1
|
Mirzaiebadizi A, Shafabakhsh R, Ahmadian MR. Modulating PAK1: Accessory Proteins as Promising Therapeutic Targets. Biomolecules 2025; 15:242. [PMID: 40001545 PMCID: PMC11852631 DOI: 10.3390/biom15020242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
The p21-activated kinase (PAK1), a serine/threonine protein kinase, is critical in regulating various cellular processes, including muscle contraction, neutrophil chemotaxis, neuronal polarization, and endothelial barrier function. Aberrant PAK1 activity has been implicated in the progression of several human diseases, including cancer, heart disease, and neurological disorders. Increased PAK1 expression is often associated with poor clinical prognosis, invasive tumor characteristics, and therapeutic resistance. Despite its importance, the cellular mechanisms that modulate PAK1 function remain poorly understood. Accessory proteins, essential for the precise assembly and temporal regulation of signaling pathways, offer unique advantages as therapeutic targets. Unlike core signaling components, these modulators can attenuate aberrant signaling without completely abolishing it, potentially restoring signaling to physiological levels. This review highlights PAK1 accessory proteins as promising and novel therapeutic targets, opening new horizons for disease treatment.
Collapse
Affiliation(s)
- Amin Mirzaiebadizi
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Rana Shafabakhsh
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany;
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| |
Collapse
|
2
|
Wang S, Castro BA, Katz JL, Arrieta V, Najem H, Vazquez-Cervantes GI, Wan H, Olson IE, Hou D, Dapash M, Billingham LK, Chia TY, Wei C, Rashidi A, Platanias LC, McCortney K, Horbinski CM, Stupp R, Zhang P, Ahmed AU, Sonabend AM, Heimberger AB, Lesniak MS, Riviere-Cazaux C, Burns T, Miska J, Fischietti M, Lee-Chang C. B cell-based therapy produces antibodies that inhibit glioblastoma growth. J Clin Invest 2024; 134:e177384. [PMID: 39207859 PMCID: PMC11473152 DOI: 10.1172/jci177384] [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: 12/01/2023] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Glioblastoma (GBM) is a highly aggressive and malignant brain tumor with limited therapeutic options and a poor prognosis. Despite current treatments, the invasive nature of GBM often leads to recurrence. A promising alternative strategy is to harness the potential of the immune system against tumor cells. Our previous data showed that the BVax (B cell-based vaccine) can induce therapeutic responses in preclinical models of GBM. In this study, we aimed to characterize the antigenic reactivity of BVax-derived Abs and evaluate their therapeutic potential. We performed immunoproteomics and functional assays in murine models and samples from patients with GBM. Our investigations revealed that BVax distributed throughout the GBM tumor microenvironment and then differentiated into Ab-producing plasmablasts. Proteomics analyses indicated that the Abs produced by BVax had unique reactivity, predominantly targeting factors associated with cell motility and the extracellular matrix. Crucially, these Abs inhibited critical processes such as GBM cell migration and invasion. These findings provide valuable insights into the therapeutic potential of BVax-derived Abs for patients with GBM, pointing toward a novel direction for GBM immunotherapy.
Collapse
Affiliation(s)
- Si Wang
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Brandyn A. Castro
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Neurological Surgery, University of Chicago Medicine, Chicago, Illinois, USA
| | - Joshua L. Katz
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Victor Arrieta
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Hinda Najem
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Gustavo I. Vazquez-Cervantes
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Hanxiao Wan
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Ian E. Olson
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - David Hou
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mark Dapash
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Leah K. Billingham
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Tzu-yi Chia
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Chao Wei
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Aida Rashidi
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Leonidas C. Platanias
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
- Department of Medicine, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Kathleen McCortney
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Craig M. Horbinski
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Roger Stupp
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Peng Zhang
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Atique U. Ahmed
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Adam M. Sonabend
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Amy B. Heimberger
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Maciej S. Lesniak
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | | | - Terry Burns
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesotta, USA
| | - Jason Miska
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Mariafausta Fischietti
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
- Department of Medicine, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Catalina Lee-Chang
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| |
Collapse
|
3
|
Zheng R, He Y, Yang L, Chen Y, Wang R, Xie S. Nischarin inhibits the epithelial-mesenchymal transition process and angiogenesis in breast cancer cells by inactivating FAK/ERK signaling pathway via EGF like repeats and discoidin domains 3. Mol Biol Rep 2024; 51:821. [PMID: 39023636 DOI: 10.1007/s11033-024-09776-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/02/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Our previous study has demonstrated that Nischarin (NISCH) exerts its antitumor effects in breast cancer (BC) by suppressing cell migration and invasion. This study aims to explore the underlying mechanism through which NISCH functions in BC. METHODS AND RESULTS The relevance between EGF Like Repeats and Discoidin Domains 3 (EDIL3) mRNA expression and the overall survival of tumor patients was depicted by the Kaplan-Meier curve. The findings revealed that overexpressed NISCH attenuated cell motility and colony-forming capacities of Hs578T cells, yet silenced NISCH in MDA-MB-231 cells led to contrasting results. Western blot (WB) analysis indicated that overexpression of NISCH significantly down-regulated the Vimentin and Slug expression, and inactivated the FAK/ERK signaling pathway. RNA sequencing (RNA-seq) was performed in NISCH-overexpressed Hs578T cells and the control cells to analyze differentially expressed genes (DeGs), and the results showed a significant down-regulation of EDIL3 mRNA level upon overexpression of NISCH. Subsequent functional analyses demonstrated that overexpression of EDIL3 attenuated the inhibitory effect of NISCH on cell migration, invasion, colony formation, and tube formation. CONCLUSION In summary, our finding preliminarily revealed that NISCH inhibits the epithelial-mesenchymal transition (EMT) process and angiogenesis in BC cells by down-regulating EDIL3 to inactivate the FAK/ERK signaling pathway, thereby suppressing the progression of BC. Our results hold promise for contributing to the deep understanding of BC pathogenesis and identifying new therapeutic strategies for clinical application.
Collapse
Affiliation(s)
- Ruzhen Zheng
- Department of Oncology, Hangzhou Cancer Hospital, Hangzhou, 310002, Zhejiang, China
| | - Yibo He
- Department of Oncology Surgery, Hangzhou Cancer Hospital, Yanguan Lane 34, Hangzhou, 310002, Zhejiang, China
| | - Lingrong Yang
- Hangzhou Cancer Institution, Hangzhou Cancer Hospital, Hangzhou, 310002, Zhejiang, China
| | - Yidan Chen
- Hangzhou Cancer Institution, Hangzhou Cancer Hospital, Hangzhou, 310002, Zhejiang, China
| | - Rui Wang
- Department of Oncology Surgery, Hangzhou Cancer Hospital, Yanguan Lane 34, Hangzhou, 310002, Zhejiang, China
| | - Shangnao Xie
- Department of Oncology Surgery, Hangzhou Cancer Hospital, Yanguan Lane 34, Hangzhou, 310002, Zhejiang, China.
| |
Collapse
|
4
|
Hua HK, Zhu HM, Zhang ZG. Clinical significance of downregulated NISCH expression in skin cutaneous melanoma: Modulation of tumor cell invasion, migration, and EMT via PAK1 inhibition. Tissue Cell 2024; 88:102399. [PMID: 38723330 DOI: 10.1016/j.tice.2024.102399] [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: 01/15/2024] [Revised: 04/16/2024] [Accepted: 04/30/2024] [Indexed: 06/17/2024]
Abstract
OBJECTIVE This study aimed to investigate the expression and functional role of NISCH in skin cutaneous melanoma (SKCM), exploring its association with clinical characteristics and its potential impact on human skin melanoma cell behavior. METHODS The research assessed differential NISCH expression in SKCM tissues using the GEPIA (Gene Expression Profiling Interactive Analysis) database and validated these findings through immunohistochemical staining of 45 clinical samples. To affirm NISCH expression at the cellular level, three human skin melanoma cell lines (RPMI-7951, A375, MEL-5), and the human normal skin cell line HEMa underwent quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blotting. Transwell experiments evaluated the migration and invasion capabilities of RPMI-7951 and A375 cells post-transduction with NISCH or PAK1 lentiviral activation particles. Additionally, qRT-PCR analysis of epithelial-mesenchymal transition (EMT)-related gene expression (Vimentin, E-cadherin, N-cadherin) was conducted in A375 and RPMI-7951 cells. RESULTS SKCM tissues exhibited significantly reduced NISCH expression compared to normal tissues. Immunohistochemical analysis revealed predominant nuclear localization of NISCH in melanoma cells, with reduced expression significantly correlating with sex, advanced stage, and lymph node metastasis. Melanoma cell lines displayed lower NISCH expression levels compared to normal skin cells. Functional experiments showcased that NISCH overexpression suppressed p-PAK1/PAK1, while PAK1 upregulation notably increased melanoma cell migration, invasion, and induced EMT. Remarkably, NISCH overexpression counteracted PAK1-induced effects on EMT, migration, and invasion in melanoma cells. CONCLUSION NISCH may significantly influence the aggressive behavior of SKCM cells via the PAK1 pathway, making it a potential therapeutic target for managing melanoma metastasis.
Collapse
Affiliation(s)
- Huai-Kang Hua
- Plastic and Reconstructive Surgery, Lishui People' s Hospital, Lishui, Zhejiang 323000, China
| | - Hong-Mei Zhu
- General practice, Xin Bi Community Health Center, Lishui, Zhejiang 321403, China
| | - Zhen-Guo Zhang
- Plastic and Reconstructive Surgery, Lishui People' s Hospital, Lishui, Zhejiang 323000, China.
| |
Collapse
|
5
|
Ostojić M, Đurić A, Živić K, Grahovac J. Analysis of the nischarin expression across human tumor types reveals its context-dependent role and a potential as a target for drug repurposing in oncology. PLoS One 2024; 19:e0299685. [PMID: 38781180 PMCID: PMC11115306 DOI: 10.1371/journal.pone.0299685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Nischarin was reported to be a tumor suppressor that plays a critical role in breast cancer initiation and progression, and a positive prognostic marker in breast, ovarian and lung cancers. Our group has found that nischarin had positive prognostic value in female melanoma patients, but negative in males. This opened up a question whether nischarin has tumor type-specific and sex-dependent roles in cancer progression. In this study, we systematically examined in the public databases the prognostic value of nischarin in solid tumors, regulation of its expression and associated signaling pathways. We also tested the effects of a nischarin agonist rilmenidine on cancer cell viability in vitro. Nischarin expression was decreased in tumors compared to the respective healthy tissues, most commonly due to the deletions of the nischarin gene and promoter methylation. Unlike in healthy tissues where it was located in the cytoplasm and at the membrane, in tumor tissues nischarin could also be observed in the nuclei, implying that nuclear translocation may also account for its cancer-specific role. Surprisingly, in several cancer types high nischarin expression was a negative prognostic marker. Gene set enrichment analysis showed that in tumors in which high nischarin expression was a negative prognostic marker, signaling pathways that regulate stemness were enriched. In concordance with the findings that nischarin expression was negatively associated with pathways that control cancer growth and progression, nischarin agonist rilmenidine decreased the viability of cancer cells in vitro. Taken together, our study lays a ground for functional studies of nischarin in a context-dependent manner and, given that nischarin has several clinically approved agonists, provides rationale for their repurposing, at least in tumors in which nischarin is predicted to be a positive prognostic marker.
Collapse
Affiliation(s)
- Marija Ostojić
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Ana Đurić
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Kristina Živić
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Jelena Grahovac
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| |
Collapse
|
6
|
Zheng P, Pan C, Zhou C, Liu B, Wang L, Duan S, Ding Y. Contribution of Nischarin/IRAS in CNS development, injury and diseases. J Adv Res 2023; 54:43-57. [PMID: 36716956 DOI: 10.1016/j.jare.2023.01.020] [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: 09/27/2022] [Revised: 12/28/2022] [Accepted: 01/24/2023] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Murine Nischarin and its human homolog IRAS are scaffold proteins highly expressed in the central nervous system (CNS). Nischarin was initially discovered as a tumor suppressor protein, and recent studies have also explored its potential value in the CNS. Research on IRAS has largely focused on its effect on opioid dependence. Although the role of Nischarin/IRAS in the physiological function and pathological process of the CNS has gradually attracted attention and the related research results are expected to be applied in clinical practice, there is no systematic review of the role and mechanisms of Nischarin/IRAS in the CNS so far. AIM OF REVIEW This review will systematically analyze the role and mechanism of Nischarin/IRAS in the CNS, and provide necessary references and possible targets for the treatment of neurological diseases, thereby broadening the direction of Nischarin/IRAS research and facilitating clinical translation. KEY SCIENTIFIC CONCEPTS OF REVIEW The pathophysiological processes affected by dysregulation of Nischarin/IRAS expression in the CNS are mainly introduced, including spinal cord injury (SCI), opioid dependence, anxiety, depression, and autism. The molecular mechanisms such as factors regulating Nischarin/IRAS expression and signal transduction pathways regulated by Nischarin/IRAS are systematically summarized. Finally, the clinical application of Nischarin/IRAS has been prospected.
