1
|
Rosas PC, Solaro RJ. p21-Activated Kinase 1 (Pak1) as an Element in Functional and Dysfunctional Interplay Among the Myocardium, Adipose Tissue, and Pancreatic Beta Cells. Compr Physiol 2025; 15:e70006. [PMID: 40065530 PMCID: PMC11894248 DOI: 10.1002/cph4.70006] [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: 12/13/2024] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025]
Abstract
This review focuses on p21-activated kinase 1 (Pak1), a multifunctional, highly conserved enzyme that regulates multiple downstream effectors present in many tissues. Upstream signaling via Ras-related small G-proteins, Cdc42/Rac1 promotes the activity of Pak1. Our hypothesis is that this signaling cascade is an important element in communication among the myocardium, adipose tissue, and pancreatic β-cells. Evidence indicates that a shared property of these tissues is that structure/function stability requires homeostatic Pak1 activity. Increases or decreases in Pak1 activity may promote dysfunction or increase susceptibility to stressors. Evidence that increased levels of Pak1 activity may be protective provides support for efforts to develop therapeutic approaches activating Pak1 with potential use in prevalent disorders associated with obesity, diabetes, and myocardial dysfunction. On the other hand, since increased Pak1 activity is associated with cancer progression, there has been a significant effort to develop Pak1 inhibitors. These opposing therapeutic approaches highlight the need for a deep understanding of Pak1 signaling in relation to the development of effective and selective therapies with minimal or absent off-target effects.
Collapse
Affiliation(s)
- Paola C. Rosas
- Department of Pharmacy Practice, College of PharmacyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - R. John Solaro
- Department of Physiology and Biophysics, College of MedicineUniversity of Illinois at ChicagoChicagoIllinoisUSA
| |
Collapse
|
2
|
He H, Jiang T, Ding M, Zhu Y, Xu X, Huang Y, Yu W, Ou H. Nox1/PAK1 is required for angiotensin II-induced vascular inflammation and abdominal aortic aneurysm formation. Redox Biol 2025; 79:103477. [PMID: 39721498 PMCID: PMC11732235 DOI: 10.1016/j.redox.2024.103477] [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: 07/04/2024] [Revised: 10/27/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
NADPH oxidase 1 (Nox1) is a major isoform of Nox in vascular smooth muscle cells (VSMCs). VSMC activation and extracellular matrix (ECM) remodelling induce abdominal aortic aneurysm (AAA). In this study, we aim to determine the role of Nox1 in the progression of AAA and explore the underling mechanism. ApoE-/-Nox1SMCko mice in which the Nox1 gene was smooth muscle cell (SMC)-specifically deleted in ApoE-/- background, were infused with angiotensin II (Ang II) for 28 days. We found the Nox1 deficiency reduced AAA formation and increased survival compared with ApoE-/-Nox1y/flox mice. Abdominal aortic ROS and monocyts/macrophages were reduced in the ApoE-/-Nox1SMCko mice after Ang II-infusion. The SMC-specific Nox1 deletion caused less elastin fragments and lower matrix metalloproteinase (MMP) activities in the abdominal aorta. Further, we found the Nox1 protein interacted with p21-activated kinase 1 (PAK1) in Ang II-stimulated VSMCs. The PAK1, controlled by Nox1/ROS, promoted VSMC proliferation, migration and differentiation; this is associated with increased activities of vimentin and cofilin, and cytoskeleton modulation. Moreover, we found that the Nox1/PAK1 activated the downstream MAPKs (ERK1/2, p38 and JNKs) and NF-κB, and upregulated Sp1-mediated MMP2 expression upon Ang II-stimulation. Finally, overexpression of PAK1 in the ApoE-/-Nox1SMCko mice increased vascular elastic fibre degradation, pro-inflammatory cytokine expression and AAA incidence. Therefore, we conclude that Nox1, together with PAK1, facilitates Ang II-induced VSMC activation, vascular inflammation and ECM remodelling, and thus potentiates the AAA formation.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/etiology
- Aortic Aneurysm, Abdominal/genetics
- Angiotensin II/adverse effects
- NADPH Oxidase 1/metabolism
- NADPH Oxidase 1/genetics
- Mice
- p21-Activated Kinases/metabolism
- p21-Activated Kinases/genetics
- Inflammation/metabolism
- Inflammation/pathology
- Reactive Oxygen Species/metabolism
- Mice, Knockout
- Myocytes, Smooth Muscle/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Disease Models, Animal
- Humans
- Male
- Signal Transduction
Collapse
Affiliation(s)
- Hui He
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Tianyu Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Meng Ding
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Yuan Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Xiaoting Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Yashuang Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Wenfeng Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Hailong Ou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China.
| |
Collapse
|
3
|
Kauer SD, Benson CA, Carrara JM, Tarafder AA, Ibrahim YH, Estacion MA, Waxman SG, Tan AM. PAK1 inhibition with Romidepsin attenuates H-reflex hyperexcitability after spinal cord injury. J Physiol 2024; 602:5061-5081. [PMID: 39231098 DOI: 10.1113/jp284976] [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: 05/12/2023] [Accepted: 08/16/2024] [Indexed: 09/06/2024] Open
Abstract
Hyperreflexia associated with spasticity is a prevalent neurological condition characterized by excessive and exaggerated reflex responses to stimuli. Hyperreflexia can be caused by several diseases including multiple sclerosis, stroke and spinal cord injury (SCI). Although we have previously identified the contribution of the RAC1-PAK1 pathway underlying spinal hyperreflexia with SCI-induced spasticity, a feasible druggable target has not been validated. To assess the utility of targeting PAK1 to attenuate H-reflex hyperexcitability, we administered Romidepsin, a clinically available PAK1 inhibitor, in Thy1-YFP reporter mice. We performed longitudinal EMG studies with a study design that allowed us to assess pathological H-reflex changes and drug intervention effects over time, before and after contusive SCI. As expected, our results show a significant loss of rate-dependent depression - an indication of hyperreflexia and spasticity - 1 month following SCI as compared with baseline, uninjured controls (or before injury). Romidepsin treatment reduced signs of hyperreflexia in comparison with control cohorts and in pre- and post-drug intervention in SCI animals. Neuroanatomical study further confirmed drug response, as romidepsin treatment also reduced the presence of SCI-induced dendritic spine dysgenesis on α-motor neurons. Taken together, our findings extend previous work demonstrating the utility of targeting PAK1 activity in SCI-induced spasticity and support the novel use of romidepsin as an effective tool for managing spasticity. KEY POINTS: PAK1 plays a role in contributing to the development of spinal cord injury (SCI)-induced spasticity by contributing to dendritic spine dysgenesis. In this study, we explored the preclinical utility of inhibiting PAK1 to reduce spasticity and dendritic spine dysgenesis in an SCI mouse model. Romidepsin is a PAK1 inhibitor approved in the US in 2009 for the treatment of cutaneous T-cell lymphoma. Here we show that romidepsin treatment after SCI reduced SCI-induced H-reflex hyperexcitability and abnormal α-motor neuron spine morphology. This study provides compelling evidence that romidepsin may be a promising therapeutic approach for attenuating SCI-induced spasticity.
Collapse
Affiliation(s)
- Sierra D Kauer
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Curtis A Benson
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Jennifer M Carrara
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Afrin A Tarafder
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Youssef H Ibrahim
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Maile A Estacion
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Andrew M Tan
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
4
|
Pagliari S, Vinarsky V, Martino F, Perestrelo AR, Oliver De La Cruz J, Caluori G, Vrbsky J, Mozetic P, Pompeiano A, Zancla A, Ranjani SG, Skladal P, Kytyr D, Zdráhal Z, Grassi G, Sampaolesi M, Rainer A, Forte G. YAP-TEAD1 control of cytoskeleton dynamics and intracellular tension guides human pluripotent stem cell mesoderm specification. Cell Death Differ 2021; 28:1193-1207. [PMID: 33116297 PMCID: PMC8027678 DOI: 10.1038/s41418-020-00643-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/24/2020] [Accepted: 10/08/2020] [Indexed: 12/22/2022] Open
Abstract
The tight regulation of cytoskeleton dynamics is required for a number of cellular processes, including migration, division and differentiation. YAP-TEAD respond to cell-cell interaction and to substrate mechanics and, among their downstream effects, prompt focal adhesion (FA) gene transcription, thus contributing to FA-cytoskeleton stability. This activity is key to the definition of adult cell mechanical properties and function. Its regulation and role in pluripotent stem cells are poorly understood. Human PSCs display a sustained basal YAP-driven transcriptional activity despite they grow in very dense colonies, indicating these cells are insensitive to contact inhibition. PSC inability to perceive cell-cell interactions can be restored by tampering with Tankyrase enzyme, thus favouring AMOT inhibition of YAP function. YAP-TEAD complex is promptly inactivated when germ layers are specified, and this event is needed to adjust PSC mechanical properties in response to physiological substrate stiffness. By providing evidence that YAP-TEAD1 complex targets key genes encoding for proteins involved in cytoskeleton dynamics, we suggest that substrate mechanics can direct PSC specification by influencing cytoskeleton arrangement and intracellular tension. We propose an aberrant activation of YAP-TEAD1 axis alters PSC potency by inhibiting cytoskeleton dynamics, thus paralyzing the changes in shape requested for the acquisition of the given phenotype.
