1
|
Li Z, Zhou T, Bao Z, Wu M, Mao Y. The Porous SilMA Hydrogel Scaffolds Carrying Dual-Sensitive Paclitaxel Nanoparticles Promote Neuronal Differentiation for Spinal Cord Injury Repair. Tissue Eng Regen Med 2024; 21:809-827. [PMID: 39004636 PMCID: PMC11286913 DOI: 10.1007/s13770-024-00659-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND In the intricate pathological milieu post-spinal cord injury (SCI), neural stem cells (NSCs) frequently differentiate into astrocytes rather than neurons, significantly limiting nerve repair. Hence, the utilization of biocompatible hydrogel scaffolds in conjunction with exogenous factors to foster the differentiation of NSCs into neurons has the potential for SCI repair. METHODS In this study, we engineered a 3D-printed porous SilMA hydrogel scaffold (SM) supplemented with pH-/temperature-responsive paclitaxel nanoparticles (PTX-NPs). We analyzed the biocompatibility of a specific concentration of PTX-NPs and its effect on NSC differentiation. We also established an SCI model to explore the ability of composite scaffolds for in vivo nerve repair. RESULTS The physical adsorption of an optimal PTX-NPs dosage can simultaneously achieve pH/temperature-responsive release and commendable biocompatibility, primarily reflected in cell viability, morphology, and proliferation. An appropriate PTX-NPs concentration can steer NSC differentiation towards neurons over astrocytes, a phenomenon that is also efficacious in simulated injury settings. Immunoblotting analysis confirmed that PTX-NPs-induced NSC differentiation occurred via the MAPK/ERK signaling cascade. The repair of hemisected SCI in rats demonstrated that the composite scaffold augmented neuronal regeneration at the injury site, curtailed astrocyte and fibrotic scar production, and enhanced motor function recovery in rat hind limbs. CONCLUSION The scaffold's porous architecture serves as a cellular and drug carrier, providing a favorable microenvironment for nerve regeneration. These findings corroborate that this strategy amplifies neuronal expression within the injury milieu, significantly aiding in SCI repair.
Collapse
Affiliation(s)
- Zhixiang Li
- School of Life Sciences, Bengbu Medical University, 2600 Donghai Road, Bengbu, 233030, China
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China
| | - Tao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China
| | - Zhengqi Bao
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China
| | - Min Wu
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China.
| | - Yingji Mao
- School of Life Sciences, Bengbu Medical University, 2600 Donghai Road, Bengbu, 233030, China.
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China.
- Anhui Engineering Research Center of Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu, China.
| |
Collapse
|
2
|
Ba-Alawi W, Kadambat Nair S, Li B, Mammoliti A, Smirnov P, Mer AS, Penn LZ, Haibe-Kains B. Bimodal gene expression in cancer patients provides interpretable biomarkers for drug sensitivity. Cancer Res 2022; 82:2378-2387. [PMID: 35536872 DOI: 10.1158/0008-5472.can-21-2395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 02/24/2022] [Accepted: 05/06/2022] [Indexed: 11/16/2022]
Abstract
Identifying biomarkers predictive of cancer cell response to drug treatment constitutes one of the main challenges in precision oncology. Recent large-scale cancer pharmacogenomic studies have opened new avenues of research to develop predictive biomarkers by profiling thousands of human cancer cell lines at the molecular level and screening them with hundreds of approved drugs and experimental chemical compounds. Many studies have leveraged these data to build predictive models of response using various statistical and machine learning methods. However, a common pitfall to these methods is the lack of interpretability as to how they make predictions, hindering the clinical translation of these models. To alleviate this issue, we used the recent logic modeling approach to develop a new machine learning pipeline that explores the space of bimodally expressed genes in multiple large in vitro pharmacogenomic studies and builds multivariate, nonlinear, yet interpretable logic-based models predictive of drug response. The performance of this approach was showcased in a compendium of the three largest in vitro pharmacogenomic data sets to build robust and interpretable models for 101 drugs that span 17 drug classes with high validation rates in independent datasets. These results along with in vivo and clinical validation, support a better translation of gene expression biomarkers between model systems using bimodal gene expression.