Collapse
Affiliation(s)
- Peijie Zheng
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou 310015, China
| | - Chenshu Pan
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou 310015, China
| | - Chuntao Zhou
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou 310015, China
| | - Bin Liu
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou 310015, China
| | - Linlin Wang
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shiwei Duan
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou 310015, China; Institute of Translational Medicine, Zhejiang University City College, Hangzhou 310015, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Zhejiang University City College, Hangzhou 310015, China.
| | - Yuemin Ding
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou 310015, China; Institute of Translational Medicine, Zhejiang University City College, Hangzhou 310015, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Zhejiang University City College, Hangzhou 310015, China.
| |
Collapse
|
7
|
Gupta S, Bazargani N, Drew J, Howden JH, Modi S, Al Awabdh S, Marie H, Attwell D, Kittler JT. The non-adrenergic imidazoline-1 receptor protein nischarin is a key regulator of astrocyte glutamate uptake. iScience 2022; 25:104127. [PMID: 35434559 PMCID: PMC9010640 DOI: 10.1016/j.isci.2022.104127] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 12/24/2021] [Accepted: 03/17/2022] [Indexed: 12/02/2022] Open
Abstract
Astrocytic GLT-1 is the main glutamate transporter involved in glutamate buffering in the brain, pivotal for glutamate removal at excitatory synapses to terminate neurotransmission and for preventing excitotoxicity. We show here that the surface expression and function of GLT-1 can be rapidly modulated through the interaction of its N-terminus with the nonadrenergic imidazoline-1 receptor protein, Nischarin. The phox domain of Nischarin is critical for interaction and internalization of surface GLT-1. Using live super-resolution imaging, we found that glutamate accelerated Nischarin-GLT-1 internalization into endosomal structures. The surface GLT-1 level increased in Nischarin knockout astrocytes, and this correlated with a significant increase in transporter uptake current. In addition, Nischarin knockout in astrocytes is neuroprotective against glutamate excitotoxicity. These data provide new molecular insights into regulation of GLT-1 surface level and function and suggest new drug targets for the treatment of neurological disorders. Nischarin phox domain interacts with the N-terminus of the glutamate transporter, GLT-1 Nischarin promotes internalization of GLT-1 to endosomes Glutamate modulates GLT-1 surface levels by regulating the Nischarin-GLT-1 interaction Nischarin loss enhances GLT-1 surface levels, transport currents, and neuroprotection
Collapse
Affiliation(s)
- Swati Gupta
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Narges Bazargani
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - James Drew
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Jack H. Howden
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Souvik Modi
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Sana Al Awabdh
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Hélène Marie
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Josef T. Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
- Corresponding author
| |
Collapse
|
8
|
Xinxin L, Zhang Q. LncRNA RP11-214F16.8 drives breast cancer tumorigenesis via a post-translational repression on NISCH expression. Cell Signal 2022; 92:110271. [DOI: 10.1016/j.cellsig.2022.110271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/27/2021] [Accepted: 01/26/2022] [Indexed: 12/24/2022]
|
9
|
p21-Activated kinase 1 (PAK1) in aging and longevity: An overview. Ageing Res Rev 2021; 71:101443. [PMID: 34390849 DOI: 10.1016/j.arr.2021.101443] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/26/2021] [Accepted: 08/10/2021] [Indexed: 02/08/2023]
Abstract
The p21-activated kinases (PAKs) belong to serine/threonine kinases family, regulated by ∼21 kDa small signaling G proteins RAC1 and CDC42. The mammalian PAK family comprises six members (PAK1-6) that are classified into two groups (I and II) based on their domain architecture and regulatory mechanisms. PAKs are implicated in a wide range of cellular functions. PAK1 has recently attracted increasing attention owing to its involvement in oncogenesis, tumor progression, and metastasis as well as several life-limiting diseases and pathological conditions. In Caenorhabditis elegans, PAK1 functions limit the lifespan under basal conditions by inhibiting forkhead transcription factor DAF-16. Interestingly, PAK depletion extended longevity and attenuated the onset of age-related phenotypes in a premature-aging mouse model and delayed senescence in mammalian fibroblasts. These observations implicate PAKs as not only oncogenic but also aging kinases. Therefore, PAK-targeting genetic and/or pharmacological interventions, particularly PAK1-targeting, could be a viable strategy for developing cancer therapies with relatively no side effects and promoting healthy longevity. This review describes PAK family proteins, their biological functions, and their role in regulating aging and longevity using C. elegans. Moreover, we discuss the effect of small-molecule PAK1 inhibitors on the lifespan and healthspan of C. elegans.
Collapse
|
10
|
Understanding the role of integrins in breast cancer invasion, metastasis, angiogenesis, and drug resistance. Oncogene 2021; 40:1043-1063. [PMID: 33420366 DOI: 10.1038/s41388-020-01588-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 11/11/2020] [Accepted: 11/26/2020] [Indexed: 12/13/2022]
Abstract
Integrins are cell adhesion receptors, which are typically transmembrane glycoproteins that connect to the extracellular matrix (ECM). The function of integrins regulated by biochemical events within the cells. Understanding the mechanisms of cell growth by integrins is important in elucidating their effects on tumor progression. One of the major events in integrin signaling is integrin binding to extracellular ligands. Another event is distant signaling that gathers chemical signals from outside of the cell and transmit the signals upon cell adhesion to the inside of the cell. In normal breast tissue, integrins function as checkpoints to monitor effects on cell proliferation, while in cancer tissue these functions altered. The combination of tumor microenvironment and its associated components determines the cell fate. Hypoxia can increase the expression of several integrins. The exosomal integrins promote the growth of metastatic cells. Expression of certain integrins is associated with increased metastasis and decreased prognosis in cancers. In addition, integrin-binding proteins promote invasion and metastasis in breast cancer. Targeting specific integrins and integrin-binding proteins may provide new therapeutic approaches for breast cancer therapies. This review will examine the current knowledge of integrins' role in breast cancer.
Collapse
|
11
|
Cai YJ, Ma B, Wang ML, Chen J, Zhao FG, Zhou JD, Guo X, Zheng L, Xu CJ, Wang Y, He YB, Liu J, Xie SN. Impact of Nischarin on EMT regulators in breast cancer cell lines. Oncol Lett 2020; 20:291. [PMID: 33101485 PMCID: PMC7576990 DOI: 10.3892/ol.2020.12154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/24/2020] [Indexed: 12/13/2022] Open
Abstract
Nischarin is an integrin-binding protein, which is well known as a novel tumor suppressor. In breast cancer, Nischarin serves a critical role in breast cancer cell migration and invasion. However, the molecular mechanism underlying the role of Nischarin remains unclear. Recent findings have demonstrated that epithelial-mesenchymal transition (EMT) increases the capacity of cell migration and invasion. As a member of the integrin family, it was hypothesized that Nischarin may regulate cellular processes via various signaling pathways associated with the EMT process. The present study detected the mRNA levels of EMT regulators via reverse transcription-quantitative PCR and related protein levels via western blotting in breast cancer cells, following NISCH-overexpression and -knockdown. The results demonstrated that Nischarin inhibits cell proliferation, migration and invasion in breast cancer cells. Furthermore, when the NISCH gene was overexpressed, the relative mRNA level of E-cadherin was increased, while the relative mRNA levels of several transcription factors, such as Snail, ZEB1, N-cadherin, Slug, Twist1 and vimentin, decreased. When NISCH was silenced, these results were reversed. The present results demonstrated that Nischarin suppresses cell migration and invasion via inhibiting the EMT process.
Collapse
Affiliation(s)
- Yuan-Jie Cai
- Department of Breast Surgery, Zhejiang Hospital, Hangzhou, Zhejiang 310030, P.R. China
| | - Bo Ma
- Department of Breast Surgery, Zhejiang Hospital, Hangzhou, Zhejiang 310030, P.R. China
| | - Mei-Li Wang
- Department of Breast Surgery, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Jie Chen
- Department of Breast Surgery, Zhejiang Hospital, Hangzhou, Zhejiang 310030, P.R. China
| | - Fu-Guang Zhao
- Department of Breast Surgery, Zhejiang Hospital, Hangzhou, Zhejiang 310030, P.R. China
| | - Juan-Di Zhou
- Department of Breast Surgery, Zhejiang Hospital, Hangzhou, Zhejiang 310030, P.R. China
| | - Xu Guo
- Department of Breast Surgery, Zhejiang Hospital, Hangzhou, Zhejiang 310030, P.R. China
| | - Lei Zheng
- Department of Breast Surgery, Zhejiang Hospital, Hangzhou, Zhejiang 310030, P.R. China
| | - Chun-Jing Xu
- Department of Breast Surgery, Zhejiang Hospital, Hangzhou, Zhejiang 310030, P.R. China
| | - Yi Wang
- Department of Breast Surgery, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Yi-Bo He
- Department of Breast Surgery, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Jian Liu
- Department of Breast Surgery, Zhejiang University Affiliated Hangzhou First People Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Shang-Nao Xie
- Department of Breast Surgery, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310000, P.R. China.,Department of Breast Surgery, Zhejiang University Affiliated Hangzhou First People Hospital, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
12
|
Yao D, Li C, Rajoka MSR, He Z, Huang J, Wang J, Zhang J. P21-Activated Kinase 1: Emerging biological functions and potential therapeutic targets in Cancer. Am J Cancer Res 2020; 10:9741-9766. [PMID: 32863957 PMCID: PMC7449905 DOI: 10.7150/thno.46913] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
The p21-Activated kinase 1 (PAK1), a member of serine-threonine kinases family, was initially identified as an interactor of the Rho GTPases RAC1 and CDC42, which affect a wide range of processes associated with cell motility, survival, metabolism, cell cycle, proliferation, transformation, stress, inflammation, and gene expression. Recently, the PAK1 has emerged as a potential therapeutic target in cancer due to its role in many oncogenic signaling pathways. Many PAK1 inhibitors have been developed as potential preclinical agents for cancer therapy. Here, we provide an overview of essential roles that PAK1 plays in cancer, including its structure and autoactivation mechanism, its crucial function from onset to progression to metastasis, metabolism, immune escape and even drug resistance in cancer; endogenous regulators; and cancer-related pathways. We also summarize the reported PAK1 small-molecule inhibitors based on their structure types and their potential application in cancer. In addition, we provide overviews on current progress and future challenges of PAK1 in cancer, hoping to provide new ideas for the diagnosis and treatment of cancer.
Collapse
|
13
|
Kanumuri R, Saravanan R, Pavithra V, Sundaram S, Rayala SK, Venkatraman G. Current trends and opportunities in targeting p21 activated kinase-1(PAK1) for therapeutic management of breast cancers. Gene 2020; 760:144991. [PMID: 32717309 DOI: 10.1016/j.gene.2020.144991] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/18/2020] [Accepted: 07/21/2020] [Indexed: 12/24/2022]
Abstract
Breast cancer is the most frequently diagnosed cancer in women worldwide. Identifying reliable biomarkers and druggable molecular targets pose to be a significant quest in breast cancer research. p21-activated kinase 1 (PAK1) is a serine/threonine kinase that direct cell motility, cytoskeletal remodelling, and has been shown to function as a downstream regulator for various cancer signalling cascades that promote cell proliferation, apoptosis deregulation and hasten mitotic abnormalities, resulting in tumor formation and progression. The heterogeneity and acquired drug resistance are important factors that challenge the treatment of breast cancer. p21-activated kinase 1 signalling is crucial for activation of the Ras/RAF/MEK/ERK, PI3K/Akt/mTOR and Wnt signalling cascades which regulate cell survival, cell cycle progression, differentiation, and proliferation. A study involving proteogenomics analysis on breast cancer tissues showed the PAK1 as outlier kinase. In addition to this, few outlier molecules were identified specific to subtypes of breast cancer. A few substrates of PAK1 in breast cancer are already known. In this paper, we have discussed a similar approach called Kinase Interacting Substrate Screening (KISS) for the identification of novel oncogenic substrates of p21-activated kinase specific to subtypes of breast cancer. Such high throughput approaches are expected to accelerate the process of identifying novel drug targets and biomarkers.