Collapse
Affiliation(s)
- Stefania Pagliari
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic.
| | - Vladimir Vinarsky
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
- Competence Center for Mechanobiology in Regenerative Medicine, INTERREG ATCZ133, CZ-62500, Brno, Czech Republic
| | - Fabiana Martino
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
- Faculty of Medicine, Department of Biology, Masaryk University, CZ-62500, Brno, Czech Republic
| | - Ana Rubina Perestrelo
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
| | - Jorge Oliver De La Cruz
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
- Competence Center for Mechanobiology in Regenerative Medicine, INTERREG ATCZ133, CZ-62500, Brno, Czech Republic
| | - Guido Caluori
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
- Central European Institute of Technology, Masaryk University, CZ-62500, Brno, Czech Republic
| | - Jan Vrbsky
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
| | - Pamela Mozetic
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
| | - Antonio Pompeiano
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic
| | | | - Sri Ganji Ranjani
- Central European Institute of Technology, Masaryk University, CZ-62500, Brno, Czech Republic
| | - Petr Skladal
- Central European Institute of Technology, Masaryk University, CZ-62500, Brno, Czech Republic
| | - Dan Kytyr
- Czech Academy of Sciences, Institute of Theoretical and Applied Mechanics, 190 00, Prague 9, Czech Republic
| | - Zbyněk Zdráhal
- Central European Institute of Technology, Masaryk University, CZ-62500, Brno, Czech Republic
| | - Gabriele Grassi
- Department of Life Sciences, Cattinara University Hospital, Trieste University, I-34149, Trieste, Italy
| | - Maurilio Sampaolesi
- Department of Development and Regeneration, KU Leuven, 3000, Leuven, Belgium
| | - Alberto Rainer
- Università Campus Bio-Medico di Roma, Rome, Italy
- Institute of Nanotechnology (NANOTEC), National Research Council, c/o Campus EcoTekne, via Monteroni, 73100, Lecce, Italy
| | - Giancarlo Forte
- International Clinical Research Center (ICRC) of St Anne's University Hospital, CZ-65691, Brno, Czech Republic.
- Competence Center for Mechanobiology in Regenerative Medicine, INTERREG ATCZ133, CZ-62500, Brno, Czech Republic.
- Department of Biomaterials Science, Institute of Dentistry, University of Turku, FI-20014, Turku, Finland.
| |
Collapse
|
5
|
Effects of acetylation on dissociation and phosphorylation of actomyosin in postmortem ovine muscle during incubation at 4 °C in vitro. Food Chem 2021; 356:129696. [PMID: 33838605 DOI: 10.1016/j.foodchem.2021.129696] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022]
Abstract
This study aimed to assess the effects of acetylation levels on actomyosin disassociation and phosphorylation of lamb during incubation at 4 °C. Samples of whole proteins from lamb longissimus thoracis muscles were prepared and assigned into three treatments (high, middle and low acetylation groups). The results showed that deacetylation of myosin heavy chain and actin was inhibited by lysine deacetylase inhibitor trichostatin A and nicotinamide in this study. Phosphorylation levels of myosin heavy chain and actin were inhibited by their acetylation during incubation in vitro. Actomyosin disassociation degree in high acetylation group was significantly lower than that in middle and low acetylation groups (P < 0.05). The ATPase activity in high acetylation group was significantly higher than that in middle and low acetylation groups (P < 0.05). In conclusion, acetylation of myosin heavy chain and actin inhibited actomyosin dissociation by inhibiting their phosphorylation at 4 °C in vitro.
Collapse
|
6
|
Feng X, Zhang H, Meng L, Song H, Zhou Q, Qu C, Zhao P, Li Q, Zou C, Liu X, Zhang Z. Hypoxia-induced acetylation of PAK1 enhances autophagy and promotes brain tumorigenesis via phosphorylating ATG5. Autophagy 2021; 17:723-742. [PMID: 32186433 PMCID: PMC8032228 DOI: 10.1080/15548627.2020.1731266] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/07/2020] [Accepted: 02/14/2020] [Indexed: 01/07/2023] Open
Abstract
Although the treatment of brain tumors by targeting kinase-regulated macroautophagy/autophagy, is under investigation, the precise mechanism underlying autophagy initiation and its significance in glioblastoma (GBM) remains to be defined. Here, we report that PAK1 (p21 [RAC1] activated kinase 1) is significantly upregulated and promotes GBM development. The Cancer Genome Atlas analysis suggests that the oncogenic role of PAK1 in GBM is mainly associated with autophagy. Subsequent experiments demonstrate that PAK1 indeed serves as a positive modulator for hypoxia-induced autophagy in GBM. Mechanistically, hypoxia induces ELP3-mediated PAK1 acetylation at K420, which suppresses the dimerization of PAK1 and enhances its activity, thereby leading to subsequent PAK1-mediated ATG5 (autophagy related 5) phosphorylation at the T101 residue. This event not only protects ATG5 from ubiquitination-dependent degradation but also increases the affinity between the ATG12-ATG5 complex and ATG16L1 (autophagy related 16 like 1). Consequently, ELP3-dependent PAK1 (K420) acetylation and PAK1-mediated ATG5 (T101) phosphorylation are required for hypoxia-induced autophagy and brain tumorigenesis by promoting autophagosome formation. Silencing PAK1 with shRNA or small molecule inhibitor FRAX597 potentially blocks autophagy and GBM growth. Furthermore, SIRT1-mediated PAK1-deacetylation at K420 hinders autophagy and GBM growth. Clinically, the levels of PAK1 (K420) acetylation significantly correlate with the expression of ATG5 (T101) phosphorylation in GBM patients. Together, this report uncovers that the acetylation modification and kinase activity of PAK1 plays an instrumental role in hypoxia-induced autophagy initiation and maintaining GBM growth. Therefore, PAK1 and its regulator in the autophagy pathway might represent potential therapeutic targets for GBM treatment.Abbreviations: 3-MA: 3-methyladenine; Ac-CoA: acetyl coenzyme A; ATG5: autophagy related 5; ATG16L1, autophagy related 16 like 1; BafA1: bafilomycin A1; CDC42: cell division cycle 42; CGGA: Chinese Glioma Genome Atlas; CHX, cycloheximide; ELP3: elongator acetyltransferase complex subunit 3; GBM, glioblastoma; HBSS: Hanks balanced salts solution; MAP1LC3B/LC3: microtubule associated protein 1 light chain 3 beta; MAP2K1: mitogen-activated protein kinase kinase 1; MAPK14, mitogen-activated protein kinase 14; PAK1: p21 (RAC1) activated kinase 1; PDK1: pyruvate dehydrogenase kinase 1; PGK1, phosphoglycerate kinase 1; PTMs: post-translational modifications; RAC1: Rac family small GTPase 1; SQSTM1: sequestosome 1; TCGA, The Cancer Genome Atlas.
Collapse
Affiliation(s)
- Xing Feng
- The Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guangxi Neurological Diseases Clinical Research Center, Guilin, Guangxi, China
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Heng Zhang
- Department of Histology and Embryology, Xiang Ya School of Medicine, Central South University, Changsha, China
| | - Lingbing Meng
- Neurology Department, Beijing Hospital, National Center of Gerontology, Beijing
| | - Huiwen Song
- Department of Cardiology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Qingxin Zhou
- Department of Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, China
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Chao Qu
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
| | - Pan Zhao
- Clinical Medical Research Center, the First Affiliated Hospital of Southern University of Science and Technology, the Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, China
- Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, China
| | - Qinghua Li
- Department of Neurology, the affiliated hospital of Guilin Medical University, Guangxi, China
| | - Chang Zou
- Clinical Medical Research Center, the First Affiliated Hospital of Southern University of Science and Technology, the Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, China
- Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, China
| | - Xing Liu
- Beijing Neurosurgical Institute, Beijing, China
| | - Zhiyong Zhang
- The Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guangxi Neurological Diseases Clinical Research Center, Guilin, Guangxi, China
- Department of Surgery, Robert-Wood-Johnson Medical School University Hospital, Rutgers University, New Brunswick, New Jersey, USA
| |
Collapse
|
7
|
Ben-Shmuel A, Sabag B, Biber G, Barda-Saad M. The Role of the Cytoskeleton in Regulating the Natural Killer Cell Immune Response in Health and Disease: From Signaling Dynamics to Function. Front Cell Dev Biol 2021; 9:609532. [PMID: 33598461 PMCID: PMC7882700 DOI: 10.3389/fcell.2021.609532] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/11/2021] [Indexed: 01/13/2023] Open
Abstract
Natural killer (NK) cells are innate lymphoid cells, which play key roles in elimination of virally infected and malignant cells. The balance between activating and inhibitory signals derived from NK surface receptors govern the NK cell immune response. The cytoskeleton facilitates most NK cell effector functions, such as motility, infiltration, conjugation with target cells, immunological synapse assembly, and cytotoxicity. Though many studies have characterized signaling pathways that promote actin reorganization in immune cells, it is not completely clear how particular cytoskeletal architectures at the immunological synapse promote effector functions, and how cytoskeletal dynamics impact downstream signaling pathways and activation. Moreover, pioneering studies employing advanced imaging techniques have only begun to uncover the architectural complexity dictating the NK cell activation threshold; it is becoming clear that a distinct organization of the cytoskeleton and signaling receptors at the NK immunological synapse plays a decisive role in activation and tolerance. Here, we review the roles of the actin cytoskeleton in NK cells. We focus on how actin dynamics impact cytolytic granule secretion, NK cell motility, and NK cell infiltration through tissues into inflammatory sites. We will also describe the additional cytoskeletal components, non-muscle Myosin II and microtubules that play pivotal roles in NK cell activity. Furthermore, special emphasis will be placed on the role of the cytoskeleton in assembly of immunological synapses, and how mutations or downregulation of cytoskeletal accessory proteins impact NK cell function in health and disease.