Collapse
Affiliation(s)
| | | | - Bo Li
- University of Toronto, Toronto, Canada
| | | | | | | | - Linda Z Penn
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | |
Collapse
|
3
|
Structural basis for p50RhoGAP BCH domain-mediated regulation of Rho inactivation. Proc Natl Acad Sci U S A 2021; 118:2014242118. [PMID: 34006635 DOI: 10.1073/pnas.2014242118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Spatiotemporal regulation of signaling cascades is crucial for various biological pathways, under the control of a range of scaffolding proteins. The BNIP-2 and Cdc42GAP Homology (BCH) domain is a highly conserved module that targets small GTPases and their regulators. Proteins bearing BCH domains are key for driving cell elongation, retraction, membrane protrusion, and other aspects of active morphogenesis during cell migration, myoblast differentiation, and neuritogenesis. We previously showed that the BCH domain of p50RhoGAP (ARHGAP1) sequesters RhoA from inactivation by its adjacent GAP domain; however, the underlying molecular mechanism for RhoA inactivation by p50RhoGAP remains unknown. Here, we report the crystal structure of the BCH domain of p50RhoGAP Schizosaccharomyces pombe and model the human p50RhoGAP BCH domain to understand its regulatory function using in vitro and cell line studies. We show that the BCH domain adopts an intertwined dimeric structure with asymmetric monomers and harbors a unique RhoA-binding loop and a lipid-binding pocket that anchors prenylated RhoA. Interestingly, the β5-strand of the BCH domain is involved in an intermolecular β-sheet, which is crucial for inhibition of the adjacent GAP domain. A destabilizing mutation in the β5-strand triggers the release of the GAP domain from autoinhibition. This renders p50RhoGAP active, thereby leading to RhoA inactivation and increased self-association of p50RhoGAP molecules via their BCH domains. Our results offer key insight into the concerted spatiotemporal regulation of Rho activity by BCH domain-containing proteins.
Collapse
|
4
|
Zhang P, Lim SB, Jiang K, Chew TW, Low BC, Lim CT. Distinct mRNAs in Cancer Extracellular Vesicles Activate Angiogenesis and Alter Transcriptome of Vascular Endothelial Cells. Cancers (Basel) 2021; 13:cancers13092009. [PMID: 33921957 PMCID: PMC8122258 DOI: 10.3390/cancers13092009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/07/2021] [Accepted: 04/19/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Cancer extracellular vesicles (EVs) are implicated in various processes of cancer development, with most of the EV-induced changes attributed to EV proteins and microRNAs. However, the knowledge about the cancer EV-mRNAs remains limited. Here, we have assessed the mRNAs of 61 diverse oncogenes and found half of them, including VEGFA and SNAIL1/2, are abundant in cancer EVs while absent in non-tumorigenic cell-derived EVs. Fluorescent trafficking shows the EV VEGFA mRNAs are translatable after being internalized by the recipient cell. Concomitantly, the cancer EVs induced VEGFA-dependent angiogenesis and upregulated epithelial-mesenchymal transition-related genes. Our findings reveal that the EV-mRNA profile can reflect the cell malignancy, and the intercellular transfer of these mRNAs can contribute toward tumor angiogenesis. Abstract Cancer-derived extracellular vesicles (EVs) have been demonstrated to be implicated in various processes of cancer development, with most of the EV-induced changes attributed to EV-proteins and EV-microRNAs. However, the knowledge about the abundance of cancer EV-mRNAs and their contribution to cancer development remain elusive. Here, we show that mRNAs prevail in cancer EVs as compared with normal EVs, and cancer EVs that carry abundant angiogenic mRNAs activate angiogenesis in human umbilical vein endothelial cells (HUVECs). Specifically, of a gene panel comprising 61 hypoxia-targeted oncogenes, a larger proportion is harbored by cancer EVs (>40%) than normal EVs (14.8%). Fluorescent trafficking indicates cancer EVs deliver translatable mRNAs such as VEGFA to HUVECs, contributing to the activation of VEGFR-dependent angiogenesis and the upregulation of epithelial-mesenchymal transition-related and metabolism-related genes. Overall, our findings provide novel insights into EV-mRNAs and their role in angiogenesis, and has potential for diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Pan Zhang
- NUS Graduate School—Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore;
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore;
| | - Su Bin Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore;
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Kuan Jiang
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore; (K.J.); (T.W.C.)
| | - Ti Weng Chew
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore; (K.J.); (T.W.C.)
| | - Boon Chuan Low
- NUS Graduate School—Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore;
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore; (K.J.); (T.W.C.)
- Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore
- University Scholars Programme, National University of Singapore, Singapore 138593, Singapore
- Correspondence: (B.C.L.); (C.T.L.)
| | - Chwee Teck Lim
- NUS Graduate School—Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore;
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore;
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore; (K.J.); (T.W.C.)