Collapse
Affiliation(s)
- Rahul Kanumuri
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India; Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamilnadu, India
| | - Roshni Saravanan
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India
| | - V Pavithra
- Department of Pathology, Sri Ramachandra Medical College & Research Institute, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India
| | - Sandhya Sundaram
- Department of Pathology, Sri Ramachandra Medical College & Research Institute, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India
| | - Suresh K Rayala
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamilnadu, India.
| | - Ganesh Venkatraman
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India.
| |
Collapse
|
14
|
Huang S, Zhu Y, Wang C, Li X, Cui X, Tu S, You L, Fu J, Chen Z, Hu W, Gong W. PAK5 facilitates the proliferation, invasion and migration in colorectal cancer cells. Cancer Med 2020; 9:4777-4790. [PMID: 32383357 PMCID: PMC7333859 DOI: 10.1002/cam4.3084] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/08/2020] [Accepted: 04/08/2020] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is the third-most common cancer around the world, accounting for approximately 10% of cancer-related mortality. Deeper molecular understanding of colorectal carcinogenesis will provide evidences for identification of early diagnostic indicators and novel therapeutic strategies for CRC treatment. The p21cdc42/rac1 -activated kinase 5 (PAK5) has been reported to be involved in a variety of tumor-promoting behaviors, whereas the underlying mechanisms of PAK5 in CRC progression are still obscure. Our current study revealed an upregulated expression of PAK5 in human CRC tissues as compared with normal adjacent biopsies, which was associated with tumor progression and metastasis. We further unraveled that inhibition of PAK5 was correlated with restrained proliferation, migration, and invasion of CRC cells in vitro and in vivo. Moreover, we showed an indispensable role of PAK5 in interacting with Cdc42 and Integrin β1, β3, thus, to facilitate the migration and invasion of CRC cells. Collectively, we pointed out a potential of PAK5 to serve as a novel therapeutic target in restricting CRC proliferation and metastasis. The uncovered mechanisms will deepen the comprehension with regard to the mechanisms of CRC progression, as well as providing new insights for therapeutic intervention in colorectal cancer.
Collapse
Affiliation(s)
- Silin Huang
- Department of GastroenterologyShenzhen HospitalSouthern Medical UniversityShenzhenGuangdongChina
| | - Ying Zhu
- Department of GastroenterologyShenzhen HospitalSouthern Medical UniversityShenzhenGuangdongChina
| | - Chunfei Wang
- Endoscopy CenterThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityShenzhenGuangdongChina
| | - Xiaxi Li
- Department of GastroenterologyShenzhen HospitalSouthern Medical UniversityShenzhenGuangdongChina
| | - Xiaobing Cui
- Department of GastroenterologyShenzhen HospitalSouthern Medical UniversityShenzhenGuangdongChina
| | - Sufang Tu
- Department of GastroenterologyShenzhen HospitalSouthern Medical UniversityShenzhenGuangdongChina
| | - Lijuan You
- Department of GastroenterologyShenzhen HospitalSouthern Medical UniversityShenzhenGuangdongChina
| | - JingWen Fu
- Department of GastroenterologyShenzhen HospitalSouthern Medical UniversityShenzhenGuangdongChina
| | - Zemin Chen
- Department of GastroenterologyShenzhen HospitalSouthern Medical UniversityShenzhenGuangdongChina
| | - Wei Hu
- Department of GastroenterologyShenzhen HospitalSouthern Medical UniversityShenzhenGuangdongChina
- Department of Anaesthesia and Intensive CareThe Chinese University of Hong KongHong Kong
| | - Wei Gong
- Department of GastroenterologyShenzhen HospitalSouthern Medical UniversityShenzhenGuangdongChina
| |
Collapse
|
15
|
Dong S, Ruiz-Calderon B, Rathinam R, Eastlack S, Maziveyi M, Alahari SK. Knockout model reveals the role of Nischarin in mammary gland development, breast tumorigenesis and response to metformin treatment. Int J Cancer 2019; 146:2576-2587. [PMID: 31525254 DOI: 10.1002/ijc.32690] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 07/30/2019] [Accepted: 09/10/2019] [Indexed: 01/05/2023]
Abstract
Previously, our lab discovered the protein Nischarin and uncovered its role in regulating cell migration and invasion via its interactions with several proteins. We subsequently described a role for Nischarin in breast cancer, in which it is frequently underexpressed. To characterize Nischarin's role in breast tumorigenesis and mammary gland development more completely, we deleted a critical region of the Nisch gene (exons 7-10) from the mouse genome and observed the effects. Mammary glands in mutant animals showed delayed terminal end bud formation but did not develop breast tumors spontaneously. Therefore, we interbred the animals with transgenic mice expressing the mouse mammary tumor virus-polyoma middle T-antigen (MMTV-PyMT) oncogene. The MMTV-PyMT mammary glands lacking Nischarin showed increased hyperplasia compared to wild-type animal tissues. Furthermore, we observed significantly increased tumor growth and metastasis in Nischarin mutant animals. Surprisingly, Nischarin deletion decreased activity of AMPK and subsequently its downstream effectors. Given this finding, we treated these animals with metformin, which enhances AMPK activity. Here, we show for the first time, metformin activates AMPK signaling and inhibits tumor growth of Nischarin lacking PyMT tumors suggesting a potential use for metformin as a cancer therapeutic, particularly in the case of Nischarin-deficient breast cancers.
Collapse
Affiliation(s)
- Shengli Dong
- Department of Biochemistry and Molecular Biology, Stanley S. Scott Cancer Center, New Orleans, LA
| | | | - Rajamani Rathinam
- Department of Biochemistry and Molecular Biology, Stanley S. Scott Cancer Center, New Orleans, LA
| | - Steven Eastlack
- Department of Biochemistry and Molecular Biology, Stanley S. Scott Cancer Center, New Orleans, LA
| | - Mazvita Maziveyi
- Department of Biochemistry and Molecular Biology, Stanley S. Scott Cancer Center, New Orleans, LA
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, Stanley S. Scott Cancer Center, New Orleans, LA
| |
Collapse
|
16
|
Abstract
Integrins are heterodimeric cell surface receptors ensuring the mechanical connection between cells and the extracellular matrix. In addition to the anchorage of cells to the extracellular matrix, these receptors have critical functions in intracellular signaling, but are also taking center stage in many physiological and pathological conditions. In this review, we provide some historical, structural, and physiological notes so that the diverse functions of these receptors can be appreciated and put into the context of the emerging field of mechanobiology. We propose that the exciting journey of the exploration of these receptors will continue for at least another new generation of researchers.
Collapse
Affiliation(s)
- Michael Bachmann
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| | - Sampo Kukkurainen
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| | - Vesa P Hytönen
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| |
Collapse
|
17
|
Development of insulin resistance in Nischarin mutant female mice. Int J Obes (Lond) 2018; 43:1046-1057. [PMID: 30546133 DOI: 10.1038/s41366-018-0241-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/18/2018] [Accepted: 09/16/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND/OBJECTIVES NISCH-STAB1 is a newly identified locus correlated to human waist-hip ratio (WHR), which is a risk indicator of developing obesity-associated diabetes. Our previous studies have shown that Nisch mutant male mice increased glucose tolerance in chow-fed conditions. Thus we hypothesized that Nisch mutant mice will have changes in insulin resistance, adipocytes, hepatic steatosis when mice are fed with high-fat diet (HFD). METHODS Insulin resistance was assessed in Nisch mutant mice and WT mice fed with high-fat diet (60% by kCal) or chow diet. Whole-body energy metabolism was examined using an indirect calorimeter. Adipose depots including inguinal white adipose tissue (WAT), perigonadal WAT, retroperitoneal WAT, and mesenteric WAT were extracted. Area and eqdiameter of each adipocyte were determined, and insulin signaling was examined as well. Paired samples of subcutaneous and omental visceral adipose tissue were obtained from 400 individuals (267 women, 133 men), and examined the expression of Nischarin. RESULTS We found that insulin signaling was impaired in major insulin-sensitive tissues of Nisch mutant female mice. When mice were fed with HFD for 15 weeks, the Nisch mutant female mice not only developed severe insulin resistance and decreased glucose tolerance compared with wild-type control mice, but also accumulated more white fat, had larger adipocytes and developed severe hepatic steatosis than wild-type control mice. To link our animal studies to human diseases, we further analyzed Nischarin expression in the paired human samples of visceral and subcutaneous adipose tissue from Caucasians. In humans, we found that Nischarin expression is attenuated in adipose tissue with obesity. More importantly, we found that Nischarin mRNA inversely correlated with parameters of obesity, fat distribution, lipid and glucose metabolism. CONCLUSIONS Taken together, our data revealed sexual dimorphism of Nischarin in body fat distribution, insulin resistance, and glucose tolerance in mice.
Collapse
|
18
|
Maziveyi M, Dong S, Baranwal S, Alahari SK. Nischarin regulates focal adhesion and Invadopodia formation in breast cancer cells. Mol Cancer 2018; 17:21. [PMID: 29415725 PMCID: PMC5803897 DOI: 10.1186/s12943-018-0764-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 02/03/2023] Open
Abstract
Background During metastasis, tumor cells move through the tracks of extracellular matrix (ECM). Focal adhesions (FAs) are the protein complexes that link the cell cytoskeleton to the ECM and their presence is necessary for cell attachment. The tumor suppressor Nischarin interacts with a number of signaling proteins such as Integrin α5, PAK1, LIMK1, LKB1, and Rac1 to prevent cancer cell migration. Although previous findings have shown that Nischarin exerts this migratory inhibition by interacting with other proteins, the effects of these interactions on the entire FA machinery are unknown. Methods RT-PCR, Western Blotting, invadopodia assays, and immunofluorescence were used to examine FA gene expression and determine whether Nischarin affects cell attachment, as well as the proteins that regulate it. Results Our data show that Nischarin prevents cell migration and invasion by altering the expression of key focal adhesion proteins. Furthermore, we have found that Nischarin-expressing cells have reduced ability to attach the ECM, which in turn leads to a decrease in invadopodia-mediated matrix degradation. Conclusions These experiments demonstrate an important role of Nischarin in regulating cell attachment, which adds to our understanding of the early events of the metastatic process in breast cancer. Electronic supplementary material The online version of this article (10.1186/s12943-018-0764-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mazvita Maziveyi
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, LA, 70112, USA
| | - Shengli Dong
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, LA, 70112, USA
| | - Somesh Baranwal
- Department of Biochemistry and Microbial Science, Central University of Punjab, Bathinda, 151001, India
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
19
|
Gstrein T, Edwards A, Přistoupilová A, Leca I, Breuss M, Pilat-Carotta S, Hansen AH, Tripathy R, Traunbauer AK, Hochstoeger T, Rosoklija G, Repic M, Landler L, Stránecký V, Dürnberger G, Keane TM, Zuber J, Adams DJ, Flint J, Honzik T, Gut M, Beltran S, Mechtler K, Sherr E, Kmoch S, Gut I, Keays DA. Mutations in Vps15 perturb neuronal migration in mice and are associated with neurodevelopmental disease in humans. Nat Neurosci 2018; 21:207-217. [PMID: 29311744 PMCID: PMC5897053 DOI: 10.1038/s41593-017-0053-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/22/2017] [Indexed: 01/31/2023]
Abstract
The formation of the vertebrate brain requires the generation, migration, differentiation and survival of neurons. Genetic mutations that perturb these critical cellular events can result in malformations of the telencephalon, providing a molecular window into brain development. Here we report the identification of an N-ethyl-N-nitrosourea-induced mouse mutant characterized by a fractured hippocampal pyramidal cell layer, attributable to defects in neuronal migration. We show that this is caused by a hypomorphic mutation in Vps15 that perturbs endosomal-lysosomal trafficking and autophagy, resulting in an upregulation of Nischarin, which inhibits Pak1 signaling. The complete ablation of Vps15 results in the accumulation of autophagic substrates, the induction of apoptosis and severe cortical atrophy. Finally, we report that mutations in VPS15 are associated with cortical atrophy and epilepsy in humans. These data highlight the importance of the Vps15-Vps34 complex and the Nischarin-Pak1 signaling hub in the development of the telencephalon.