Collapse
Affiliation(s)
- Aviad Ben-Shmuel
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Batel Sabag
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Guy Biber
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Mira Barda-Saad
- Laboratory of Molecular and Applied Immunology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
8
|
PAK1 Positively Regulates Oligodendrocyte Morphology and Myelination. J Neurosci 2021; 41:1864-1877. [PMID: 33478987 DOI: 10.1523/jneurosci.0229-20.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 01/04/2021] [Accepted: 01/09/2021] [Indexed: 02/06/2023] Open
Abstract
The actin cytoskeleton is crucial for oligodendrocyte differentiation and myelination. Here we show that p21-activated kinase 1 (PAK1), a well-known actin regulator, promotes oligodendrocyte morphologic change and myelin production in the CNS. A combination of in vitro and in vivo models demonstrated that PAK1 is expressed throughout the oligodendrocyte lineage with highest expression in differentiated oligodendrocytes. Inhibiting PAK1 early in oligodendrocyte development decreased oligodendrocyte morphologic complexity and altered F-actin spreading at the tips of oligodendrocyte progenitor cell processes. Constitutively activating AKT in oligodendrocytes in male and female mice, which leads to excessive myelin wrapping, increased PAK1 expression, suggesting an impact of PAK1 during active myelin wrapping. Furthermore, constitutively activating PAK1 in oligodendrocytes in zebrafish led to an increase in myelin internode length while inhibiting PAK1 during active myelination decreased internode length. As myelin parameters influence conduction velocity, these data suggest that PAK1 may influence communication within the CNS. These data support a model in which PAK1 is a positive regulator of CNS myelination.SIGNIFICANCE STATEMENT Myelin is a critical component of the CNS that provides metabolic support to neurons and also facilitates communication between cells in the CNS. Recent data demonstrate that actin dynamics drives myelin wrapping, but how actin is regulated during myelin wrapping is unknown. The authors investigate the role of the cytoskeletal modulator PAK1 during differentiation and myelination by oligodendrocytes, the myelinating cells of the CNS. They demonstrate that PAK1 promotes oligodendrocyte differentiation and myelination by modulating the cytoskeleton and thereby internode length, thus playing a critical role in the function of the CNS.
Collapse
|
9
|
Parreno J, Amadeo MB, Kwon EH, Fowler VM. Tropomyosin 3.1 Association With Actin Stress Fibers is Required for Lens Epithelial to Mesenchymal Transition. Invest Ophthalmol Vis Sci 2021; 61:2. [PMID: 32492110 PMCID: PMC7415280 DOI: 10.1167/iovs.61.6.2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Purpose Epithelial to mesenchymal transition (EMT) is a cause of anterior and posterior subcapsular cataracts. Central to EMT is the formation of actin stress fibers. Selective targeting of actin stress fiber-associated tropomyosin (Tpm) in epithelial cells may be a means to prevent stress fiber formation and repress lens EMT. Methods We identified Tpm isoforms in mouse immortalized lens epithelial cells and epithelial and fiber cells from whole lenses by semi-quantitative reverse transcription-polymerase chain reaction (RT-PCR) followed Sanger sequencing. We focused on the role of one particular tropomyosin isoform, Tpm3.1, in EMT. To induce EMT, we treated cells or native lenses with TGFβ2. To test the function of Tpm3.1, we exposed cells or whole lenses to a Tpm3.1-specific chemical inhibitor, TR100, as well as investigated lenses from Tpm3.1 knockout mice. We examined stress fiber formation by confocal microscopy and assessed EMT progression by analysis of alpha-smooth muscle actin (αSMA) mRNA (real-time RT-PCR), and protein (Western immunoassay [WES]). Results Lens epithelial cells express eight Tpm isoforms. Cell culture studies showed that TGFβ2 treatment results in the upregulation of Tpm3.1, which associates with actin in stress fibers. TR100 prevents stress fiber formation and reduces αSMA in TGFβ2-treated cells. Using an ex vivo lens culture model, TGFβ2 treatment results in stress fiber formation at the basal regions of the epithelial cells. Genetic knockout of Tpm3.1 or treatment of lenses with TR100 prevents basal stress fiber formation and reduces epithelial αSMA levels. Conclusions Targeting specific stress fiber associated tropomyosin isoform, Tpm3.1, is a means to repress lens EMT.
Collapse
|
10
|
Vermeulen S, Roumans N, Honig F, Carlier A, Hebels DG, Eren AD, Dijke PT, Vasilevich A, de Boer J. Mechanotransduction is a context-dependent activator of TGF-β signaling in mesenchymal stem cells. Biomaterials 2020; 259:120331. [DOI: 10.1016/j.biomaterials.2020.120331] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/15/2020] [Accepted: 08/13/2020] [Indexed: 02/08/2023]
|
11
|
Nair MG, Prabhu JS, Korlimarla A, Rajarajan S, P S H, Kaul R, Alexander A, Raghavan R, B S S, T S S. miR-18a activates Wnt pathway in ER-positive breast cancer and is associated with poor prognosis. Cancer Med 2020; 9:5587-5597. [PMID: 32543775 PMCID: PMC7402845 DOI: 10.1002/cam4.3183] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 04/30/2020] [Accepted: 05/02/2020] [Indexed: 12/24/2022] Open
Abstract
Despite the established benefits of long‐term endocrine therapy, women with hormone receptor‐positive breast cancer remain at risk for late relapse. The basis of this is multi‐factorial including genetic, epigenetic, and host factors. In this study we have explored the epigenetic regulation of estrogen receptor (ER)‐dependent molecular and cellular phenotype by hsa‐miR‐18a‐5p using well‐established human ER‐positive (ER+) breast cancer cell lines. miR‐18a was overexpressed in MCF7 and ZR‐75‐1 and this led to an increase in the proliferative ability of the cells and concurrently resulted in decreased expression of luminal markers and higher expression of the basal marker, cytokeratin 14. The cells became more migratory with a significant repression of E‐cadherin and activation of the Wnt noncanonical pathway. We observed an activation of the planar cell polarity (PCP) pathway with increased activation of JNK pathway and eventually change in actin dynamics. There was increased F‐actin polymerization in cells with higher expression of miR‐18a. Examination of miR‐18a expression in a set of human ER+ breast cancer specimens showed a negative correlation between miR‐18a and ESR1 transcripts as well as ER protein. Kaplan‐Meier survival analysis of the cohort stratified by tumor hsa‐miR‐18a‐5p levels produced significant differences in disease‐free survival (log rank P < .05). This observation was independently validated in the METABRIC cohort. These data provide support for a role of hsa‐miR‐18a‐5p in altering the proliferative and migratory behavior of ER+ cells and its potential utility as a prognostic marker in clinical ER+ breast cancers.
Collapse
Affiliation(s)
- Madhumathy G Nair
- Division of Molecular Medicine, St. John's Research Institute, Bangalore, India
| | - Jyothi S Prabhu
- Division of Molecular Medicine, St. John's Research Institute, Bangalore, India
| | - Aruna Korlimarla
- Division of Molecular Medicine, St. John's Research Institute, Bangalore, India
| | - Savitha Rajarajan
- Division of Molecular Medicine, St. John's Research Institute, Bangalore, India
| | - Hari P S
- Division of Molecular Medicine, St. John's Research Institute, Bangalore, India
| | - Roma Kaul
- Division of Molecular Medicine, St. John's Research Institute, Bangalore, India
| | - Annie Alexander
- Division of Molecular Medicine, St. John's Research Institute, Bangalore, India
| | - Rohini Raghavan
- Division of Molecular Medicine, St. John's Research Institute, Bangalore, India
| | - Srinath B S
- Sri Shankara Cancer Hospital and Research Centre, Bangalore, India
| | - Sridhar T S
- Division of Molecular Medicine, St. John's Research Institute, Bangalore, India
| |
Collapse
|
12
|
High expression of OSR1 as a predictive biomarker for poor prognosis and lymph node metastasis in breast cancer. Breast Cancer Res Treat 2020; 182:35-46. [PMID: 32424721 DOI: 10.1007/s10549-020-05671-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 05/06/2020] [Indexed: 01/22/2023]
Abstract
PURPOSE Oxidative stress-responsive kinase 1 (OSR1) plays a crucial role in regulating diverse cellular pathophysiologic functions, including ion homeostasis, development, differentiation, angiogenesis, invasive migration, and metastasis. Regardless, the clinical significance of OSR1 in breast cancer is scarce. The current study was conducted to evaluate the effect of OSR1 on the prognosis of patients with breast cancer with a long-term follow-up. METHODS OSR1 expression in formalin-fixed and paraffin-embedded tissue specimens was analyzed. These specimens were collected from 551 evaluable breast cancer cases by immunohistochemistry (IHC). OSR1 expression was dichotomized based on the H-score in IHC. The effects of OSR1 levels on the clinicopathological attributes and survival prediction in patients with breast cancer were explored. RESULTS Among 551 specimens, 183 (33.2%) exhibited high expression of OSR1 in tumor cells. High OSR1 levels were markedly correlated with histologic grade (P = 0.035), ER (P < 0.001) and PgR (P = 0.043) expression, lymph node involvement (P < 0.001), TNM stage (P < 0.001), and axillary surgery procedures (P = 0.003). Univariate analysis results indicate that patients with high OSR1 expression had significantly poor overall survival (P < 0.001), distant disease-free survival (P < 0.001), and breast cancer-specific survival (P < 0.001). Multivariable Cox regression analyses suggest that OSR1 expression was an independent predictive marker of poor prognosis and lymph node metastasis (HR 3.224, 95% CI 1.182-8.702, P = 0.023) in patients with breast cancer. CONCLUSIONS Our findings indicate that OSR1 is a significantly independent prognosis index for patients with breast cancer with respect to distant disease-free survival, overall survival, and breast cancer-specific survival. High OSR1 expression caused an increase in deaths specifically attributed to breast cancer and was related to increased lymph node metastasis. However, the precise cellular mechanisms for OSR1 in breast cancer require further research.
