stimulating effect of Mark4 on oxidative stress and inflammation. Finally, we demonstrated that the IKKα/NF-κB signal pathway was involved in Mark4 induced oxidative stress and inflammation, while PTDC, a special inhibitor of NF-κB signal pathway, reduced oxidative stress and inflammation. Thus, our study indicated that Mark4 was a potential drug target for treating metabolic diseases.
Collapse
Affiliation(s)
- Zhenjiang Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Lu Gan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yizhe Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Dan Luo
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zhenzhen Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Weina Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zhongjie Zhou
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xueting Lin
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| |
Collapse
|
33
|
Tang EI, Mruk DD, Cheng CY. Regulation of microtubule (MT)-based cytoskeleton in the seminiferous epithelium during spermatogenesis. Semin Cell Dev Biol 2016; 59:35-45. [PMID: 26791048 DOI: 10.1016/j.semcdb.2016.01.004] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 01/05/2016] [Indexed: 12/31/2022]
Abstract
In rodents and humans, testicular cells, similar to other mammalian cells, are supported by actin-, microtubule (MT)- and intermediate filament-based cytoskeletons. Although the cytoskeletal network of the testis serves an important role in regulating spermatogenesis during the epithelial cycle, most of the published findings in the literature are limited to studies that only visualize these cytoskeletons in the seminiferous epithelium. Few focus on the underlying molecular mechanism that regulates their organization in the epithelium in response to changes in the stages of the epithelial cycle. Functional studies in the last decade have begun to focus on the role of binding proteins that regulate these cytoskeletons, with some interesting findings rapidly emerging in the field. Since the actin- and intermediate filament-based cytoskeletons have been recently reviewed, herein we focus on the MT-based cytoskeleton for two reasons. First, besides serving as a structural support cytoskeleton, MTs are known to serve as the track to support and facilitate the transport of germ cells, such as preleptotene spermatocytes connected in clones and elongating/elongated spermatids during spermiogenesis, across the blood-testis barrier (BTB) and the adluminal compartment, respectively, during spermatogenesis. While these cellular events are crucial to the completion of spermatogenesis, they have been largely ignored in the past. Second, MT-based cytoskeleton is working in concert with the actin-based cytoskeleton to provide structural support for the transport of intracellular organelles across the cell cytosol, such as endosome-based vesicles, and phagosomes, which contain residual bodies detached from spermatids, to maintain the cellular homeostasis in the seminiferous epithelium. We critically evaluate some recent published findings herein to support a hypothesis regarding the role of MT in conferring germ cell transport in the seminiferous epithelium.
Collapse
Affiliation(s)
- Elizabeth I Tang
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, United States
| | - Dolores D Mruk
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, United States
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, United States.
| |
Collapse
|
34
|
Coles GL, Baglia LA, Ackerman KG. KIF7 Controls the Proliferation of Cells of the Respiratory Airway through Distinct Microtubule Dependent Mechanisms. PLoS Genet 2015; 11:e1005525. [PMID: 26439735 PMCID: PMC4595342 DOI: 10.1371/journal.pgen.1005525] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 08/24/2015] [Indexed: 12/12/2022] Open
Abstract
The cell cycle must be tightly coordinated for proper control of embryonic development and for the long-term maintenance of organs such as the lung. There is emerging evidence that Kinesin family member 7 (Kif7) promotes Hedgehog (Hh) signaling during embryonic development, and its misregulation contributes to diseases such as ciliopathies and cancer. Kif7 encodes a microtubule interacting protein that controls Hh signaling through regulation of microtubule dynamics within the primary cilium. However, whether Kif7 has a function in nonciliated cells remains largely unknown. The role Kif7 plays in basic cell biological processes like cell proliferation or cell cycle progression also remains to be elucidated. Here, we show that Kif7 is required for coordination of the cell cycle, and inactivation of this gene leads to increased cell proliferation in vivo and in vitro. Immunostaining and transmission electron microscopy experiments show that Kif7dda/dda mutant lungs are hyperproliferative and exhibit reduced alveolar epithelial cell differentiation. KIF7 depleted C3H10T1/2 fibroblasts and Kif7dda/dda mutant mouse embryonic fibroblasts have increased growth rates at high cellular densities, suggesting that Kif7 may function as a general regulator of cellular proliferation. We ascertained that in G1, Kif7 and microtubule dynamics regulate the expression and activity of several components of the cell cycle machinery known to control entry into S phase. Our data suggest that Kif7 may function to regulate the maintenance of the respiratory airway architecture by controlling cellular density, cell proliferation, and cycle exit through its role as a microtubule associated protein. Respiratory diseases such as lung cancer, COPD, and asthma are the second leading cause of death in the United States. These diseases are heterogeneous and arise from genetic factors, environmental hazards, or developmental abnormalities that persist throughout life. An increased understanding of the genes and cellular mechanisms regulating respiratory system homeostasis and regeneration should provide information for the development of future therapeutics. We show that the gene Kif7 regulates cell proliferation, cellular density, and intracellular signaling within the epithelial and mesenchymal cells of the respiratory airway. We expand on the known role for Kif7 in regulating microtubule architecture within ciliated cells by showing that this protein regulates cell signaling in non-ciliated secretory cells. Furthermore, we show that microtubules function to regulate the abundance and activity of several factors known to be required for proper cell cycle timing. We propose that Kif7 and microtubule dynamics hone cellular signaling necessary for control of the balance between cell proliferation and cell cycle exit, and we provide evidence that Kif7 has a critical role in the maintenance of the respiratory system in postnatal life.