Collapse
Affiliation(s)
- Thomas Gstrein
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Andrew Edwards
- Wellcome Trust Center for Human Genetics (WTCHG), Oxford, UK
| | - Anna Přistoupilová
- Institute of Inherited Metabolic Disorders, Charles University, Prague, Czech Republic
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Ines Leca
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Martin Breuss
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | | | - Andi H Hansen
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Ratna Tripathy
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Anna K Traunbauer
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Tobias Hochstoeger
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Gavril Rosoklija
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Marco Repic
- Institute for Molecular Biotechnology (IMBA), Vienna, Austria
| | - Lukas Landler
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Viktor Stránecký
- Institute of Inherited Metabolic Disorders, Charles University, Prague, Czech Republic
| | - Gerhard Dürnberger
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Thomas M Keane
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Johannes Zuber
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - David J Adams
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Jonathan Flint
- Wellcome Trust Center for Human Genetics (WTCHG), Oxford, UK
| | - Tomas Honzik
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marta Gut
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Sergi Beltran
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Karl Mechtler
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Elliott Sherr
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Stanislav Kmoch
- Institute of Inherited Metabolic Disorders, Charles University, Prague, Czech Republic
| | - Ivo Gut
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - David A Keays
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria.
| |
Collapse
|
20
|
Maffioli E, Schulte C, Nonnis S, Grassi Scalvini F, Piazzoni C, Lenardi C, Negri A, Milani P, Tedeschi G. Proteomic Dissection of Nanotopography-Sensitive Mechanotransductive Signaling Hubs that Foster Neuronal Differentiation in PC12 Cells. Front Cell Neurosci 2018; 11:417. [PMID: 29354032 PMCID: PMC5758595 DOI: 10.3389/fncel.2017.00417] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/12/2017] [Indexed: 12/11/2022] Open
Abstract
Neuronal cells are competent in precisely sensing nanotopographical features of their microenvironment. The perceived microenvironmental information will be “interpreted” by mechanotransductive processes and impacts on neuronal functioning and differentiation. Attempts to influence neuronal differentiation by engineering substrates that mimic appropriate extracellular matrix (ECM) topographies are hampered by the fact that profound details of mechanosensing/-transduction complexity remain elusive. Introducing omics methods into these biomaterial approaches has the potential to provide a deeper insight into the molecular processes and signaling cascades underlying mechanosensing/-transduction but their exigence in cellular material is often opposed by technical limitations of major substrate top-down fabrication methods. Supersonic cluster beam deposition (SCBD) allows instead the bottom-up fabrication of nanostructured substrates over large areas characterized by a quantitatively controllable ECM-like nanoroughness that has been recently shown to foster neuron differentiation and maturation. Exploiting this capacity of SCBD, we challenged mechanosensing/-transduction and differentiative behavior of neuron-like PC12 cells with diverse nanotopographies and/or changes of their biomechanical status, and analyzed their phosphoproteomic profiles in these settings. Versatile proteins that can be associated to significant processes along the mechanotransductive signal sequence, i.e., cell/cell interaction, glycocalyx and ECM, membrane/f-actin linkage and integrin activation, cell/substrate interaction, integrin adhesion complex, actomyosin organization/cellular mechanics, nuclear organization, and transcriptional regulation, were affected. The phosphoproteomic data suggested furthermore an involvement of ILK, mTOR, Wnt, and calcium signaling in these nanotopography- and/or cell mechanics-related processes. Altogether, potential nanotopography-sensitive mechanotransductive signaling hubs participating in neuronal differentiation were dissected.
Collapse
Affiliation(s)
- Elisa Maffioli
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | - Carsten Schulte
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Simona Nonnis
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Francesca Grassi Scalvini
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Claudio Piazzoni
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Cristina Lenardi
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Armando Negri
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Paolo Milani
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Gabriella Tedeschi
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| |
Collapse
|
21
|
Dong S, Baranwal S, Garcia A, Serrano-Gomez SJ, Eastlack S, Iwakuma T, Mercante D, Mauvais-Jarvis F, Alahari SK. Nischarin inhibition alters energy metabolism by activating AMP-activated protein kinase. J Biol Chem 2017; 292:16833-16846. [PMID: 28842496 DOI: 10.1074/jbc.m117.784256] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 08/22/2017] [Indexed: 11/06/2022] Open
Abstract
Nischarin (Nisch) is a key protein functioning as a molecular scaffold and thereby hosting interactions with several protein partners. To explore the physiological importance of Nisch, here we generated Nisch loss-of-function mutant mice and analyzed their metabolic phenotype. Nisch-mutant embryos exhibited delayed development, characterized by small size and attenuated weight gain. We uncovered the reason for this phenotype by showing that Nisch binds to and inhibits the activity of AMP-activated protein kinase (AMPK), which regulates energy homeostasis by suppressing anabolic and activating catabolic processes. The Nisch mutations enhanced AMPK activation and inhibited mechanistic target of rapamycin signaling in mouse embryonic fibroblasts as well as in muscle and liver tissues of mutant mice. Nisch-mutant mice also exhibited increased rates of glucose oxidation with increased energy expenditure, despite reduced overall food intake. Moreover, the Nisch-mutant mice had reduced expression of liver markers of gluconeogenesis associated with increased glucose tolerance. As a result, these mice displayed decreased growth and body weight. Taken together, our results indicate that Nisch is an important AMPK inhibitor and a critical regulator of energy homeostasis, including lipid and glucose metabolism.
Collapse
Affiliation(s)
- Shengli Dong
- From the Department of Biochemistry and Molecular Biology, School of Medicine, and
| | - Somesh Baranwal
- From the Department of Biochemistry and Molecular Biology, School of Medicine, and.,the Center for Biochemistry and Microbial Sciences, Central University of Punjab, City Campus Mansa Rd., Bathinda-151001, India
| | - Anapatricia Garcia
- the Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia 30322
| | - Silvia J Serrano-Gomez
- From the Department of Biochemistry and Molecular Biology, School of Medicine, and.,the Pontificia Universidad Javeriana, 11001000 Bogotá, Colombia
| | - Steven Eastlack
- From the Department of Biochemistry and Molecular Biology, School of Medicine, and
| | - Tomoo Iwakuma
- the Department of Cancer Biology, Kansas University Medical Center, Kansas City, Kansas 66160, and
| | - Donald Mercante
- Department of Biostatistics, School of Public Health, Louisiana State University Health Science Center, New Orleans, Louisiana 70112
| | - Franck Mauvais-Jarvis
- the Division of Endocrinology, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - Suresh K Alahari
- From the Department of Biochemistry and Molecular Biology, School of Medicine, and
| |
Collapse
|
22
|
A mutation in Nischarin causes otitis media via LIMK1 and NF-κB pathways. PLoS Genet 2017; 13:e1006969. [PMID: 28806779 PMCID: PMC5570507 DOI: 10.1371/journal.pgen.1006969] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 08/24/2017] [Accepted: 08/08/2017] [Indexed: 01/18/2023] Open
Abstract
Otitis media (OM), inflammation of the middle ear (ME), is a common cause of conductive hearing impairment. Despite the importance of the disease, the aetiology of chronic and recurrent forms of middle ear inflammatory disease remains poorly understood. Studies of the human population suggest that there is a significant genetic component predisposing to the development of chronic OM, although the underlying genes are largely unknown. Using N-ethyl-N-nitrosourea mutagenesis we identified a recessive mouse mutant, edison, that spontaneously develops a conductive hearing loss due to chronic OM. The causal mutation was identified as a missense change, L972P, in the Nischarin (NISCH) gene. edison mice develop a serous or granulocytic effusion, increasingly macrophage and neutrophil rich with age, along with a thickened, inflamed mucoperiosteum. We also identified a second hypomorphic allele, V33A, with only modest increases in auditory thresholds and reduced incidence of OM. NISCH interacts with several proteins, including ITGA5 that is thought to have a role in modulating VEGF-induced angiogenesis and vascularization. We identified a significant genetic interaction between Nisch and Itga5; mice heterozygous for Itga5-null and homozygous for edison mutations display a significantly increased penetrance and severity of chronic OM. In order to understand the pathological mechanisms underlying the OM phenotype, we studied interacting partners to NISCH along with downstream signalling molecules in the middle ear epithelia of edison mouse. Our analysis implicates PAK1 and RAC1, and downstream signalling in LIMK1 and NF-κB pathways in the development of chronic OM. Otitis media (OM) is the most common cause of deafness in children and is primarily characterised by inflammation of the middle ear. It is the most common cause of surgery in children in the developed world, with many children developing recurrent and chronic forms of OM undergoing tympanostomy tube insertion. There is evidence that a significant genetic component contributes towards the development of recurrent and chronic forms of OM. The mouse has been a powerful tool for identifying the genes involved in chronic OM. In this study we identified and characterised edison, a novel mouse model of chronic OM that shares important features with the chronic disease in humans. A mutation in the Nisch gene causes edison mice to spontaneously develop OM following birth and subsequently develop chronic OM, with an associated hearing loss. Our molecular analysis of the mutation reveals the underlying pathological mechanisms and pathways involved in OM in the edison mouse, involving PAK1, RAC1 and downstream signalling in LIMK1 and NF-κB pathways. Identification of the edison mutant provides an important genetic disease model of chronic OM and implicates a new gene and genetic pathways involved in predisposition to OM.
Collapse
|
23
|
The traditional Chinese medicine Achyranthes bidentata and our de novo conception of its metastatic chemoprevention: from phytochemistry to pharmacology. Sci Rep 2017. [PMID: 28634392 PMCID: PMC5478643 DOI: 10.1038/s41598-017-02054-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Our recent biosystems analysis revealed similarities between embryonic implantation and cancer cell adhesion, which suggests that abortifacients may be good for safe and effective metastatic chemoprevention targeting circulating tumor cells (CTC). Here we test the hypothesis by using the well-known abortion herb Achyranthes bidentata Blume (A. bidentata). Five compounds were separated from the herb root. Among them, ginsenoside Ro was the most potent in inhibiting embryonic implantation within non-cytotoxic concentrations. It specifically inhibited the metastatic dissemination capability of colon cancer cells HT29, including the migration and invasion ability, and their adhesion to human endothelium through inhibiting integrin αvβ6, MMP-2, MMP-9, and ERK phosphorylation by HT29. Pretreatment of nude mice with oral ginsenoside Ro followed by HT29 intravenous inoculation and 40-day oral ginsenoside Ro significantly prevented lung metastasis with downregulation of integrin αvβ6 and no toxicity. The present study firstly introduces the new conception of utilizing safe and effective abortion botanic medicines for CTC-based metastatic chemoprevention.
Collapse
|
24
|
Expression of Nischarin negatively correlates with estrogen receptor and alters apoptosis, migration and invasion in human breast cancer. Biochem Biophys Res Commun 2017; 484:536-542. [PMID: 28131840 DOI: 10.1016/j.bbrc.2017.01.109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 01/21/2017] [Indexed: 11/22/2022]
Abstract
Nischarin, a novel integrin binding protein, has been demonstrated its negative effects on cell migration and invasion. However, the biological role of Nischarin in breast cancer has not been fully elucidated yet. Our study aimed to analyze the association between Nischarin expression and clinical features of breast cancer patients, and further investigate the role of Nischarin in breast cancer cells apoptosis, migration and invasion. Results showed that Nischarin expression was significantly lower in breast cancer tissues (37.8%, 23/67) than in normal tissues (61.8%, 21/34; P < 0.05), and the expression of Nischarin significantly negatively correlated with estrogen receptor status. Similarly, Nischarin expression was highest in normal breast cell line HBL-100 while triple-negative breast cancer cell line MDA-MB-231 had the lowest expression of Nischarin. Further experiments demonstrated that overexpression of Nischarin may induce apoptosis, and inhibit cell migration and invasion. The present data confirmed that Nishcharin might be a novel tumor suppressor and plays an important role in breast cancer cell apoptosis and metastasis, which can be used as a potential therapeutic target for breast cancer treatment.