Collapse
|
13
|
de la Ballina NR, Villalba A, Cao A. Differences in proteomic profile between two haemocyte types, granulocytes and hyalinocytes, of the flat oyster Ostrea edulis. FISH & SHELLFISH IMMUNOLOGY 2020; 100:456-466. [PMID: 32205190 DOI: 10.1016/j.fsi.2020.03.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/05/2020] [Accepted: 03/17/2020] [Indexed: 06/10/2023]
Abstract
Haemocytes play a dominant role in shellfish immunity, being considered the main defence effector cells in molluscs. These cells are known to be responsible for many functions, including chemotaxis, cellular recognition, attachment, aggregation, shell repair and nutrient transport and digestion. There are two basic cell types of bivalve haemocytes morphologically distinguishable, hyalinocytes and granulocytes; however, functional differences and specific abilities are poorly understood: granulocytes are believed to be more efficient in killing microorganisms, while hyalinocytes are thought to be more specialised in clotting and wound healing. A proteomic approach was implemented to find qualitative differences in the protein profile between granulocytes and hyalinocytes of the European flat oyster, Ostrea edulis, as a way to evaluate functional differences. Oyster haemolymph cells were differentially separated by Percoll® density gradient centrifugation. Granulocyte and hyalinocyte proteins were separated by 2D-PAGE and their protein profiles were analysed and compared with PD Quest software; the protein spots exclusive for each haemocyte type were excised from gels and analysed by MALDI-TOF/TOF with a combination of mass spectrometry (MS) and MS/MS for sequencing and protein identification. A total of 34 proteins were identified, 20 unique to granulocytes and 14 to hyalinocytes. The results suggested differences between the haemocyte types in signal transduction, apoptosis, oxidation reduction processes, cytoskeleton, phagocytosis and pathogen recognition. These results contribute to identify differential roles of each haemocyte type and to better understand the oyster immunity mechanisms, which should help to fight oyster diseases.
Collapse
Affiliation(s)
- Nuria R de la Ballina
- Centro de Investigacións Mariñas (CIMA), Consellería do Mar, Xunta de Galicia, 36620, Vilanova de Arousa, Spain
| | - Antonio Villalba
- Centro de Investigacións Mariñas (CIMA), Consellería do Mar, Xunta de Galicia, 36620, Vilanova de Arousa, Spain; Departamento de Ciencias de la Vida, Universidad de Alcalá, 28871, Alcalá de Henares, Spain; Research Centre for Experimental Marine Biology and Biotechnology (PIE), University of the Basque Country (UPV/EHU), 48620, Plentzia, Spain.
| | - Asunción Cao
- Centro de Investigacións Mariñas (CIMA), Consellería do Mar, Xunta de Galicia, 36620, Vilanova de Arousa, Spain
| |
Collapse
|
14
|
Brown TL, Macklin WB. The Actin Cytoskeleton in Myelinating Cells. Neurochem Res 2020; 45:684-693. [PMID: 30847860 PMCID: PMC6732044 DOI: 10.1007/s11064-019-02753-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/11/2019] [Accepted: 02/12/2019] [Indexed: 12/15/2022]
Abstract
Myelinating cells of both the peripheral and central nervous systems (CNSs) undergo dramatic cytoskeletal reorganization in order to differentiate and produce myelin. Myelinating oligodendrocytes in the CNS show a periodic actin pattern, demonstrating tight regulation of actin. Furthermore, recent data demonstrate that actin polymerization drives early cell differentiation and that actin depolymerization drives myelin wrapping. Dysregulation of the actin cytoskeleton in myelinating cells is seen in some disease states. This review highlights the cytoskeletal molecules that regulate differentiation of and myelination by cells of the PNS and CNS, informing our understanding of neural development, in particular myelination.
Collapse
Affiliation(s)
- Tanya L Brown
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Cell Biology, Stem Cells, and Development Graduate Program, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
15
|
Alauddin M, Okumura T, Rajaxavier J, Khozooei S, Pöschel S, Takeda S, Singh Y, Brucker SY, Wallwiener D, Koch A, Salker MS. Gut Bacterial Metabolite Urolithin A Decreases Actin Polymerization and Migration in Cancer Cells. Mol Nutr Food Res 2020; 64:e1900390. [PMID: 31976617 DOI: 10.1002/mnfr.201900390] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 12/11/2019] [Indexed: 12/12/2022]
Abstract
SCOPE Urolithin A (UA) is a gut-derived bacterial metabolite from ellagic acid found in pomegranates, berries, and nuts can downregulate cell proliferation and migration. Cell proliferation and cell motility require actin reorganization, which is under control of ras-related C3 botulinum toxin substrate 1 (Rac1) and p21 protein-activated kinase 1 (PAK1). The present study explores whether UA can modify actin cytoskeleton in cancer cells. METHODS The effect of UA on globular over filamentous actin ratio is determined utilizing Western blotting, immunofluorescence, and flow cytometry. Rac1 and PAK1 levels are measured by quantitative RT-PCR and immunoblotting. As a result, a 24 h treatment with UA (20 µm) significantly decreased Rac1 and PAK1 transcript levels and activity, depolymerized actin and wound healing. The effect of UA on actin polymerization is mimicked by pharmacological inhibition of Rac1 and PAK1. The effect is also mirrored by knock down using siRNA. CONCLUSION UA leads to disruption of Rac1 and Pak1 activity with subsequent actin depolymerization and migration. Thus, use of dietary UA in cancer prevention or as adjuvant therapy is promising.
Collapse
Affiliation(s)
- Md Alauddin
- Department of Women's Health, Eberhard Karls University of Tuebingen, Tübingen, 72076, Germany
| | - Toshiyuki Okumura
- Department of Women's Health, Eberhard Karls University of Tuebingen, Tübingen, 72076, Germany.,Department of Obstetrics and Gynecology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
| | - Janet Rajaxavier
- Department of Women's Health, Eberhard Karls University of Tuebingen, Tübingen, 72076, Germany
| | - Shayan Khozooei
- Department of Women's Health, Eberhard Karls University of Tuebingen, Tübingen, 72076, Germany
| | - Simone Pöschel
- Image Stream Core Facility, Eberhard Karls University of Tuebingen, Tübingen, 72076, Germany
| | - Satoru Takeda
- Department of Obstetrics and Gynecology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
| | - Yogesh Singh
- Institute of Medical Genetics and Applied Genomics, Eberhard Karls University of Tuebingen, Tübingen, 72076, Germany
| | - Sara Y Brucker
- Department of Women's Health, Eberhard Karls University of Tuebingen, Tübingen, 72076, Germany
| | - Diethelm Wallwiener
- Department of Women's Health, Eberhard Karls University of Tuebingen, Tübingen, 72076, Germany
| | - André Koch
- Department of Women's Health, Eberhard Karls University of Tuebingen, Tübingen, 72076, Germany
| | - Madhuri S Salker
- Department of Women's Health, Eberhard Karls University of Tuebingen, Tübingen, 72076, Germany
| |
Collapse
|
16
|
Sabadashka M, Nagalievska M, Sybirna N. Tyrosine nitration as a key event of signal transduction that regulates functional state of the cell. Cell Biol Int 2020; 45:481-497. [PMID: 31908104 DOI: 10.1002/cbin.11301] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/04/2020] [Indexed: 12/21/2022]
Abstract
This review is dedicated to the role of nitration of proteins by tyrosine residues in physiological and pathological conditions. First of all, we analyze the biochemical evidence of peroxynitrite formation and reactions that lead to its formation, types of posttranslational modifications (PTMs) induced by reactive nitrogen species, as well as three biological pathways of tyrosine nitration. Then, we describe two possible mechanisms of protein nitration that are involved in intracellular signal transduction, as well as its interconnection with phosphorylation/dephosphorylation of tyrosine. Next part of the review is dedicated to the role of proteins nitration in different pathological conditions. In this section, special attention is devoted to the role of nitration in changes of functional properties of actin-protein that undergoes PTMs both in normal and pathological conditions. Overall, this review is devoted to the main features of protein nitration by tyrosine residue and the role of this process in intracellular signal transduction in basal and pathological conditions.
Collapse
Affiliation(s)
- Mariya Sabadashka
- Department of Biochemistry, Faculty of Biology, Ivan Franko National University of Lviv, 4, Hrushevskyi St., Lviv, 79005, Ukraine
| | - Mariia Nagalievska
- Department of Biochemistry, Faculty of Biology, Ivan Franko National University of Lviv, 4, Hrushevskyi St., Lviv, 79005, Ukraine
| | - Nataliia Sybirna
- Department of Biochemistry, Faculty of Biology, Ivan Franko National University of Lviv, 4, Hrushevskyi St., Lviv, 79005, Ukraine
| |
Collapse
|
17
|
Zhou Y, Zhang J, Wang J, Cheng M, Zhao D, Li F. Targeting PAK1 with the Small Molecule Drug AK963/40708899 Suppresses Gastric Cancer Cell Proliferation and Invasion by Downregulation of PAK1 Activity and PAK1‐Related Signaling Pathways. Anat Rec (Hoboken) 2019; 302:1571-1579. [DOI: 10.1002/ar.24095] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/06/2018] [Accepted: 11/18/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Ying Zhou
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell BiologyMinistry of Education, China Medical University Shenyang China
| | - Jian Zhang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell BiologyMinistry of Education, China Medical University Shenyang China
| | - Jian Wang
- Key Laboratory of Structure‐Based Drug Design and Discovery of Ministry of EducationShenyang Pharmaceutical University Shenyang China
| | - Mao‐Sheng Cheng
- Key Laboratory of Structure‐Based Drug Design and Discovery of Ministry of EducationShenyang Pharmaceutical University Shenyang China
| | - Dong‐Mei Zhao
- Key Laboratory of Structure‐Based Drug Design and Discovery of Ministry of EducationShenyang Pharmaceutical University Shenyang China
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell BiologyMinistry of Education, China Medical University Shenyang China
| |
Collapse
|
18
|
Lobo V, Parte P. Membrane-bound Glucose regulated protein 78 interacts with alpha-2-macroglobulin to promote actin reorganization in sperm during epididymal maturation. ACTA ACUST UNITED AC 2018; 25:137-155. [DOI: 10.1093/molehr/gay055] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/06/2018] [Accepted: 12/24/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Vivian Lobo
- Department of Gamete Immunobiology, ICMR—National Institute for Research in Reproductive Health, Mumbai, India
| | - Priyanka Parte
- Department of Gamete Immunobiology, ICMR—National Institute for Research in Reproductive Health, Mumbai, India
| |
Collapse
|
19
|
Delve E, Parreno J, Co V, Wu PH, Chong J, Di Scipio M, Kandel RA. CDC42 regulates the expression of superficial zone molecules in part through the actin cytoskeleton and myocardin-related transcription factor-A. J Orthop Res 2018. [PMID: 29537109 DOI: 10.1002/jor.23892] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Osteoarthritis (OA) is a degenerative disease that initially manifests as loss of the superficial zone (SZ) of articular cartilage. SZ chondrocytes (SZC) differ in morphology from other chondrocytes as they are elongated and oriented parallel to the tissue surface. Proteoglycan 4 (PRG4) and tenascin C (TNC) are molecules expressed by SZC, which have been shown to be chondroprotective. Identification of the signalling pathway(s) regulating expression of SZ molecules may lead to a therapeutic target that can be used to delay or prevent the onset of OA. The hypothesis of this study is that expression of SZ molecules are regulated in part, by the CDC42-actin-myocardin-related transcription factor-A (MRTF-A) signaling pathway. SZC from bovine metacarpal-phalangeal joints were isolated and grown in monolayer culture. Each target in the CDC42-actin-MRTF-A pathway was inhibited and the effect on cell shape, actin cytoskeleton status, and expression of PRG4 and TNC were determined. Treatment with the CDC42 inhibitor ML141 decreased PRG4 and TNC expression, and correlated with increased cell circularity and G-/F-actin ratio. PRG4 and TNC expression were differentially regulated by actin depolymerizing agents, latrunculin B and cytochalasin D. Chemical inhibition of MRTF-A resulted in decreased expression of both PRG4 and TNC; however, specific knockdown by small interfering RNA only decreased expression of TNC indicating that TNC, but not PRG4, is regulated by MRTF-A. Although PRG4 and TNC expression are both regulated by CDC42 and actin, it appears to occur through different downstream signaling pathways. Further study is required to elucidate the pathway regulating PRG4. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:2421-2430, 2018.