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Correspondence: (B.C.L.); (C.T.L.)
| |
Collapse
|
5
|
Pan M, Chew TW, Wong DCP, Xiao J, Ong HT, Chin JFL, Low BC. BNIP-2 retards breast cancer cell migration by coupling microtubule-mediated GEF-H1 and RhoA activation. SCIENCE ADVANCES 2020; 6:eaaz1534. [PMID: 32789168 PMCID: PMC7399486 DOI: 10.1126/sciadv.aaz1534] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 06/18/2020] [Indexed: 06/11/2023]
Abstract
Microtubules display dynamic turnover during cell migration, leading to cell contractility and focal adhesion maturation regulated by Rho guanosine triphosphatase activity. This interplay between microtubules and actomyosin is mediated by guanine nucleotide exchange factor (GEF)-H1 released after microtubule depolymerization or microtubule disconnection from focal adhesions. However, how GEF-H1 activates Rho upon microtubule disassembly remains elusive. Here, we found that BNIP-2, a BCH domain-containing protein that binds both RhoA and GEF-H1 and traffics with kinesin-1 on microtubules, is important for GEF-H1-driven RhoA activation upon microtubule disassembly. Depletion of BNIP-2 in MDA-MB-231 breast cancer cells decreases RhoA activity and promotes cell migration. Upon nocodazole-induced microtubule disassembly, the interaction between BNIP-2 and GEF-H1 increases, while knockdown of BNIP-2 reduces RhoA activation and cell rounding via uncoupling RhoA-GEF-H1 interaction. Together, these findings revealed that BNIP-2 couples microtubules and focal adhesions via scaffolding GEF-H1 and RhoA, fine-tuning RhoA activity and cell migration.
Collapse
Affiliation(s)
- Meng Pan
- Mechanobiology Institute, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Ti Weng Chew
- Mechanobiology Institute, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Darren Chen Pei Wong
- Mechanobiology Institute, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Jingwei Xiao
- Mechanobiology Institute, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Hui Ting Ong
- Mechanobiology Institute, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Jasmine Fei Li Chin
- Mechanobiology Institute, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Boon Chuan Low
- Mechanobiology Institute, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
- Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543, Singapore
- University Scholars Programme, 18 College Avenue East, Singapore 138593, Singapore
| |
Collapse
|
6
|
Interaction of Nerve Growth Factor β with Adiponectin and SPARC Oppositely Modulates its Biological Activity. Int J Mol Sci 2019; 20:ijms20071541. [PMID: 30934765 PMCID: PMC6479836 DOI: 10.3390/ijms20071541] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 03/22/2019] [Accepted: 03/25/2019] [Indexed: 12/31/2022] Open
Abstract
Both adiponectin and secreted protein, acidic and rich in cysteine (SPARC) inhibit platelet-derived growth factor-BB (PDGF-BB)-induced and basic fibroblast growth factor (FGF2)-induced angiogenic activities through direct and indirect interactions. Although SPARC enhances nerve growth factor (NGF)-dependent neurogenesis, the physical interaction of NGFβ with adiponectin and SPARC remains obscure. Therefore, we first examined their intermolecular interaction by surface plasmon resonance method. NGFβ bound to immobilized SPARC with the binding constant of 59.4 nM, comparable with that of PDGF-BB (24.5 nM) but far less than that of FGF2 (14.4 µM). NGFβ bound to immobilized full length adiponectin with the binding constant of 103 nM, slightly higher than those of PDGF-BB (24.3 nM) and FGF2 (80.2 nM), respectively. Treatment of PC12 cells with SPARC did not cause mitogen-activated protein kinase (MAPK) activation and neurite outgrowth. However, simultaneous addition of SPARC with NGFβ enhanced NGFβ-induced MAPK phosphorylation and neurite outgrowth. Treatment of the cells with adiponectin increased AMP-activated protein kinase (AMPK) phosphorylation but failed to induce neurite outgrowth. Simultaneous treatment with NGFβ and adiponectin significantly reduced cell size and the number of cells with neurite, even after silencing the adiponectin receptors by their siRNA. These results indicate that NGFβ directly interacts with adiponectin and SPARC, whereas these interactions oppositely regulate NGFβ functions.