Collapse
|
25
|
Ding YM, Li YY, Wang C, Huang H, Zheng CC, Huang SH, Xuan Y, Sun XY, Zhang X. Nischarin-siRNA delivered by polyethylenimine-alginate nanoparticles accelerates motor function recovery after spinal cord injury. Neural Regen Res 2017; 12:1687-1694. [PMID: 29171434 PMCID: PMC5696850 DOI: 10.4103/1673-5374.217348] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
A previous study by our group found that inhibition of nischarin promotes neurite outgrowth and neuronal regeneration in Neuro-2a cells and primary cortical neurons. In recent years, more and more studies have shown that nanomaterials have good prospects in treatment of spinal cord injury. We proposed that small interfering RNA targeting nischarin (Nis-siRNA) delivered by polyethyleneimine-alginate (PEI-ALG) nanoparticles promoted motor function recovery in rats with spinal cord injury. Direct microinjection of 5 μL PEI-ALG/Nis-siRNA into the spinal cord lesion area of spinal cord injury rats was performed. From day 7 after surgery, Basso, Beattie and Bresnahan score was significantly higher in rats from the PEI-ALG/Nis-siRNA group compared with the spinal cord injury group and PEI-ALG/Control-siRNA group. On day 21 after injection, hematoxylin-eosin staining showed that the necrotic area was reduced in the PEI-ALG/Nis-siRNA group. Immunohistochemistry and western blot assay results confirmed successful inhibition of nischarin expression and increased protein expression of growth-associated protein-43 in the PEI-ALG/Nis-siRNA group. These findings suggest that a complex of PEI-ALG nanoparticles and Nis-siRNA effectively suppresses nischarin expression, induces expression of growth-associated protein-43, and accelerates motor function recovery after spinal cord injury.
Collapse
Affiliation(s)
- Yue-Min Ding
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang Province, China
| | - Yu-Ying Li
- Department of Physiology, School of Medicine, Quzhou College of Technology, Quzhou, Zhejiang Province, China
| | - Chu Wang
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang Province, China
| | - Hao Huang
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang Province, China
| | - Chen-Chen Zheng
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang Province, China
| | - Shao-Han Huang
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang Province, China
| | - Yang Xuan
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang Province, China
| | - Xiao-Yi Sun
- Department of Pharmacy, Zhejiang University City College, Hangzhou, Zhejiang Province, China
| | - Xiong Zhang
- Department of Basic Medicine, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
26
|
Kumar R, Sanawar R, Li X, Li F. Structure, biochemistry, and biology of PAK kinases. Gene 2016; 605:20-31. [PMID: 28007610 DOI: 10.1016/j.gene.2016.12.014] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/24/2016] [Accepted: 12/14/2016] [Indexed: 02/07/2023]
Abstract
PAKs, p21-activated kinases, play central roles and act as converging junctions for discrete signals elicited on the cell surface and for a number of intracellular signaling cascades. PAKs phosphorylate a vast number of substrates and act by remodeling cytoskeleton, employing scaffolding, and relocating to distinct subcellular compartments. PAKs affect wide range of processes that are crucial to the cell from regulation of cell motility, survival, redox, metabolism, cell cycle, proliferation, transformation, stress, inflammation, to gene expression. Understandably, their dysregulation disrupts cellular homeostasis and severely impacts key cell functions, and many of those are implicated in a number of human diseases including cancers, neurological disorders, and cardiac disorders. Here we provide an overview of the members of the PAK family and their current status. We give special emphasis to PAK1 and PAK4, the prototypes of groups I and II, for their profound roles in cancer, the nervous system, and the heart. We also highlight other family members. We provide our perspective on the current advancements, their growing importance as strategic therapeutic targets, and our vision on the future of PAKs.
Collapse
Affiliation(s)
- Rakesh Kumar
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20037, USA; Cancer Biology Program, Rajiv Gandhi Center of Biotechnology, Thiruvananthapuram 695014, India.
| | - Rahul Sanawar
- Cancer Biology Program, Rajiv Gandhi Center of Biotechnology, Thiruvananthapuram 695014, India
| | - Xiaodong Li
- Department of Cell Biology, Key Laboratory of Medical Cell Biology, Chinese Ministry of Education, China Medical University, Shenyang 110122, China
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Medical Cell Biology, Chinese Ministry of Education, China Medical University, Shenyang 110122, China.
| |
Collapse
|
27
|
Mervin LH, Cao Q, Barrett IP, Firth MA, Murray D, McWilliams L, Haddrick M, Wigglesworth M, Engkvist O, Bender A. Understanding Cytotoxicity and Cytostaticity in a High-Throughput Screening Collection. ACS Chem Biol 2016; 11:3007-3023. [PMID: 27571164 DOI: 10.1021/acschembio.6b00538] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
While mechanisms of cytotoxicity and cytostaticity have been studied extensively from the biological side, relatively little is currently understood regarding areas of chemical space leading to cytotoxicity and cytostasis in large compound collections. Predicting and rationalizing potential adverse mechanism-of-actions (MoAs) of small molecules is however crucial for screening library design, given the link of even low level cytotoxicity and adverse events observed in man. In this study, we analyzed results from a cell-based cytotoxicity screening cascade, comprising 296 970 nontoxic, 5784 cytotoxic and cytostatic, and 2327 cytostatic-only compounds evaluated on the THP-1 cell-line. We employed an in silico MoA analysis protocol, utilizing 9.5 million active and 602 million inactive bioactivity points to generate target predictions, annotate predicted targets with pathways, and calculate enrichment metrics to highlight targets and pathways. Predictions identify known mechanisms for the top ranking targets and pathways for both phenotypes after review and indicate that while processes involved in cytotoxicity versus cytostaticity seem to overlap, differences between both phenotypes seem to exist to some extent. Cytotoxic predictions highlight many kinases, including the potentially novel cytotoxicity-related target STK32C, while cytostatic predictions outline targets linked with response to DNA damage, metabolism, and cytoskeletal machinery. Fragment analysis was also employed to generate a library of toxicophores to improve general understanding of the chemical features driving toxicity. We highlight substructures with potential kinase-dependent and kinase-independent mechanisms of toxicity. We also trained a cytotoxic classification model on proprietary and public compound readouts, and prospectively validated these on 988 novel compounds comprising difficult and trivial testing instances, to establish the applicability domain of models. The proprietary model performed with precision and recall scores of 77.9% and 83.8%, respectively. The MoA results and top ranking substructures with accompanying MoA predictions are available as a platform to assess screening collections.
Collapse
Affiliation(s)
- Lewis H. Mervin
- Centre
for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Qing Cao
- Discovery Sciences, AstraZeneca R&D, Waltham, United States
| | - Ian P. Barrett
- Discovery Sciences, AstraZeneca R&D, Cambridge Science Park, Cambridge, United Kingdom
| | - Mike A. Firth
- Discovery Sciences, AstraZeneca R&D, Cambridge Science Park, Cambridge, United Kingdom
| | - David Murray
- Discovery Sciences, AstraZeneca R&D, Alderley Park, Macclesfield, United Kingdom
| | - Lisa McWilliams
- Discovery Sciences, AstraZeneca R&D, Alderley Park, Macclesfield, United Kingdom
| | - Malcolm Haddrick
- Discovery Sciences, AstraZeneca R&D, Alderley Park, Macclesfield, United Kingdom
| | - Mark Wigglesworth
- Discovery Sciences, AstraZeneca R&D, Alderley Park, Macclesfield, United Kingdom
| | - Ola Engkvist
- Discovery Sciences, AstraZeneca R&D, Mölndal, Sweden
| | - Andreas Bender
- Centre
for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
28
|
Fan Z, Ley K. Leukocyte arrest: Biomechanics and molecular mechanisms of β2 integrin activation. Biorheology 2016; 52:353-77. [PMID: 26684674 DOI: 10.3233/bir-15085] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Integrins are a group of heterodimeric transmembrane receptors that play essential roles in cell-cell and cell-matrix interaction. Integrins are important in many physiological processes and diseases. Integrins acquire affinity to their ligand by undergoing molecular conformational changes called activation. Here we review the molecular biomechanics during conformational changes of integrins, integrin functions in leukocyte biorheology (adhesive functions during rolling and arrest) and molecules involved in integrin activation.
Collapse
Affiliation(s)
- Zhichao Fan
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.,Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
29
|
Wang P, Du X, Xiong M, Cui J, Yang Q, Wang W, Chen Y, Zhang T. Ginsenoside Rd attenuates breast cancer metastasis implicating derepressing microRNA-18a-regulated Smad2 expression. Sci Rep 2016; 6:33709. [PMID: 27641158 PMCID: PMC5027393 DOI: 10.1038/srep33709] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 09/01/2016] [Indexed: 12/25/2022] Open
Abstract
Metastasis remains a major cause of mortality and poor prognosis in breast cancer patients. Anti-metastatic therapies are in great need to achieve optimal clinical outcome in breast cancer patients. Panax Notoginseng Saponins (PNS) has previously been shown to inhibit breast cancer metastasis in mouse. Here the potential anti-metastatic effect of one of the chemical compounds of PNS, ginsenoside Rd (Rd), was further evaluated in mouse mammary carcinoma 4T1 cells. The results revealed that Rd treatment dose-dependently suppressed cell migration and invasion in cultured 4T1 cells. In 4T1 cell-inoculated mice, Rd treatment led to decreased number of tumor lesions in lungs in both spontaneous and experimental metastasis models. Rd treatment resulted in increased expression of Smad2 in cultured 4T1 cells and in tumors grown from inoculated 4T1 cells. Rd treatment decreased the expression of microRNA (miR)-18a in cultured 4T1 cells and in tumors derived from inoculated 4T1 cells. Smad2 was further verified to be a direct target of miR-18a in 4T1 cells. The significant impact of Rd on counteracting miR-18a-medidated downregulation of Smad2 expression was also demonstrated. Together, the current work shows for the first time that Rd treatment attenuates breast cancer metastasis in part through derepressing miR-18a-mediated Smad2 expression regulation.
Collapse
Affiliation(s)
- Peiwei Wang
- Yueyang Hospital &Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xiaoye Du
- Yueyang Hospital &Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Minqi Xiong
- Yueyang Hospital &Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Jingang Cui
- Yueyang Hospital &Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Qinbo Yang
- Yueyang Hospital &Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Wenjian Wang
- Yueyang Hospital &Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yu Chen
- Yueyang Hospital &Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Teng Zhang
- Yueyang Hospital &Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| |
Collapse
|
30
|
Ding Y, Li Y, Lu L, Zhang R, Zeng L, Wang L, Zhang X. Inhibition of Nischarin Expression Promotes Neurite Outgrowth through Regulation of PAK Activity. PLoS One 2015; 10:e0144948. [PMID: 26670864 PMCID: PMC4682924 DOI: 10.1371/journal.pone.0144948] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 11/25/2015] [Indexed: 11/19/2022] Open
Abstract
Nischarin is a cytoplasmic protein expressed in various organs that plays an inhibitory role in cell migration and invasion and the carcinogenesis of breast cancer cells. We previously reported that Nischarin is highly expressed in neuronal cell lines and is differentially expressed in the brain tissue of adult rats. However, the physiological function of Nischarin in neural cells remains unknown. Here, we show that Nischarin is expressed in rat primary cortical neurons but not in astrocytes. Nischarin is localized around the nucleus and dendrites. Using shRNA to knockdown the expression of endogenous Nischarin significantly increases the percentage of neurite-bearing cells, remarkably increases neurite length, and accelerates neurite extension in neuronal cells. Silencing Nischarin expression also promotes dendrite elongation in rat cortical neurons where Nischarin interacts with p21-activated kinase 1/2 (PAK1/2) and negatively regulates phosphorylation of both PAK1 and PAK2. The stimulation of neurite growth observed in cells with decreased levels of Nischarin is partially abolished by IPA3-mediated inhibition of PAK1 activity. Our findings indicate that endogenous Nischarin inhibits neurite outgrowth by blocking PAK1 activation in neurons.