Collapse
Affiliation(s)
- Elizabeth Delve
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario
| | - Justin Parreno
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Vivian Co
- University of Ontario Institute of Technology, Oshawa, Ontario
| | - Po-Han Wu
- The Department of Human Biology, University of Toronto, Toronto, Ontario
| | - Jasmine Chong
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario
| | - Matteo Di Scipio
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario
| | - Rita A Kandel
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario.,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario.,Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario
| |
Collapse
|
20
|
14-3-3 Regulates Actin Filament Formation in the Deep-Branching Eukaryote Giardia lamblia. mSphere 2017; 2:mSphere00248-17. [PMID: 28932813 PMCID: PMC5597967 DOI: 10.1128/msphere.00248-17] [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] [Received: 05/26/2017] [Accepted: 08/21/2017] [Indexed: 01/30/2023] Open
Abstract
The phosphoserine/phosphothreonine-binding protein 14-3-3 is known to regulate actin; this function has been previously attributed to sequestration of phosphorylated cofilin. 14-3-3 was identified as an actin-associated protein in the deep-branching eukaryote Giardia lamblia; however, Giardia lacks cofilin and all other canonical actin-binding proteins (ABPs). Thus, the role of G. lamblia 14-3-3 (Gl-14-3-3) in actin regulation was unknown. Gl-14-3-3 depletion resulted in an overall disruption of actin organization characterized by ectopically distributed short actin filaments. Using phosphatase and kinase inhibitors, we demonstrated that actin phosphorylation correlated with destabilization of the actin network and increased complex formation with 14-3-3, while blocking actin phosphorylation stabilized actin filaments and attenuated complex formation. Giardia's sole Rho family GTPase, Gl-Rac, modulates Gl-14-3-3's association with actin, providing the first connection between Gl-Rac and the actin cytoskeleton in Giardia. Giardia actin (Gl-actin) contains two putative 14-3-3 binding motifs, one of which (S330) is conserved in mammalian actin. Mutation of these sites reduced, but did not completely disrupt, the association with 14-3-3. Native gels and overlay assays indicate that intermediate proteins are required to support complex formation between 14-3-3 and actin. Overall, our results support a role for 14-3-3 as a regulator of actin; however, the presence of multiple 14-3-3-actin complexes suggests a more complex regulatory relationship than might be expected for a minimalistic parasite. IMPORTANCEGiardia lacks canonical actin-binding proteins. Gl-14-3-3 was identified as an actin interactor, but the significance of this interaction was unknown. Loss of Gl-14-3-3 results in ectopic short actin filaments, indicating that Gl-14-3-3 is an important regulator of the actin cytoskeleton in Giardia. Drug studies indicate that Gl-14-3-3 complex formation is in part phospho-regulated. We demonstrate that complex formation is downstream of Giardia's sole Rho family GTPase, Gl-Rac. This result provides the first mechanistic connection between Gl-Rac and Gl-actin in Giardia. Native gels and overlay assays indicate intermediate proteins are required to support the interaction between Gl-14-3-3 and Gl-actin, suggesting that Gl-14-3-3 is regulating multiple Gl-actin complexes.
Collapse
|
21
|
Lee J, Ahn E, Park WK, Park S. Phosphoproteome Profiling of SH-SY5y Neuroblastoma Cells Treated with Anesthetics: Sevoflurane and Isoflurane Affect the Phosphorylation of Proteins Involved in Cytoskeletal Regulation. PLoS One 2016; 11:e0162214. [PMID: 27611435 PMCID: PMC5017685 DOI: 10.1371/journal.pone.0162214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 08/18/2016] [Indexed: 12/11/2022] Open
Abstract
Inhalation anesthetics are used to decrease the spinal cord transmission of painful stimuli. However, the molecular or biochemical processes within cells that regulate anesthetic-induced responses at the cellular level are largely unknown. Here, we report the phosphoproteome profile of SH-SY5y human neuroblastoma cells treated with sevoflurane, a clinically used anesthetic. Phosphoproteins were isolated from cell lysates and analyzed using two-dimensional gel electrophoresis. The phosphorylation of putative anesthetic-responsive marker proteins was validated using western blot analysis in cells treated with both sevoflurane and isoflurane. A total of 25 phosphoproteins were identified as differentially phosphorylated proteins. These included key regulators that signal cytoskeletal remodeling steps in pathways related to vesicle trafficking, axonal growth, and cell migration. These proteins included the Rho GTPase, Ras-GAP SH3 binding protein, Rho GTPase activating protein, actin-related protein, and actin. Sevoflurane and isoflurane also resulted in the dissolution of F-actin fibers in SH-SY5y cells. Our results show that anesthetics affect the phosphorylation of proteins involved in cytoskeletal remodeling pathways.
Collapse
Affiliation(s)
- Joomin Lee
- Department of Food and Nutrition, Chosun University, Gwangju 61452, Korea
| | - Eunsook Ahn
- Department of Applied Chemistry, Dongduk Women’s University, Seoul 02748, Korea
| | - Wyun Kon Park
- Department of Anesthesia and Pain, College of Medicine, Department of Anesthesia and Pain, Yonsei University, Seoul 03722, Korea
| | - Seyeon Park
- Department of Applied Chemistry, Dongduk Women’s University, Seoul 02748, Korea
- * E-mail:
| |
Collapse
|
22
|
LeftyA decreases Actin Polymerization and Stiffness in Human Endometrial Cancer Cells. Sci Rep 2016; 6:29370. [PMID: 27404958 PMCID: PMC4941646 DOI: 10.1038/srep29370] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 06/16/2016] [Indexed: 12/27/2022] Open
Abstract
LeftyA, a cytokine regulating stemness and embryonic differentiation, down-regulates cell proliferation and migration. Cell proliferation and motility require actin reorganization, which is under control of ras-related C3 botulinum toxin substrate 1 (Rac1) and p21 protein-activated kinase 1 (PAK1). The present study explored whether LeftyA modifies actin cytoskeleton, shape and stiffness of Ishikawa cells, a well differentiated endometrial carcinoma cell line. The effect of LeftyA on globular over filamentous actin ratio was determined utilizing Western blotting and flow cytometry. Rac1 and PAK1 transcript levels were measured by qRT-PCR as well as active Rac1 and PAK1 by immunoblotting. Cell stiffness (quantified by the elastic modulus), cell surface area and cell volume were studied by atomic force microscopy (AFM). As a result, 2 hours treatment with LeftyA (25 ng/ml) significantly decreased Rac1 and PAK1 transcript levels and activity, depolymerized actin, and decreased cell stiffness, surface area and volume. The effect of LeftyA on actin polymerization was mimicked by pharmacological inhibition of Rac1 and PAK1. In the presence of the Rac1 or PAK1 inhibitor LeftyA did not lead to significant further actin depolymerization. In conclusion, LeftyA leads to disruption of Rac1 and Pak1 activity with subsequent actin depolymerization, cell softening and cell shrinkage.