Collapse
|
7
|
Karabiyik C, Fernandes R, Figueiredo FR, Socodato R, Brakebusch C, Lambertsen KL, Relvas JB, Santos SD. Neuronal Rho GTPase Rac1 elimination confers neuroprotection in a mouse model of permanent ischemic stroke. Brain Pathol 2017; 28:569-580. [PMID: 28960571 DOI: 10.1111/bpa.12562] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 09/21/2017] [Indexed: 01/08/2023] Open
Abstract
The Rho GTPase Rac1 is a multifunctional protein involved in distinct pathways ranging from development to pathology. The aim of the present study was to unravel the contribution of neuronal Rac1 in regulating the response to brain injury induced by permanent focal cerebral ischemia (pMCAO). Our results show that pMCAO significantly increased total Rac1 levels in wild type mice, mainly through rising nuclear Rac1, while a reduction in Rac1 activation was observed. Such changes preceded cell death induced by excitotoxic stress. Pharmacological inhibition of Rac1 in primary neuronal cortical cells prevented the increase in oxidative stress induced after overactivation of glutamate receptors. However, this was not sufficient to prevent the associated neuronal cell death. In contrast, RNAi-mediated knock down of Rac1 in primary cortical neurons prevented cell death elicited by glutamate excitotoxicity and decreased the activity of NADPH oxidase. To test whether in vivo down regulation of neuronal Rac1 was neuroprotective after pMCAO, we used tamoxifen-inducible neuron-specific conditional Rac1-knockout mice. We observed a significant 50% decrease in brain infarct volume of knockout mice and a concomitant increase in HIF-1α expression compared to littermate control mice, demonstrating that ablation of Rac1 in neurons is neuroprotective. Transmission electron microscopy performed in the ischemic brain showed that lysosomes in the infarct of Rac1- knockout mice were preserved at similar levels to those of non-infarcted tissue, while littermate mice displayed a decrease in the number of lysosomes, further corroborating the notion that Rac1 ablation in neurons is neuroprotective. Our results demonstrate that Rac1 plays important roles in the ischemic pathological cascade and that modulation of its levels is of therapeutic interest.
Collapse
Affiliation(s)
- Cansu Karabiyik
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Glial Cell Biology, IBMC- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Rui Fernandes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,HEMS, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Francisco Rosário Figueiredo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,HEMS, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Renato Socodato
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Glial Cell Biology, IBMC- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Cord Brakebusch
- Biotech Research and Innovation Center, University of Copenhagen, Denmark
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark.,Department of Neurology, Odense University Hospital, Odence C, Denmark.,BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - João Bettencourt Relvas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Glial Cell Biology, IBMC- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Sofia Duque Santos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Glial Cell Biology, IBMC- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| |
Collapse
|
8
|
Azoulay-Alfaguter I, Strazza M, Mor A. Chaperone-mediated specificity in Ras and Rap signaling. Crit Rev Biochem Mol Biol 2014; 50:194-202. [PMID: 25488471 DOI: 10.3109/10409238.2014.989308] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ras and Rap proteins are closely related small guanosine triphosphatase (GTPases) that share similar effector-binding domains but operate in a very different signaling networks; Ras has a dominant role in cell proliferation, while Rap mediates cell adhesion. Ras and Rap proteins are regulated by several shared processes such as post-translational modification, phosphorylation, activation by guanine exchange factors and inhibition by GTPase-activating proteins. Sub-cellular localization and trafficking of these proteins to and from the plasma membrane are additional important regulatory features that impact small GTPases function. Despite its importance, the trafficking mechanisms of Ras and Rap proteins are not completely understood. Chaperone proteins play a critical role in trafficking of GTPases and will be the focus of the discussion in this work. We will review several aspects of chaperone biology focusing on specificity toward particular members of the small GTPase family. Understanding this specificity should provide key insights into drug development targeting individual small GTPases.