Collapse
Affiliation(s)
- Yuemin Ding
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, 310015, China
| | - Yuying Li
- Department of Physiology, School of Medicine, Quzhou College of Technology, Quzhou, 324000, China
| | - Lingchao Lu
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, 310015, China
| | - Ruyi Zhang
- Department of Pathology, Jiaxing Second Hospital, Jiaxing, 314000, China
| | - Linghui Zeng
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, 310015, China
| | - Linlin Wang
- Department of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
- * E-mail: (XZ); (LW)
| | - Xiong Zhang
- Department of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
- * E-mail: (XZ); (LW)
| |
Collapse
|
31
|
Maziveyi M, Alahari SK. Breast Cancer Tumor Suppressors: A Special Emphasis on Novel Protein Nischarin. Cancer Res 2015; 75:4252-9. [PMID: 26392073 DOI: 10.1158/0008-5472.can-15-1395] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 07/02/2015] [Indexed: 11/16/2022]
Abstract
Tumor suppressor genes regulate cell growth and prevent spontaneous proliferation that could lead to aberrant tissue function. Deletions and mutations of these genes typically lead to progression through the cell-cycle checkpoints, as well as increased cell migration. Studies of these proteins are important as they may provide potential treatments for breast cancers. In this review, we discuss a comprehensive overview on Nischarin, a novel protein discovered by our laboratory. Nischarin, or imidazoline receptor antisera-selected protein, is a protein involved in a vast number of cellular processes, including neuronal protection and hypotension. The NISCH promoter experiences hypermethylation in several cancers, whereas some highly aggressive breast cancer cells exhibit genomic loss of the NISCH locus. Furthermore, we discuss data illustrating a novel role of Nischarin as a tumor suppressor in breast cancer. Analysis of this new paradigm may shed light on various clinical questions. Finally, the therapeutic potential of Nischarin is discussed.
Collapse
Affiliation(s)
- Mazvita Maziveyi
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, New Orleans, Louisiana.
| |
Collapse
|
32
|
Li J, He X, Dong R, Wang Y, Yu J, Qiu H. Frequent Loss of NISCH Promotes Tumor Proliferation and Invasion in Ovarian Cancer via Inhibiting the FAK Signal Pathway. Mol Cancer Ther 2015; 14:1202-12. [PMID: 25724667 DOI: 10.1158/1535-7163.mct-14-0911] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 02/17/2015] [Indexed: 11/16/2022]
Abstract
NISCH encodes the imidazoline receptor Nischarin and is a known tumor suppressor in many human malignancies; however, its roles in ovarian cancer are still largely unknown. Here, we aim to investigate the biologic functions of NISCH in ovarian cancer. We found that NISCH was significantly downregulated, which correlated considerably with advanced tumor stage, poor differentiation, lymph node metastasis, and the serous/mucinous subtypes in a panel of ovarian cancer tissues. Moreover, NISCH gene silencing was mainly the product of promoter hypermethylation, which could be reversed by treatment with 5-aza-dC. In vitro, NISCH overexpression suppressed cell proliferation and colony formation by hindering cell-cycle progression, whereas the opposite was observed in NISCH knockdown counterparts. In vivo, abundant NISCH expression hindered the growth of HO8910 xenografts, whereas NISCH knockdown accelerated the growth of SKOV3 xenografts. In addition, NISCH significantly attenuated cell invasion by inhibiting the phosphorylation of FAK and ERK, which could be neutralized by PF-562271 (a FAK/Pyk2 inhibitor). Accordingly, NISCH knockdown xenografts exhibited increased peritoneal/pelvic metastases that were not present in counterparts treated with PF-562271. Furthermore, NISCH expression in primary ovarian cancer cells predicted a cellular resistance to PF-562271. In conclusion, we showed that NISCH was frequently silenced by promoter hypermethylation in human ovarian cancer. NISCH manipulated cellular proliferation and invasion by arresting cell cycle and inhibiting the FAK signal. Our findings revealed the biologic functions of NISCH in ovarian cancer, and might be useful for treating patients with aberrant expression of NISCH.
Collapse
Affiliation(s)
- Jing Li
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China. Key-Discipline Laboratory of Clinical Medicine Henan, Zhengzhou, China
| | - Xiaoying He
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ruofan Dong
- Department of Obstetrics and Gynecology, the Affiliated Hospital of Jiangnan University and The 4th People's Hospital of Wuxi, Jiangsu Province, China
| | - Yuan Wang
- Department of Obstetrics and Gynecology, the Affiliated Hospital of Jiangnan University and The 4th People's Hospital of Wuxi, Jiangsu Province, China
| | - Jinjin Yu
- Department of Obstetrics and Gynecology, the Affiliated Hospital of Jiangnan University and The 4th People's Hospital of Wuxi, Jiangsu Province, China
| | - Haifeng Qiu
- Key-Discipline Laboratory of Clinical Medicine Henan, Zhengzhou, China. Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
33
|
Chen J, Feng WL, Mo WJ, Ding XW, Xie SN. Expression of integrin-binding protein Nischarin in metastatic breast cancer. Mol Med Rep 2015; 12:77-82. [PMID: 25695373 PMCID: PMC4438937 DOI: 10.3892/mmr.2015.3373] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 12/19/2014] [Indexed: 11/24/2022] Open
Abstract
The present study aimed to investigate the expression of Nischarin protein in primary breast cancer (PBC), and to evaluate its role in tumor metastasis. Paired specimens of breast cancer tissues and adjacent normal tissues were surgically obtained from 60 patients with PBC at the Zhejiang Cancer Hospital (Hangzhou, China). Nischarin protein concentrations were determined by an ELISA assay. Breast cancer tissues exhibited a significantly lower concentration of Nischarin (5.86±3.19 ng/ml) compared with that of the adjacent noncancerous tissues (9.25±3.65 ng/ml; P<0.001). Furthermore, cancer tissue from patients with lymph node metastasis had significantly lower levels of Nischarin protein (4.69±2.40 ng/ml) than those of patients without lymph node metastasis (7.04±3.47 ng/ml; P=0.004). There was no significant difference in Nischarin protein expression levels between patients with grade I, II or III PBC (grade I, 5.44±3.57 ng/ml; grade II, 6.42±3.85 ng/ml and grade III, 5.10±1.18 ng/ml; P=0.765). The significant differences in the expression of Nischarin between: i) Cancer tissue and noncancerous tissue and ii) patients with and without lymph node metastasis, suggested that Nischarin may have a significant role in tumor occurrence and metastasis of breast cancer. Nischarin expression may therefore be used as a marker to predict the invasiveness and metastasis of PBC.
Collapse
Affiliation(s)
- Jie Chen
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Wei-Liang Feng
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Wen-Ju Mo
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Xiao-Wen Ding
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Shang-Nao Xie
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| |
Collapse
|
34
|
Hammer A, Diakonova M. Tyrosyl phosphorylated serine-threonine kinase PAK1 is a novel regulator of prolactin-dependent breast cancer cell motility and invasion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 846:97-137. [PMID: 25472536 DOI: 10.1007/978-3-319-12114-7_5] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite efforts to discover the cellular pathways regulating breast cancer metastasis, little is known as to how prolactin (PRL) cooperates with extracellular environment and cytoskeletal proteins to regulate breast cancer cell motility and invasion. We implicated serine-threonine kinase p21-activated kinase 1 (PAK1) as a novel target for PRL-activated Janus-kinase 2 (JAK2). JAK2-dependent PAK1 tyrosyl phosphorylation plays a critical role in regulation of both PAK1 kinase activity and scaffolding properties of PAK1. Tyrosyl phosphorylated PAK1 facilitates PRL-dependent motility via at least two mechanisms: formation of paxillin/GIT1/βPIX/pTyr-PAK1 complexes resulting in increased adhesion turnover and phosphorylation of actin-binding protein filamin A. Increased adhesion turnover is the basis for cell migration and phosphorylated filamin A stimulates the kinase activity of PAK1 and increases actin-regulating activity to facilitate cell motility. Tyrosyl phosphorylated PAK1 also stimulates invasion of breast cancer cells in response to PRL and three-dimensional (3D) collagen IV via transcription and secretion of MMP-1 and MMP-3 in a MAPK-dependent manner. These data illustrate the complex interaction between PRL and the cell microenvironment in breast cancer cells and suggest a pivotal role for PRL/PAK1 signaling in breast cancer metastasis.
Collapse
Affiliation(s)
- Alan Hammer
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | | |
Collapse
|
35
|
Parrini MC. Untangling the complexity of PAK1 dynamics: The future challenge. CELLULAR LOGISTICS 2014; 2:78-83. [PMID: 23125950 PMCID: PMC3485744 DOI: 10.4161/cl.19817] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PAK1 kinase is a crucial regulator of a variety of cellular processes, such as motility, cell division, gene transcription and apoptosis. Its deregulation is involved in several pathologies, including cancer, viral infection and neurodegenerative diseases. Due to this strong implication in human health, the complex network of signaling pathways centered on PAK1 is a subject of intensive investigations. This review summarizes the present knowledge on the multiple PAK1 intracellular localizations and on its shuttling between different compartments. The dynamics of PAK1 localization and activation are finely tuned by the cell and it is this tight control that underlies the capacity of PAK1 to participate in the regulation of many fundamental cell functions. Recently, PAK1 biosensors have been developed to visualize PAK1 activation in live cells. These new imaging tools should be of great help to better understand PAK1 biology and to conceive strategies for efficient and specific PAK1 inhibitors.
Collapse
Affiliation(s)
- Maria Carla Parrini
- Institut Curie; Centre de Recherche; Paris, France; Inserm U830; Paris, France
| |
Collapse
|
36
|
Grassot V, Da Silva A, Saliba J, Maftah A, Dupuy F, Petit JM. Highlights of glycosylation and adhesion related genes involved in myogenesis. BMC Genomics 2014; 15:621. [PMID: 25051993 PMCID: PMC4223822 DOI: 10.1186/1471-2164-15-621] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 07/14/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Myogenesis is initiated by myoblast differentiation and fusion to form myotubes and muscle fibres. A population of myoblasts, known as satellite cells, is responsible for post-natal growth of muscle and for its regeneration. This differentiation requires many changes in cell behaviour and its surrounding environment. These modifications are tightly regulated over time and can be characterized through the study of changes in gene expression associated with this process. During the initial myogenesis steps, using the myoblast cell line C2C12 as a model, Janot et al. (2009) showed significant variations in expression for genes involved in pathways of glycolipid synthesis. In this study we used murine satellite cells (MSC) and their ability to differentiate into myotubes or early fat storage cells to select glycosylation related genes whose variation of expression is myogenesis specific. RESULTS The comparison of variant genes in both MSC differentiation pathways identified 67 genes associated with myogenesis. Comparison with data obtained for C2C12 revealed that only 14 genes had similar expression profiles in both cell types and that 17 genes were specifically regulated in MSC. Results were validated statistically by without a priori clustering. Classification according to protein function encoded by these 31 genes showed that the main regulated cellular processes during this differentiation were (i) remodeling of the extracellular matrix, particularly, sulfated structures, (ii) down-regulation of O-mannosyl glycan biosynthesis, and (iii) an increase in adhesion protein expression. A functional study was performed on Itga11 and Chst5 encoding two highly up-regulated proteins. The inactivation of Chst5 by specific shRNA delayed the fusion of MSC. By contrast, the inactivation of Itga11 by specific shRNA dramatically decreased the fusion ability of MSC. This result was confirmed by neutralization of Itga11 product by specific antibodies. CONCLUSIONS Our screening method detected 31 genes specific for myogenic differentiation out of the 383 genes studied. According to their function, interaction networks of the products of these selected genes converged to cell fusion. Functional studies on Itga11 and Chst5 demonstrated the robustness of this screening.
Collapse
Affiliation(s)
- Vincent Grassot
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| | - Anne Da Silva
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| | - James Saliba
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| | - Abderrahman Maftah
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| | - Fabrice Dupuy
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| | - Jean-Michel Petit
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| |
Collapse
|
37
|
Wang P, Cui J, Du X, Yang Q, Jia C, Xiong M, Yu X, Li L, Wang W, Chen Y, Zhang T. Panax notoginseng saponins (PNS) inhibits breast cancer metastasis. JOURNAL OF ETHNOPHARMACOLOGY 2014; 154:663-671. [PMID: 24793216 DOI: 10.1016/j.jep.2014.04.037] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 03/27/2014] [Accepted: 04/22/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax notoginseng (Burkill) F.H. Chen (Araliaceae) has been extensively used as a therapeutic agent to treat a variety of diseases. Panax notoginseng saponins (PNS) consist of major therapeutically active components of Panax notoginseng. PNS inhibit the growth of a variety of tumor cells in vitro and in vivo. The aim of the study is to investigate the effects and underlying mechanisms of PNS on breast cancer metastasis. MATERIALS AND METHODS 4T1 cell, a highly metastatic mouse breast carcinoma cell line, was utilized for in vitro and in vivo assays. In vitro assays were first performed to examine the effects of PNS on 4T1 cell viability, migration and invasion, respectively. Real-time PCR analyses were also performed to examine the effects of PNS on the expression of genes associated with tumor metastasis. The effect of PNS on 4T1 tumor cell metastasis was further assessed in spontaneous and experimental metastasis models in vivo. RESULTS PNS treatment exhibited a dose-dependent effect on impairing 4T1 cell viability in vitro. However, when examined at a lower dose that did not affect cell viability, the migration and invasion of 4T1 cell was remarkably inhibited in vitro. Meanwhile, PNS treatment led to upregulated expression of genes known to inhibit metastasis and downregulated expression of genes promoting metastasis in cultured 4T1 cells. These results suggested a selective effect of PNS on 4T1 migration and invasion. This hypothesis was further addressed in 4T1 metastasis models in vivo. The results showed that the lung metastasis was significantly inhibited by PNS treatment in both spontaneous and experimental metastasis models. CONCLUSION Taken together, our results demonstrated an inhibitory effect of PNS on 4T1 tumor metastasis, warranting further evaluation of PNS as a therapeutic agent for treating breast cancer metastasis.