Collapse
|
23
|
Fajol A, Honisch S, Zhang B, Schmidt S, Alkahtani S, Alarifi S, Lang F, Stournaras C, Föller M. Fibroblast growth factor (Fgf) 23 gene transcription depends on actin cytoskeleton reorganization. FEBS Lett 2016; 590:705-15. [DOI: 10.1002/1873-3468.12096] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 01/27/2016] [Accepted: 02/11/2016] [Indexed: 11/12/2022]
Affiliation(s)
- Abul Fajol
- Department of Physiology; University of Tübingen; Germany
| | - Sabina Honisch
- Department of Physiology; University of Tübingen; Germany
| | - Bingbing Zhang
- Department of Physiology; University of Tübingen; Germany
| | | | - Saad Alkahtani
- Department of Zoology; Science College; King Saud University; Riyadh Saudi Arabia
- Department of Biochemistry; University of Crete Medical School; Heraklion Greece
| | - Saud Alarifi
- Department of Zoology; Science College; King Saud University; Riyadh Saudi Arabia
- Department of Biochemistry; University of Crete Medical School; Heraklion Greece
| | - Florian Lang
- Department of Physiology; University of Tübingen; Germany
| | - Christos Stournaras
- Department of Physiology; University of Tübingen; Germany
- Department of Biochemistry; University of Crete Medical School; Heraklion Greece
| | - Michael Föller
- Institute of Agricultural and Nutritional Sciences; Martin-Luther University Halle-Wittenberg; Halle (Saale) Germany
| |
Collapse
|
24
|
Hernandez K, Swiatkowski P, Patel MV, Liang C, Dudzinski NR, Brzustowicz LM, Firestein BL. Overexpression of Isoforms of Nitric Oxide Synthase 1 Adaptor Protein, Encoded by a Risk Gene for Schizophrenia, Alters Actin Dynamics and Synaptic Function. Front Cell Neurosci 2016; 10:6. [PMID: 26869880 PMCID: PMC4735351 DOI: 10.3389/fncel.2016.00006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 01/11/2016] [Indexed: 11/13/2022] Open
Abstract
Proper communication between neurons depends upon appropriate patterning of dendrites and correct distribution and structure of spines. Schizophrenia is a neuropsychiatric disorder characterized by alterations in dendrite branching and spine density. Nitric oxide synthase 1 adaptor protein (NOS1AP), a risk gene for schizophrenia, encodes proteins that are upregulated in the dorsolateral prefrontal cortex (DLPFC) of individuals with schizophrenia. To elucidate the effects of NOS1AP overexpression observed in individuals with schizophrenia, we investigated changes in actin dynamics and spine development when a long (NOS1AP-L) or short (NOS1AP-S) isoform of NOS1AP is overexpressed. Increased NOS1AP-L protein promotes the formation of immature spines when overexpressed in rat cortical neurons from day in vitro (DIV) 14 to DIV 17 and reduces the amplitude of miniature excitatory postsynaptic currents (mEPSCs). In contrast, increased NOS1AP-S protein increases the rate of actin polymerization and the number of immature and mature spines, which may be attributed to a decrease in total Rac1 expression and a reduction in the levels of active cofilin. The increase in the number of mature spines by overexpression of NOS1AP-S is accompanied by an increase in the frequency of mEPSCs. Our findings show that overexpression of NOS1AP-L or NOS1AP-S alters the actin cytoskeleton and synaptic function. However, the mechanisms by which these isoforms induce these changes are distinct. These results are important for understanding how increased expression of NOS1AP isoforms can influence spine development and synaptic function.
Collapse
Affiliation(s)
- Kristina Hernandez
- Department of Cell Biology and Neuroscience, Human Genetics Institute of New Jersey, Rutgers—The State University of New JerseyPiscataway, NJ, USA
| | - Przemyslaw Swiatkowski
- Department of Cell Biology and Neuroscience, Rutgers—The State University of New JerseyPiscataway, NJ, USA
| | - Mihir V. Patel
- Department of Cell Biology and Neuroscience, Rutgers—The State University of New JerseyPiscataway, NJ, USA
| | - Chen Liang
- Department of Cell Biology and Neuroscience, Rutgers—The State University of New JerseyPiscataway, NJ, USA
| | - Natasha R. Dudzinski
- Department of Cell Biology and Neuroscience, Rutgers—The State University of New JerseyPiscataway, NJ, USA
| | - Linda M. Brzustowicz
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers—The State University of New JerseyPiscataway, NJ, USA
| | - Bonnie L. Firestein
- Department of Cell Biology and Neuroscience, Human Genetics Institute of New Jersey, Rutgers—The State University of New JerseyPiscataway, NJ, USA
| |
Collapse
|
25
|
Kim YB, Shin YJ, Roy A, Kim JH. The Role of the Pleckstrin Homology Domain-containing Protein CKIP-1 in Activation of p21-activated Kinase 1 (PAK1). J Biol Chem 2015; 290:21076-21085. [PMID: 26160174 DOI: 10.1074/jbc.m115.675124] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Indexed: 11/06/2022] Open
Abstract
Upon growth factor stimulation, PAK1 is recruited to the plasma membrane and activated by a mechanism that requires its phosphorylation at Ser-223 by the protein kinase CK2. However, the upstream signaling molecules that regulate this phosphorylation event are not clearly defined. Here, we demonstrate a major role of the CK2α-interacting protein CKIP-1 in activation of PAK1. CK2α, CKIP-1, and PAK1 are translocated to membrane ruffles in response to the epidermal growth factor (EGF), where CKIP-1 mediates the interaction between CK2α and PAK1 in a PI3K-dependent manner. Consistently, PAK1 mediates phosphorylation and modulation of the activity of p41-Arc, one of its plasma membrane substrate, in a fashion that requires PI3K and CKIP-1. Moreover, CKIP-1 knockdown or PI3K inhibition suppresses PAK1-mediated cell migration and invasion, demonstrating the physiological significance of the PI3K-CKIP-1-CK2-PAK1 signaling pathway. Taken together, these findings identify a novel mechanism for the activation of PAK1 at the plasma membrane, which is critical for cell migration and invasion.
Collapse
Affiliation(s)
- Yong-Bae Kim
- Department of Biochemistry and Molecular Medicine, The George Washington University Medical Center, Washington, DC 20037 and
| | - Yong Jae Shin
- Samsung Biomedical Research Institute and Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
| | - Adhiraj Roy
- Department of Biochemistry and Molecular Medicine, The George Washington University Medical Center, Washington, DC 20037 and
| | - Jeong-Ho Kim
- Department of Biochemistry and Molecular Medicine, The George Washington University Medical Center, Washington, DC 20037 and.
| |
Collapse
|
26
|
PAK1 regulates RUFY3-mediated gastric cancer cell migration and invasion. Cell Death Dis 2015; 6:e1682. [PMID: 25766321 PMCID: PMC4385928 DOI: 10.1038/cddis.2015.50] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 01/21/2015] [Accepted: 01/28/2015] [Indexed: 01/01/2023]
Abstract
Actin protrusion at the cell periphery is central to the formation of invadopodia during tumor cell migration and invasion. Although RUFY3 (RUN and FYVE domain containing 3)/SINGAR1 (single axon-related1)/RIPX (Rap2 interacting protein X) has an important role in neuronal development, its pathophysiologic role and relevance to cancer are still largely unknown. The purpose of this study was to elucidate the molecular mechanisms by which RUFY3 involves in gastric cancer cell migration and invasion. Here, our data show that overexpression of RUFY3 leads to the formation of F-actin-enriched protrusive structures at the cell periphery and induces gastric cancer cell migration. Furthermore, P21-activated kinase-1 (PAK1) interacts with RUFY3, and promotes RUFY3 expression and RUFY3-induced gastric cancer cell migration; inhibition of PAK1 attenuates RUFY3-induced SGC-7901 cell migration and invasion. Importantly, we found that the inhibitory effect of cell migration and invasion is significantly enhanced by knockdown of both PAK1 and RUFY3 compared with knockdown of RUFY3 alone or PAK1 alone. Strikingly, we found significant upregulation of RUFY3 in gastric cancer samples with invasive carcinoma at pathologic TNM III and TNM IV stages, compared with their non-tumor counterparts. Moreover, an obvious positive correlation was observed between the protein expression of RUFY3 and PAK1 in 40 pairs of gastric cancer samples. Therefore, these findings provide important evidence that PAK1 can positively regulate RUFY3 expression, which contribute to the metastatic potential of gastric cancer cells, maybe blocking PAK1-RUFY3 signaling would become a potential metastasis therapeutic strategy for gastric cancer.
Collapse
|
27
|
Gq-mediated Akt translocation to the membrane: a novel PIP3-independent mechanism in platelets. Blood 2014; 125:175-84. [PMID: 25331114 DOI: 10.1182/blood-2014-05-576306] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Akt is an important signaling molecule regulating platelet aggregation. Akt is phosphorylated after translocation to the membrane through Gi signaling pathways by a phosphatidylinositol-3,4,5-trisphosphate (PIP3)-dependent mechanism. However, Akt is more robustly phosphorylated by thrombin compared with adenosine 5'-diphosphate in platelets. This study investigated the mechanisms of Akt translocation as a possible explanation for this difference. Stimulation of washed human platelets with protease-activated receptor agonists caused translocation of Akt to the membrane rapidly, whereas phosphorylation occurred later. The translocation of Akt was abolished in the presence of a Gq-selective inhibitor or in Gq-deficient murine platelets, indicating that Akt translocation is regulated downstream of Gq pathways. Interestingly, phosphatidylinositol 3-kinase (PI3K) inhibitors or P2Y12 antagonist abolished Akt phosphorylation without affecting Akt translocation to the membrane, suggesting that Akt translocation occurs through a PI3K/PIP3/Gi-independent mechanism. An Akt scaffolding protein, p21-activated kinase (PAK), translocates to the membrane after stimulation with protease-activated receptor agonists in a Gq-dependent manner, with the kinetics of translocation similar to that of Akt. Coimmunoprecipitation studies showed constitutive association of PAK and Akt, suggesting a possible role of PAK in Akt translocation. These results show, for the first time, an important role of the Gq pathway in mediating Akt translocation to the membrane in a novel Gi/PI3K/PIP3-independent mechanism.
Collapse
|
28
|
Parreno J, Raju S, Niaki MN, Andrejevic K, Jiang A, Delve E, Kandel R. Expression of type I collagen and tenascin C is regulated by actin polymerization through MRTF in dedifferentiated chondrocytes. FEBS Lett 2014; 588:3677-84. [PMID: 25150168 DOI: 10.1016/j.febslet.2014.08.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 08/05/2014] [Accepted: 08/11/2014] [Indexed: 12/12/2022]
Abstract
This study examined actin regulation of fibroblast matrix genes in dedifferentiated chondrocytes. We demonstrated that dedifferentiated chondrocytes exhibit increased actin polymerization, nuclear localization of myocardin related transcription factor (MRTF), increased type I collagen (col1) and tenascin C (Tnc) gene expression, and decreased Sox9 gene expression. Induction of actin depolymerization by latrunculin treatment or cell rounding, reduced MRTF nuclear localization, repressed col1 and Tnc expression, and increased Sox9 gene expression in dedifferentiated chondrocytes. Treatment of passaged chondrocytes with MRTF inhibitor repressed col1 and Tnc expression, but did not affect Sox9 expression. Our results show that actin polymerization regulates fibroblast matrix gene expression through MRTF in passaged chondrocytes.