Collapse
|
9
|
Yi P, Chew LL, Zhang Z, Ren H, Wang F, Cong X, Zheng L, Luo Y, Ouyang H, Low BC, Zhou YT. KIF5B transports BNIP-2 to regulate p38 mitogen-activated protein kinase activation and myoblast differentiation. Mol Biol Cell 2014; 26:29-42. [PMID: 25378581 PMCID: PMC4279227 DOI: 10.1091/mbc.e14-03-0797] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cdo bridges scaffold proteins BNIP-2 and JLP to activate p38MAPK during myoblast differentiation. KIF5B is a novel interacting partner of BNIP-2 and promotes myogenic differentiation. KIF5B-dependent transport of BNIP-2 is essential for its promyogenic effects. The Cdo-p38MAPK (p38 mitogen-activated protein kinase) signaling pathway plays important roles in regulating skeletal myogenesis. During myogenic differentiation, the cell surface receptor Cdo bridges scaffold proteins BNIP-2 and JLP and activates p38MAPK, but the spatial-temporal regulation of this process is largely unknown. We here report that KIF5B, the heavy chain of kinesin-1 motor, is a novel interacting partner of BNIP-2. Coimmunoprecipitation and far-Western study revealed that BNIP-2 directly interacted with the motor and tail domains of KIF5B via its BCH domain. By using a range of organelle markers and live microscopy, we determined the endosomal localization of BNIP-2 and revealed the microtubule-dependent anterograde transport of BNIP-2 in C2C12 cells. The anterograde transport of BNIP-2 was disrupted by a dominant-negative mutant of KIF5B. In addition, knockdown of KIF5B causes aberrant aggregation of BNIP-2, confirming that KIF5B is critical for the anterograde transport of BNIP-2 in cells. Gain- and loss-of-function experiments further showed that KIF5B modulates p38MAPK activity and in turn promotes myogenic differentiation. Of importance, the KIF5B-dependent anterograde transport of BNIP-2 is critical for its promyogenic effects. Our data reveal a novel role of KIF5B in the spatial regulation of Cdo–BNIP-2–p38MAPK signaling and disclose a previously unappreciated linkage between the intracellular transporting system and myogenesis regulation.
Collapse
Affiliation(s)
- Peng Yi
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Li Li Chew
- Department of Biological Sciences and Mechanobiology Institute, National University of Singapore, 117411 Singapore
| | - Ziwang Zhang
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hao Ren
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Feiya Wang
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoxia Cong
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Liling Zheng
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and
| | - Yan Luo
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and
| | - Hongwei Ouyang
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Boon Chuan Low
- Department of Biological Sciences and Mechanobiology Institute, National University of Singapore, 117411 Singapore
| | - Yi Ting Zhou
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
10
|
Chew TW, Liu XJ, Liu L, Spitsbergen JM, Gong Z, Low BC. Crosstalk of Ras and Rho: activation of RhoA abates Kras-induced liver tumorigenesis in transgenic zebrafish models. Oncogene 2013; 33:2717-27. [PMID: 23812423 DOI: 10.1038/onc.2013.240] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 04/22/2013] [Accepted: 05/03/2013] [Indexed: 12/15/2022]
Abstract
RAS and Rho small GTPases are key molecular switches that control cell dynamics, cell growth and tissue development through their distinct signaling pathways. Although much has been learnt about their individual functions in both cell and animal models, the physiological and pathophysiological consequences of their signaling crosstalk in multi-cellular context in vivo remain largely unknown, especially in liver development and liver tumorigenesis. Furthermore, the roles of RhoA in RAS-mediated transformation and their crosstalk in vitro remain highly controversial. When challenged with carcinogens, zebrafish developed liver cancer that resembles the human liver cancer both molecularly and histopathologically. Capitalizing on the growing importance and relevance of zebrafish (Danio rerio) as an alternate cancer model, we have generated liver-specific, Tet-on-inducible transgenic lines expressing oncogenic Kras(G12V), RhoA, constitutively active RhoA(G14V) or dominant-negative RhoA(T19N). Double-transgenic lines expressing Kras(G12V) with one of the three RhoA genes were also generated. Based on quantitative bioimaging and molecular markers for genetic and signaling aberrations, we showed that the induced expression of oncogenic Kras during early development led to liver enlargement and hepatocyte proliferation, associated with elevated Erk phosphorylation, activation of Akt2 and modulation of its two downstream targets, p21Cip and S6 kinase. Such an increase in liver size and Akt2 expression was augmented by dominant-negative RhoA(T19N), but was abrogated by the constitutive-active RhoA(G14V). Consequently, induced expression of the oncogenic Kras in adult transgenic fish led to the development of hepatocellular carcinomas. Survival studies further revealed that the co-expression of dominant-negative RhoA(T19N) with oncogenic Kras increased the mortality rate compared with the other single or double-transgenic lines. This study provides evidence of the previously unappreciated signaling crosstalk between Kras and RhoA in regulating liver overgrowth and liver tumorigenesis. Our results also implicate that activating Rho could be beneficial to suppress the Kras-induced liver malignancies.
Collapse
Affiliation(s)
- T W Chew
- 1] Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore, Singapore [2] Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - X J Liu
- Molecular Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - L Liu
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - J M Spitsbergen
- Department of Microbiology and Marine and Freshwater Biomedical Sciences Center, Oregon State University, Corvallis, OR, USA
| | - Z Gong
- Molecular Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - B C Low
- 1] Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore, Singapore [2] Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|