Collapse
Affiliation(s)
- Peiwei Wang
- Clinical Research Institute of Integrative Medicine & Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China
| | - Jingang Cui
- Clinical Research Institute of Integrative Medicine & Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China
| | - Xiaoye Du
- Clinical Research Institute of Integrative Medicine & Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China
| | - Qinbo Yang
- Clinical Research Institute of Integrative Medicine & Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China
| | - Chenglin Jia
- Clinical Research Institute of Integrative Medicine & Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China
| | - Minqi Xiong
- Clinical Research Institute of Integrative Medicine & Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China
| | - Xintong Yu
- Clinical Research Institute of Integrative Medicine & Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China
| | - Li Li
- Clinical Research Institute of Integrative Medicine & Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China
| | - Wenjian Wang
- Clinical Research Institute of Integrative Medicine & Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China
| | - Yu Chen
- Clinical Research Institute of Integrative Medicine & Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China.
| | - Teng Zhang
- Clinical Research Institute of Integrative Medicine & Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China.
| |
Collapse
|
38
|
Abstract
p21-Activated protein kinases (PAKs) are centrally involved in a plethora of cellular processes and functions. Their function as effectors of small GTPases Rac1 and Cdc42 has been extensively studied during the past two decades, particularly in the realms of cell proliferation, apoptosis, and hence tumorigenesis, as well as cytoskeletal remodeling and related cellular events in health and disease. In recent years, a large number of studies have shed light onto the fundamental role of group I PAKs, most notably PAK1, in metabolic homeostasis. In skeletal muscle, PAK1 was shown to mediate the function of insulin on stimulating GLUT4 translocation and glucose uptake, while in pancreatic β-cells, PAK1 participates in insulin granule localization and vesicle release. Furthermore, we demonstrated that PAK1 mediates the cross talk between insulin and Wnt/β-catenin signaling pathways and hence regulates gut proglucagon gene expression and the production of the incretin hormone glucagon-like peptide-1 (GLP-1). The utilization of chemical inhibitors of PAK and the characterization of Pak1(-/-) mice enabled us to gain mechanistic insights as well as to assess the overall contribution of PAKs in metabolic homeostasis. This review summarizes our current understanding of PAKs, with an emphasis on the emerging roles of PAK1 in glucose homeostasis.
Collapse
|
39
|
The expression pattern of Nischarin after lipopolysaccharides (LPS)-induced neuroinflammation in rats brain cortex. Inflamm Res 2013; 62:929-40. [PMID: 24064952 DOI: 10.1007/s00011-013-0631-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 04/02/2013] [Accepted: 04/30/2013] [Indexed: 10/26/2022] Open
Abstract
OBJECTIVE To investigate whether Nischarin participated in neuronal apoptosis induced by neuroinflammation and via the phosphatidylinositol 3-kinase (PI3K) and PKB-dependent pathway. MATERIAL Use of male Sprague-Dawley rats, rat pheochromocytoma (PC12), and murine microglial cells (BV-2). Treatment lipopolysaccharides (LPS) were injected into the brain lateral ventricle of the rat. The BV-2 cells were treated by LPS. The PC12 cells were pretreated by or not pretreated by conditioned media and siRNA. METHODS Western blotting was used for analyzing the expression level of Nischarin, pAKT, BAD and Bcl-2. Immunohistochemistry and immunofluorescence were used to perform the morphology and localization of Nischarin. The siRNA could down-regulate the protein level of endogenous Nischarin. RESULTS The expression level of Nischarin was elevated after LPS injection; meanwhile, Nischarin was located in the neuron. Nischarin was involved in regulating the PI3K/PKB patway. CONCLUSION Nischarin might be involved in mediating the process of PI3K/PKB pathway-dependent neuronal apoptosis. After the silencing of Nischarin in cultured PC12 (pheochromocytoma) by siRNA, these results showed that it would induce a reduction of pAKT and Bcl-2 proteins expression; meanwhile, it induces an increase of BAD and active caspase-3.
Collapse
|
40
|
Jain P, Baranwal S, Dong S, Struckhoff AP, Worthylake RA, Alahari SK. Integrin-binding protein nischarin interacts with tumor suppressor liver kinase B1 (LKB1) to regulate cell migration of breast epithelial cells. J Biol Chem 2013; 288:15495-509. [PMID: 23572524 DOI: 10.1074/jbc.m112.418103] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Biallelic inactivation of LKB1, a serine/threonine kinase, has been detected in 30% of lung adenocarcinomas, and inhibition of breast tumor growth has been demonstrated. We have identified the tumor suppressor, Nischarin, as a novel binding partner of LKB1. Our mapping analysis shows that the N terminus of Nischarin interacts with amino acids 44-436 of LKB1. Time lapse microscopy and Transwell migration data show that the absence of both Nischarin and LKB1 from an invasive breast cancer cell line (MDA-MB-231) enhances migration as measured by increased distance and speed of migrating cells. Our data suggest that this is a result of elevated PAK1 and LIMK1 phosphorylation. Moreover, the absence of Nischarin and LKB1 increased tumor growth in vivo. Consistent with this, the percentage of S phase cells was increased, as demonstrated by flow cytometry and enhanced cyclin D1. The absence of Nischarin and LKB1 also led to a dramatic increase in the formation of lung metastases. Our studies, for the first time, demonstrate functional interaction between LKB1 and Nischarin to inhibit cell migration and breast tumor progression. Mechanistically, we show that these two proteins together regulate PAK-LIMK-Cofilin and cyclin D1/CDK4 pathways.
Collapse
Affiliation(s)
- Prachi Jain
- Department of Biochemistry and Molecular Biology, School of Medicine, Louisiana State University Health Science Center, New Orleans, Louisiana 70112, USA
| | | | | | | | | | | |
Collapse
|
41
|
Ghai R, Bugarcic A, Liu H, Norwood SJ, Skeldal S, Coulson EJ, Li SSC, Teasdale RD, Collins BM. Structural basis for endosomal trafficking of diverse transmembrane cargos by PX-FERM proteins. Proc Natl Acad Sci U S A 2013; 110:E643-52. [PMID: 23382219 PMCID: PMC3581954 DOI: 10.1073/pnas.1216229110] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Transit of proteins through the endosomal organelle following endocytosis is critical for regulating the homeostasis of cell-surface proteins and controlling signal transduction pathways. However, the mechanisms that control these membrane-transport processes are poorly understood. The Phox-homology (PX) domain-containing proteins sorting nexin (SNX) 17, SNX27, and SNX31 have emerged recently as key regulators of endosomal recycling and bind conserved Asn-Pro-Xaa-Tyr-sorting signals in transmembrane cargos via an atypical band, 4.1/ezrin/radixin/moesin (FERM) domain. Here we present the crystal structure of the SNX17 FERM domain bound to the sorting motif of the P-selectin adhesion protein, revealing both the architecture of the atypical FERM domain and the molecular basis for recognition of these essential sorting sequences. We further show that the PX-FERM proteins share a promiscuous ability to bind a wide array of putative cargo molecules, including receptor tyrosine kinases, and propose a model for their coordinated molecular interactions with membrane, cargo, and regulatory proteins.
Collapse
Affiliation(s)
| | | | - Huadong Liu
- Department of Biochemistry and
- Siebens Drake Medical Research Institute, University of Western Ontario, London, ON, Canada N6A 5C1
| | | | - Sune Skeldal
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD 4072, Australia; and
| | - Elizabeth J. Coulson
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD 4072, Australia; and
| | - Shawn Shun-Cheng Li
- Department of Biochemistry and
- Siebens Drake Medical Research Institute, University of Western Ontario, London, ON, Canada N6A 5C1
| | | | | |
Collapse
|
42
|
Rac and Rab GTPases dual effector Nischarin regulates vesicle maturation to facilitate survival of intracellular bacteria. EMBO J 2013; 32:713-27. [PMID: 23386062 DOI: 10.1038/emboj.2013.10] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 01/08/2013] [Indexed: 12/31/2022] Open
Abstract
The intracellular pathogenic bacterium Salmonella enterica serovar typhimurium (Salmonella) relies on acidification of the Salmonella-containing vacuole (SCV) for survival inside host cells. The transport and fusion of membrane-bound compartments in a cell is regulated by small GTPases, including Rac and members of the Rab GTPase family, and their effector proteins. However, the role of these components in survival of intracellular pathogens is not completely understood. Here, we identify Nischarin as a novel dual effector that can interact with members of Rac and Rab GTPase (Rab4, Rab14 and Rab9) families at different endosomal compartments. Nischarin interacts with GTP-bound Rab14 and PI(3)P to direct the maturation of early endosomes to Rab9/CD63-containing late endosomes. Nischarin is recruited to the SCV in a Rab14-dependent manner and enhances acidification of the SCV. Depletion of Nischarin or the Nischarin binding partners--Rac1, Rab14 and Rab9 GTPases--reduced the intracellular growth of Salmonella. Thus, interaction of Nischarin with GTPases may regulate maturation and subsequent acidification of vacuoles produced after phagocytosis of pathogens.
Collapse
|
43
|
Jin L, Wessely O, Marcusson EG, Ivan C, Calin GA, Alahari SK. Prooncogenic factors miR-23b and miR-27b are regulated by Her2/Neu, EGF, and TNF-α in breast cancer. Cancer Res 2013; 73:2884-96. [PMID: 23338610 DOI: 10.1158/0008-5472.can-12-2162] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
miRNAs (miR) are a critical class of small (21-25 nucleotides) noncoding endogenous RNAs implicated in gene expression regulation. We identified miR-23b and miR-27b as miRNAs that are highly upregulated in human breast cancer. We found that engineered knockdown of miR-23b and miR-27b substantially repressed breast cancer growth. Nischarin (NISCH) expression was augmented by knockdown of miR-23b as well as miR-27b. Notably, these miRNAs and Nischarin were inversely expressed in human breast cancers, underscoring their biologic relevance. We showed the clinical relevance of the expression of these miRNAs and showed that high expression of miR-23b and miR-27b correlates with poor outcome in breast cancer. Moreover, intraperitoneally delivered anti-miR-27b restored Nischarin expression and decreased tumor burden in a mouse xenograft model of human mammary tumor. Also, we report for the first time that HER2/neu (ERBB2), EGF, and TNF-α promote miR-23b/27b expression through the AKT/NF-κB signaling cascade. Nischarin was found to regulate miR-27b/23b expression through a feedback loop mechanism by suppressing NF-κB phosphorylation. Because anti-miR-27b compounds that suppress miR-27b inhibit tumor growth, the anti-miR-27b seems to be a good candidate for the development of new antitumor therapies.
Collapse
Affiliation(s)
- Lianjin Jin
- Department of Biochemistry and Molecular Biology and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | | | | | | | |
Collapse
|
44
|
Ding Y, Zhang R, Zhang K, Lv X, Chen Y, Li A, Wang L, Zhang X, Xia Q. Nischarin is differentially expressed in rat brain and regulates neuronal migration. PLoS One 2013; 8:e54563. [PMID: 23342172 PMCID: PMC3546990 DOI: 10.1371/journal.pone.0054563] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 12/13/2012] [Indexed: 11/19/2022] Open
Abstract
Nischarin is a protein known to inhibit breast cancer cell motility by regulating the signaling of the Rho GTPase family. However, little is known about its location and function in the nervous system. The aim of the present study was to investigate the regional and cellular expression and functions of Nischarin in the adult rodent brain. As assessed by real-time PCR, Western blot analysis and immunostaining, we found that Nischarin was widely distributed throughout the brain, with a higher expression in the cerebral cortex and hippocampus. Double-labeling showed that Nischarin was expressed in neurons and was mainly located in the perinuclear region and F-actin-rich protrusions. The expression pattern of Nischarin in the brain was thought to be closely associated with its function. This was verified by our findings from cell migration assays that Nischarin regulated neuronal migration. These results provide a preliminary survey of the distribution of Nischarin in different regions and cell types in the rat brain. This might help to elucidate its physiological roles, and to evaluate its potential clinical implications.