Collapse
Affiliation(s)
- Garry L. Coles
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Laurel A. Baglia
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States of America
- Center for Pediatric Biomedical Research, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Kate G. Ackerman
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States of America
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States of America
- Center for Pediatric Biomedical Research, University of Rochester Medical Center, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
35
|
Fontana L, Rovina D, Novielli C, Maffioli E, Tedeschi G, Magnani I, Larizza L. Suggestive evidence on the involvement of polypyrimidine-tract binding protein in regulating alternative splicing of MAP/microtubule affinity-regulating kinase 4 in glioma. Cancer Lett 2015; 359:87-96. [PMID: 25578778 DOI: 10.1016/j.canlet.2014.12.049] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 12/23/2014] [Accepted: 12/26/2014] [Indexed: 01/01/2023]
Abstract
MAP/microtubule affinity-regulating kinase 4 (MARK4) is a serine-threonine kinase that phosphorylates microtubule-associated proteins taking part in the regulation of microtubule dynamics. MARK4 is expressed in two spliced isoforms characterized by inclusion (MARK4S) or exclusion (MARK4L) of exon 16. The distinct expression profiles in the central nervous system and their imbalance in gliomas point to roles of MARK4L and MARK4S in cell proliferation and cell differentiation, respectively. Having ruled out mutations and transcription defects, we hypothesized that alterations in the expression of splicing factors may underlie deregulated MARK4 expression in gliomas. Bioinformatic analysis revealed four putative polypyrimidine-tract binding (PTB) protein binding sites in MARK4 introns 15 and 16. Glioma tissues and glioblastoma-derived cancer stem cells showed, compared with normal brain, significant overexpression of PTB, correlated with high MARK4L mRNA expression. Splicing minigene assays revealed a functional intronic splicing silencer in MARK4 intron 15, but mutagenesis of the PTB binding site in this region did not affect minigene splicing, suggesting that PTB may bind to a splicing silencer other than the predicted one and synergistically acting with the other predicted PTB sites. Electrophoretic mobility shift assays coupled with mass spectrometry confirmed binding of PTB to the polypyrimidine tract of intron 15, and thus its involvement in MARK4 alternative splicing. This finding, along with evidence of PTB overexpression in gliomas and glioblastoma-derived cancer stem cells and differentiated progeny, merged in pointing out the involvement of PTB in the switch to MARK4L, consistent with its established role in driving oncogenic splicing in brain tumors.
Collapse
Affiliation(s)
- L Fontana
- Department of Health Sciences, Medical Genetics, Università degli Studi di Milano, via Antonio di Rudinì 8, 20142 Milan, Italy
| | - D Rovina
- Department of Health Sciences, Medical Genetics, Università degli Studi di Milano, via Antonio di Rudinì 8, 20142 Milan, Italy
| | - C Novielli
- Department of Health Sciences, Medical Genetics, Università degli Studi di Milano, via Antonio di Rudinì 8, 20142 Milan, Italy
| | - E Maffioli
- Department of Animal Pathology, Hygiene and Veterinary Public Health, Università degli Studi di Milano, Via Celoria, 10, 20133 Milan, Italy; Fondazione Filarete, Viale Ortles 22/4, 20139 Milan, Italy
| | - G Tedeschi
- Department of Animal Pathology, Hygiene and Veterinary Public Health, Università degli Studi di Milano, Via Celoria, 10, 20133 Milan, Italy; Fondazione Filarete, Viale Ortles 22/4, 20139 Milan, Italy
| | - I Magnani
- Department of Health Sciences, Medical Genetics, Università degli Studi di Milano, via Antonio di Rudinì 8, 20142 Milan, Italy
| | - L Larizza
- Department of Health Sciences, Medical Genetics, Università degli Studi di Milano, via Antonio di Rudinì 8, 20142 Milan, Italy; Laboratory of Medical Cytogenetics and Molecular Genetics, IRCCS Istituto Auxologico Italiano, Via Zucchi, 18, 20095 Cusano Milanino, Italy.
| |
Collapse
|