Collapse
Affiliation(s)
- Justin Parreno
- CIHR-BioEngineering of Skeletal Tissues Team, Mount Sinai Hospital, University of Toronto, Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada; Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
| | - Sneha Raju
- CIHR-BioEngineering of Skeletal Tissues Team, Mount Sinai Hospital, University of Toronto, Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Mortah Nabavi Niaki
- CIHR-BioEngineering of Skeletal Tissues Team, Mount Sinai Hospital, University of Toronto, Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Canada
| | - Katarina Andrejevic
- CIHR-BioEngineering of Skeletal Tissues Team, Mount Sinai Hospital, University of Toronto, Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada; Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
| | - Amy Jiang
- CIHR-BioEngineering of Skeletal Tissues Team, Mount Sinai Hospital, University of Toronto, Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Elizabeth Delve
- CIHR-BioEngineering of Skeletal Tissues Team, Mount Sinai Hospital, University of Toronto, Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Canada
| | - Rita Kandel
- CIHR-BioEngineering of Skeletal Tissues Team, Mount Sinai Hospital, University of Toronto, Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada; Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Canada.
| |
Collapse
|
29
|
Huynh N, Liu KH, Yim M, Shulkes A, Baldwin GS, He H. Demonstration and biological significance of a gastrin-P21-activated kinase 1 feedback loop in colorectal cancer cells. Physiol Rep 2014; 2:2/6/e12048. [PMID: 24963032 PMCID: PMC4208650 DOI: 10.14814/phy2.12048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gastrins, including amidated gastrin17 and glycine-extended gastrin17, are important growth factors in colorectal cancer (CRC). The p21-activated kinase 1 (PAK1) plays key roles in cellular processes including proliferation, survival, and motility, and in cell transformation and tumor progression. PAK1 expression increases with the progression of CRC, and knockdown of PAK1 blocks CRC cell growth and metastasis both in vitro and in vivo. The aim of this study was to determine the interaction between PAK1 and gastrins in CRC cells. PAK1 expression and activation were assayed by Western blots, and concentrations of gastrin mRNA and peptides by real-time PCR and radioimmunoassay, respectively. Proliferation of CRC cells was measured by (3)H-thymidine incorporation, and vascular endothelial growth factor : VEGF) secretion was measured by ELISA. Gastrins activated PAK1 via PI3K-dependent pathways. Activated PAK1 in turn mediated gastrin-stimulated activation of β-catenin and VEGF secretion in CRC cells, as knockdown of PAK1 blocked stimulation of these cellular processes by gastrins. Downregulation of gastrin reduced the expression and activity of PAK1, but in contrast there was a compensatory increase in gastrins either when PAK1 was downregulated, or after treatment with a PAK inhibitor. Our results indicate that PAK1 is required for the stimulation of CRC cells by gastrins, and suggest the existence of an inhibitory feedback loop by which PAK1 downregulates gastrin production in CRC cells.
Collapse
Affiliation(s)
- Nhi Huynh
- Department of Surgery, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | - Kevin H Liu
- Department of Surgery, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | - Mildred Yim
- Department of Surgery, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | - Arthur Shulkes
- Department of Surgery, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | - Graham S Baldwin
- Department of Surgery, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | - Hong He
- Department of Surgery, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| |
Collapse
|
30
|
Kou B, Gao Y, Du C, Shi Q, Xu S, Wang CQ, Wang X, He D, Guo P. miR-145 inhibits invasion of bladder cancer cells by targeting PAK1. Urol Oncol 2014; 32:846-54. [PMID: 24954107 DOI: 10.1016/j.urolonc.2014.01.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 01/02/2014] [Accepted: 01/02/2014] [Indexed: 11/26/2022]
Abstract
OBJECTIVES MicroRNAs play important roles in cancer. In many cancers, miR-145 acts as a tumor suppressor, and it is down-regulated in bladder cancer. In the present study, we explored the modulation of oncogenic gene PAK1 by miR-145 in bladder cancer. MATERIAL AND METHODS Expression of miR-145 was detected in bladder cancer tissues and cell lines by quantitative real-time polymerase chain reaction. Through the bioinformatics approach, PAK1 has been predicted to be a direct target of miR-145 and was confirmed by the PAK1 messenger RNA 3'-untranslated region luciferase activity assay. To investigate whether miR-145 regulates PAK1 expression, it was overexpressed in J82 and T24 bladder cancer cells. In 10 paired bladder normal and tumor tissues, we determined the relationship between miR-145 and PAK1 through quantitative real-time polymerase chain reaction and western blot. By using transwell invasion assay and western blotting analysis, we investigated the effects of miR-145 and PAK1 on bladder cancer cell invasion and expression of invasion marker genes. RESULTS The level of miR-145 decreases and PAK1 protein expression up-regulates in bladder cancer tissue, as compared with the paired normal bladder tissue. Moreover, miR-145 directly targets PAK1 in bladder cancer cells. The level of miR-145 negatively correlates with PAK1 protein expression in bladder cancer. In addition, PAK1 promotes invasion and enhances the expression and activity of MMP-9, whereas miR-145 inhibits bladder cancer cell invasion and expressions of PAK1 and MMP-9. CONCLUSIONS Our results indicate that miR-145 inhibits bladder cancer cell invasion, at least partly through targeting PAK1. Restoration or replacement of miR-145 could be an efficient approach to inhibit PAK1 and bladder cancer development in the tumor therapy.
Collapse
Affiliation(s)
- Bo Kou
- Department of Urology, The First Hospital of Xi׳an Jiaotong University, Xi׳an, Shaanxi, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi׳an, Shaanxi, P.R. China
| | - Yang Gao
- Department of Urology, The First Hospital of Xi׳an Jiaotong University, Xi׳an, Shaanxi, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi׳an, Shaanxi, P.R. China
| | - Chong Du
- Department of Urology, The First Hospital of Xi׳an Jiaotong University, Xi׳an, Shaanxi, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi׳an, Shaanxi, P.R. China
| | - Qi Shi
- Department of Urology, The First Hospital of Xi׳an Jiaotong University, Xi׳an, Shaanxi, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi׳an, Shaanxi, P.R. China
| | - Shan Xu
- Department of Urology, The First Hospital of Xi׳an Jiaotong University, Xi׳an, Shaanxi, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi׳an, Shaanxi, P.R. China
| | - Chen-Qing Wang
- Department of Urology, The First Hospital of Xi׳an Jiaotong University, Xi׳an, Shaanxi, P.R. China
| | - Xinyang Wang
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi׳an, Shaanxi, P.R. China
| | - Dalin He
- Department of Urology, The First Hospital of Xi׳an Jiaotong University, Xi׳an, Shaanxi, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi׳an, Shaanxi, P.R. China
| | - Peng Guo
- Department of Urology, The First Hospital of Xi׳an Jiaotong University, Xi׳an, Shaanxi, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi׳an, Shaanxi, P.R. China.
| |
Collapse
|
31
|
Pink M, Verma N, Rettenmeier AW, Schmitz-Spanke S. Integrated proteomic and metabolomic analysis to assess the effects of pure and benzo[a]pyrene-loaded carbon black particles on energy metabolism and motility in the human endothelial cell line EA.hy926. Arch Toxicol 2014; 88:913-34. [DOI: 10.1007/s00204-014-1200-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 01/14/2014] [Indexed: 12/20/2022]
|
32
|
Roy R, Parashar V, Chauhan LKS, Shanker R, Das M, Tripathi A, Dwivedi PD. Mechanism of uptake of ZnO nanoparticles and inflammatory responses in macrophages require PI3K mediated MAPKs signaling. Toxicol In Vitro 2013; 28:457-67. [PMID: 24368203 DOI: 10.1016/j.tiv.2013.12.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 11/13/2013] [Accepted: 12/11/2013] [Indexed: 11/30/2022]
Abstract
The inflammatory responses after exposure to zinc oxide nanoparticles (ZNPs) are known, however, the molecular mechanisms and direct consequences of particle uptake are still unclear. Dose and time-dependent increase in the uptake of ZNPs by macrophages has been observed by flow cytometry. Macrophages treated with ZNPs showed a significantly enhanced phagocytic activity. Inhibition of different internalization receptors caused a reduction in uptake of ZNPs in macrophages. The strongest inhibition in internalization was observed by blocking clathrin, caveolae and scavenger receptor mediated endocytic pathways. However, FcR and complement receptor-mediated phagocytic pathways also contributed significantly to control. Further, exposure of primary macrophages to ZNPs (2.5 μg/ml) caused (i) significant enhancement of Ras, PI3K, (ii) enhanced phosphorylation and subsequent activation of its downstream signaling pathways via ERK1/2, p38 and JNK MAPKs (iii) overexpression of c-Jun, c-Fos and NF-κB. Our results demonstrate that ZNPs induce the generation of reactive nitrogen species and overexpression of Cox-2, iNOS, pro-inflammatory cytokines (IL-6, IFN-γ, TNF-α, IL-17 and regulatory cytokine IL-10) and MAPKs which were found to be inhibited after blocking internalization of ZNPs through caveolae receptor pathway. These results indicate that ZNPs are internalized through caveolae pathway and the inflammatory responses involve PI3K mediated MAPKs signaling cascade.