Collapse
Affiliation(s)
- Yuemin Ding
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Ruyi Zhang
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Kena Zhang
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyou Lv
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanan Chen
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Aiqing Li
- Gastroenterology Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Linlin Wang
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiong Zhang
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiang Xia
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
- * E-mail:
| |
Collapse
|
45
|
Chua GL, Tang XY, Patra AT, Tan SM, Bhattacharjya S. Structure and binding interface of the cytosolic tails of αXβ2 integrin. PLoS One 2012; 7:e41924. [PMID: 22844534 PMCID: PMC3406025 DOI: 10.1371/journal.pone.0041924] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 06/26/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Integrins are signal transducer proteins involved in a number of vital physiological processes including cell adhesion, proliferation and migration. Integrin molecules are hetero-dimers composed of two distinct subunits, α and β. In humans, 18 α and 8 β subunits are combined into 24 different integrin molecules. Each of the subunit comprises a large extracellular domain, a single pass transmembrane segment and a cytosolic tail (CT). The CTs of integrins are vital for bidirectional signal transduction and in maintaining the resting state of the receptors. A large number of intracellular proteins have been found to interact with the CTs of integrins linking integrins to the cytoskeleton. METHODOLOGY/PRINCIPAL FINDINGS In this work, we have investigated structure and interactions of CTs of the leukocyte specific integrin αXβ2. We determined the atomic resolution structure of a myristoylated CT of αX in perdeuterated dodecylphosphocholine (DPC) by NMR spectroscopy. Our results reveal that the 35-residue long CT of αX adopts an α-helical conformation for residues F4-N17 at the N-terminal region. The remaining residues located at the C-terminal segment of αX delineate a long loop of irregular conformations. A segment of the loop maintains packing interactions with the helical structure by an extended non-polar surface of the αX CT. Interactions between αX and β2 CTs are demonstrated by (15)N-(1)H HSQC NMR experiments. We find that residues constituting the polar face of the helical conformation of αX are involved in interactions with the N-terminal residues of β2 CT. A docked structure of the CT complex indicates that a network of polar and/or salt-bridge interactions may sustain the heteromeric interactions. CONCLUSIONS/SIGNIFICANCE The current study provides important insights into the conservation of interactions and structures among different CTs of integrins.
Collapse
Affiliation(s)
- Geok-Lin Chua
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Xiao-Yan Tang
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Alok Tanala Patra
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Suet-Mien Tan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Surajit Bhattacharjya
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
46
|
Eswaran J, Li DQ, Shah A, Kumar R. Molecular pathways: targeting p21-activated kinase 1 signaling in cancer--opportunities, challenges, and limitations. Clin Cancer Res 2012; 18:3743-9. [PMID: 22595609 DOI: 10.1158/1078-0432.ccr-11-1952] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The evolution of cancer cells involves deregulation of highly regulated fundamental pathways that are central to normal cellular architecture and functions. p21-activated kinase 1 (PAK1) was initially identified as a downstream effector of the GTPases Rac and Cdc42. Subsequent studies uncovered a variety of new functions for this kinase in growth factor and steroid receptor signaling, cytoskeleton remodeling, cell survival, oncogenic transformation, and gene transcription, largely through systematic discovery of its direct, physiologically relevant substrates. PAK1 is widely upregulated in several human cancers, such as hormone-dependent cancer, and is intimately linked to tumor progression and therapeutic resistance. These exciting developments combined with the kinase-independent role of PAK1-centered phenotypic signaling in cancer cells elevated PAK1 as an attractive drug target. Structural and biochemical studies revealed the precise mechanism of PAK1 activation, offering the possibility to develop PAK1-targeted cancer therapeutic approaches. In addition, emerging reports suggest the potential of PAK1 and its specific phosphorylated substrates as cancer prognostic markers. Here, we summarize recent findings about the PAK1 molecular pathways in human cancer and discuss the current status of PAK1-targeted anticancer therapies.
Collapse
Affiliation(s)
- Jeyanthy Eswaran
- McCormick Genomic and Proteomics Center, George Washington University, Washington, DC, USA
| | | | | | | |
Collapse
|
47
|
Briones N, Dinu V. Data mining of high density genomic variant data for prediction of Alzheimer's disease risk. BMC MEDICAL GENETICS 2012; 13:7. [PMID: 22273362 PMCID: PMC3355044 DOI: 10.1186/1471-2350-13-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 01/25/2012] [Indexed: 11/25/2022]
Abstract
Background The discovery of genetic associations is an important factor in the understanding of human illness to derive disease pathways. Identifying multiple interacting genetic mutations associated with disease remains challenging in studying the etiology of complex diseases. And although recently new single nucleotide polymorphisms (SNPs) at genes implicated in immune response, cholesterol/lipid metabolism, and cell membrane processes have been confirmed by genome-wide association studies (GWAS) to be associated with late-onset Alzheimer's disease (LOAD), a percentage of AD heritability continues to be unexplained. We try to find other genetic variants that may influence LOAD risk utilizing data mining methods. Methods Two different approaches were devised to select SNPs associated with LOAD in a publicly available GWAS data set consisting of three cohorts. In both approaches, single-locus analysis (logistic regression) was conducted to filter the data with a less conservative p-value than the Bonferroni threshold; this resulted in a subset of SNPs used next in multi-locus analysis (random forest (RF)). In the second approach, we took into account prior biological knowledge, and performed sample stratification and linkage disequilibrium (LD) in addition to logistic regression analysis to preselect loci to input into the RF classifier construction step. Results The first approach gave 199 SNPs mostly associated with genes in calcium signaling, cell adhesion, endocytosis, immune response, and synaptic function. These SNPs together with APOE and GAB2 SNPs formed a predictive subset for LOAD status with an average error of 9.8% using 10-fold cross validation (CV) in RF modeling. Nineteen variants in LD with ST5, TRPC1, ATG10, ANO3, NDUFA12, and NISCH respectively, genes linked directly or indirectly with neurobiology, were identified with the second approach. These variants were part of a model that included APOE and GAB2 SNPs to predict LOAD risk which produced a 10-fold CV average error of 17.5% in the classification modeling. Conclusions With the two proposed approaches, we identified a large subset of SNPs in genes mostly clustered around specific pathways/functions and a smaller set of SNPs, within or in proximity to five genes not previously reported, that may be relevant for the prediction/understanding of AD.
Collapse
Affiliation(s)
- Natalia Briones
- Computational Biosciences Program, School of Mathematics and Statistical Sciences, Arizona State University, 1711 South Rural Road, Tempe, Arizona 85287-1804, USA
| | | |
Collapse
|
48
|
Insights into the PX (phox-homology) domain and SNX (sorting nexin) protein families: structures, functions and roles in disease. Biochem J 2011; 441:39-59. [DOI: 10.1042/bj20111226] [Citation(s) in RCA: 212] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The mammalian genome encodes 49 proteins that possess a PX (phox-homology) domain, responsible for membrane attachment to organelles of the secretory and endocytic system via binding of phosphoinositide lipids. The PX domain proteins, most of which are classified as SNXs (sorting nexins), constitute an extremely diverse family of molecules that play varied roles in membrane trafficking, cell signalling, membrane remodelling and organelle motility. In the present review, we present an overview of the family, incorporating recent functional and structural insights, and propose an updated classification of the proteins into distinct subfamilies on the basis of these insights. Almost all PX domain proteins bind PtdIns3P and are recruited to early endosomal membranes. Although other specificities and localizations have been reported for a select few family members, the molecular basis for binding to other lipids is still not clear. The PX domain is also emerging as an important protein–protein interaction domain, binding endocytic and exocytic machinery, transmembrane proteins and many other molecules. A comprehensive survey of the molecular interactions governed by PX proteins highlights the functional diversity of the family as trafficking cargo adaptors and membrane-associated scaffolds regulating cell signalling. Finally, we examine the mounting evidence linking PX proteins to different disorders, in particular focusing on their emerging importance in both pathogen invasion and amyloid production in Alzheimer's disease.
Collapse
|
49
|
Chua GL, Tang XY, Amalraj M, Tan SM, Bhattacharjya S. Structures and interaction analyses of integrin αMβ2 cytoplasmic tails. J Biol Chem 2011; 286:43842-43854. [PMID: 22052909 DOI: 10.1074/jbc.m111.280164] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Integrins are heterodimeric (α and β subunits) signal transducer proteins involved in cell adhesions and migrations. The cytosolic tails of integrins are essential for transmitting bidirectional signaling and also implicated in maintaining the resting states of the receptors. In addition, cytosolic tails of integrins often undergo post-translation modifications like phosphorylation. However, the consequences of phosphorylation on the structures and interactions are not clear. The leukocyte-specific integrin αMβ2 is essential for myeloid cell adhesion, phagocytosis, and degranulation. In this work, we determined solution structures of the myristoylated cytosolic tail of αM and a Ser phosphorylated variant in dodecylphosphocholine micelles by NMR spectroscopy. Furthermore, the interactions between non-phosphorylated and phosphorylated αM tails with β2 tail were investigated by NMR and fluorescence resonance energy transfer (FRET). The three-dimensional structures of the 24-residue cytosolic tail of αM or phosphorylated αM are characterized by an N-terminal amphipathic helix and a loop at the C terminus. The residues at the loop are involved in packing interactions with the hydrophobic face of the helix. 15N-1H heteronuclear single quantum coherence experiments identified residues of αM and β2 tails that may be involved in the formation of a tail-tail heterocomplex. We further examined interactions between myristoylated β2 tail in dodecylphosphocholine micelles with dansylated αM tail peptides by FRET. These studies revealed enhanced interactions between αM or phosphorylated αM tails with β2 tail with Kd values ∼5.2±0.6 and ∼4.4±0.7 μm, respectively. Docked structures of tail-tail complexes delineated that the αM/β2 interface at the cytosolic region could be sustained by a network of polar interactions, ionic interactions, and/or hydrogen bonds.
Collapse
Affiliation(s)
- Geok-Lin Chua
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Xiao-Yan Tang
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Monalisa Amalraj
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Suet-Mien Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Surajit Bhattacharjya
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
50
|
Darie CC, Deinhardt K, Zhang G, Cardasis HS, Chao MV, Neubert TA. Identifying transient protein-protein interactions in EphB2 signaling by blue native PAGE and mass spectrometry. Proteomics 2011; 11:4514-28. [PMID: 21932443 DOI: 10.1002/pmic.201000819] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 08/23/2011] [Accepted: 08/31/2011] [Indexed: 11/11/2022]
Abstract
Receptor tyrosine kinases (RTKs) are proteins that upon ligand stimulation undergo dimerization and autophosphorylation. Eph receptors (EphRs) are RTKs that are found in different cell types, from both tissues that are developing and from mature tissues, and play important roles in the development of the central nervous system and peripheral nervous system. EphRs also play roles in synapse formation, neural crest formation, angiogenesis and in remodeling the vascular system. Interaction of EphRs with their ephrin ligands lead to activation of signal transduction pathways and formation of many transient protein-protein interactions that ultimately leads to cytoskeletal remodeling. However, the sequence of events at the molecular level is not well understood. We used blue native PAGE and MS to analyze the transient protein-protein interactions that resulted from the stimulation of EphB2 receptors by their ephrinB1-Fc ligands. We analyzed the phosphotyrosine-containing protein complexes immunoprecipitated from the cell lysates of both unstimulated (-) and ephrinB1-Fc-stimulated (+) NG108 cells. Our experiments allowed us to identify many signaling proteins, either known to be part of EphB2 signaling or new for this pathway, which are involved in transient protein-protein interactions upon ephrinB1-Fc stimulation. These data led us to investigate the roles of proteins such as FAK, WAVEs and Nischarin in EphB2 signaling.
Collapse
Affiliation(s)
- Costel C Darie
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York, NY, USA.
| | | | | | | | | | | |
Collapse
|