Collapse
Affiliation(s)
- Ruchi Roy
- Food, Drug and Chemical Toxicology Group, Indian Institute of Toxicology Research, M.G. Marg, Post Box No. 80, Lucknow 226001, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Vyom Parashar
- Nanotechnology Application Centre, Faculty of Science, University of Allahabad, Allahabad 211002, India
| | - L K S Chauhan
- Electron Microscopy Facility, CSIR-Indian Institute of Toxicology Research, M.G. Marg, Post Box No. 80, Lucknow 226001, India
| | - Rishi Shanker
- Food, Drug and Chemical Toxicology Group, Indian Institute of Toxicology Research, M.G. Marg, Post Box No. 80, Lucknow 226001, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India; Environmental Toxicology Division, CSIR-Indian Institute of Toxicology Research, M.G. Marg, Post Box No. 80, Lucknow 226001, India
| | - Mukul Das
- Food, Drug and Chemical Toxicology Group, Indian Institute of Toxicology Research, M.G. Marg, Post Box No. 80, Lucknow 226001, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Anurag Tripathi
- Food, Drug and Chemical Toxicology Group, Indian Institute of Toxicology Research, M.G. Marg, Post Box No. 80, Lucknow 226001, India.
| | - Premendra Dhar Dwivedi
- Food, Drug and Chemical Toxicology Group, Indian Institute of Toxicology Research, M.G. Marg, Post Box No. 80, Lucknow 226001, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India.
| |
Collapse
|
33
|
Axl gene knockdown inhibits the metastasis properties of hepatocellular carcinoma via PI3K/Akt-PAK1 signal pathway. Tumour Biol 2013; 35:3809-17. [PMID: 24347489 DOI: 10.1007/s13277-013-1521-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 11/29/2013] [Indexed: 01/08/2023] Open
Abstract
The objective of this study is to clarify the possible role and mechanism of Axl in the tumorigenicity and metastasis process of hepatocellular carcinoma. The mRNA and protein expression levels of Axl in MHCC97-H and MHCC97-L cell lines were evaluated by real-time PCR and Western blot analysis. The key factor of phosphatidylinositol-3-kinase (PI3K)/Akt-p21-activated kinases-1 (PAK1) signaling pathway was studied after Axl expression was downregulated by shRNA. Finally, we analyzed the expression status of Axl protein expression in hepatocellular carcinoma tissues and its relationship with the prognosis of hepatocellular carcinoma. Axl was observed to be higher expressed in MHCC97-H cell lines compared to MHCC97-L cell lines. The downregulation of Axl in MHCC97-H cell lines resulted in the inhibition of the invasion ability of MHCC97-H cells both in vitro and in vivo. Interestingly, blocking PI3K/Akt signaling pathway by LY294002 or Akt siRNA could remarkably inhibit the PAK1 activation and cell invasion. Finally, the Axl protein expression was positively correlated with differentiation, lymph node metastasis, and clinical stage in patients with hepatocellular carcinoma patients (all P < 0.01). These findings suggest that Axl can also regulate the metastasis process of hepatocellular carcinoma and may serve as a new prognostic marker and therapeutic target for treating hepatocellular carcinoma metastasis.
Collapse
|
34
|
Wang H, Zhuang X, Cai Y, Cheung AY, Jiang L. Apical F-actin-regulated exocytic targeting of NtPPME1 is essential for construction and rigidity of the pollen tube cell wall. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2013; 76:367-79. [PMID: 23906068 DOI: 10.1111/tpj.12300] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 07/15/2013] [Accepted: 07/18/2013] [Indexed: 05/03/2023]
Abstract
In tip-confined growing pollen tubes, delivery of newly synthesized cell wall materials to the rapidly expanding apical surface requires spatial organization and temporal regulation of the apical F-actin filament and exocytosis. In this study, we demonstrate that apical F-actin is essential for the rigidity and construction of the pollen tube cell wall by regulating exocytosis of Nicotiana tabacum pectin methylesterase (NtPPME1). Wortmannin disrupts the spatial organization of apical F-actin in the pollen tube tip and inhibits polar targeting of NtPPME1, which subsequently alters the rigidity and pectic composition of the pollen tube cell wall, finally causing growth arrest of the pollen tube. In addition to mechanistically linking cell wall construction and apical F-actin, wortmannin can be used as a useful tool for studying endomembrane trafficking and cytoskeletal organization in pollen tubes.
Collapse
Affiliation(s)
- Hao Wang
- School of Life Sciences, Centre for Cell and Developmental Biology and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | | | | | | | | |
Collapse
|
35
|
Yang Y, Du J, Hu Z, Liu J, Tian Y, Zhu Y, Wang L, Gu L. Activation of Rac1-PI3K/Akt is required for epidermal growth factor-induced PAK1 activation and cell migration in MDA-MB-231 breast cancer cells. J Biomed Res 2013; 25:237-45. [PMID: 23554696 PMCID: PMC3597073 DOI: 10.1016/s1674-8301(11)60032-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 05/16/2011] [Accepted: 06/02/2011] [Indexed: 11/11/2022] Open
Abstract
Epidermal growth factor (EGF) may increase cell motility, an event implicated in cancer cell invasion and metastasis. However, the underlying mechanisms for EGF-induced cell motility remain elusive. In this study, we found that EGF treatment could activate Ras-related C3 botulinum toxin substrate 1 (Rac1), PI3K/Akt and p21-actived kinase (PAK1) along with cell migration. Ectopic expression of PAK1 K299R, a dominant negative PAK1 mutant, could largely abolish EGF-induced cell migration. Blocking PI3K/Akt signalling with LY294002 or Akt siRNA remarkably inhibited both EGF-induced PAK1 activation and cell migration. Furthermore, expression of dominant-negative Rac1 (T17N) could largely block EGF-induced PI3K/Akt-PAK1 activation and cell migration. Interestingly, EGF could induce a significant production of ROS, and N-acetyl-L-cysteine, a scavenger of ROS which abolished the EGF-induced ROS generation, cell migration, as well as activation of PI3K/Akt and PAK, but not Rac1. Our study demonstrated that EGF-induced cell migration involves a cascade of signalling events, including activation of Rac1, generation of ROS and subsequent activation of PI3K/Akt and PAK1.
Collapse
|
36
|
The B55α regulatory subunit of protein phosphatase 2A mediates fibroblast growth factor-induced p107 dephosphorylation and growth arrest in chondrocytes. Mol Cell Biol 2013; 33:2865-78. [PMID: 23716589 DOI: 10.1128/mcb.01730-12] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Fibroblast growth factor (FGF)-induced growth arrest of chondrocytes is a unique cell type-specific response which contrasts with the proliferative response of most cell types and underlies several genetic skeletal disorders caused by activating FGF receptor (FGFR) mutations. We have shown that one of the earliest key events in FGF-induced growth arrest is dephosphorylation of the retinoblastoma protein (Rb) family member p107 by protein phosphatase 2A (PP2A), a ubiquitously expressed multisubunit phosphatase. In this report, we show that the PP2A-B55α holoenzyme (PP2A containing the B55α subunit) is responsible for this phenomenon. Only the B55α (55-kDa regulatory subunit, alpha isoform) regulatory subunit of PP2A was able to bind p107, and this interaction was induced by FGF in chondrocytes but not in other cell types. Small interfering RNA (siRNA)-mediated knockdown of B55α prevented p107 dephosphorylation and FGF-induced growth arrest of RCS (rat chondrosarcoma) chondrocytes. Importantly, the B55α subunit bound with higher affinity to dephosphorylated p107. Since the p107 region interacting with B55α is also the site of cyclin-dependent kinase (CDK) binding, B55α association may also prevent p107 phosphorylation by CDKs. FGF treatment induces dephosphorylation of the B55α subunit itself on several serine residues that drastically increases the affinity of B55α for the PP2A A/C dimer and p107. Together these observations suggest a novel mechanism of p107 dephosphorylation mediated by activation of PP2A through B55α dephosphorylation. This mechanism might be a general signal transduction pathway used by PP2A to initiate cell cycle arrest when required by external signals.
Collapse
|
37
|
Schmidt EM, Schmid E, Münzer P, Hermann A, Eyrich AK, Russo A, Walker B, Gu S, vom Hagen JM, Faggio C, Schaller M, Föller M, Schöls L, Gawaz M, Borst O, Storch A, Stournaras C, Lang F. Chorein sensitivity of cytoskeletal organization and degranulation of platelets. FASEB J 2013; 27:2799-806. [PMID: 23568775 DOI: 10.1096/fj.13-229286] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Chorea-acanthocytosis (ChAc), a lethal disease caused by defective chorein, is characterized by neurodegeneration and erythrocyte acanthocytosis. The functional significance of chorein in other cell types remained ill-defined. The present study revealed chorein expression in blood platelets. As compared to platelets from healthy volunteers, platelets from patients with ChAc displayed a 47% increased globular/filamentous actin ratio, indicating actin depolymerization. Moreover, phosphoinositide-3-kinase subunit p85 phosphorylation, p21 protein-activated kinase (PAK1) phosphorylation, as well as vesicle-associated membrane protein 8 (VAMP8) expression were significantly reduced in platelets from patients with ChAc (by 17, 22, and 39%, respectively) and in megakaryocytic (MEG-01) cells following chorein silencing (by 16, 54, and 11%, respectively). Activation-induced platelet secretion from dense granules (ATP release) and α granules (P-selectin exposure) were significantly less (by 55% after stimulation with 1 μg/ml CRP and by 33% after stimulation with 5 μM TRAP, respectively) in ChAc platelets than in control platelets. Furthermore, platelet aggregation following stimulation with different platelet agonists was significantly impaired. These observations reveal a completely novel function of chorein, i.e., regulation of secretion and aggregation of blood platelets.
Collapse
Affiliation(s)
- Eva-Maria Schmidt
- Department of Physiology, University of Tübingen, Gmelinstrasse 5, 72076 Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Singhal R, Kandel ES. The response to PAK1 inhibitor IPA3 distinguishes between cancer cells with mutations in BRAF and Ras oncogenes. Oncotarget 2013; 3:700-8. [PMID: 22869096 PMCID: PMC3443253 DOI: 10.18632/oncotarget.587] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
While new drugs aimed at BRAF-mutated cancers are entering clinical practice, cells and tumors with activating Ras mutations are relatively resistant to those and quite a few other anti-cancer agents. This inspires the effort to reverse this resistance or to uncover new vulnerabilities in such resistant cancers. IPA3 has been originally identified as a small molecule inhibitor of p21-activated protein kinase 1 (PAK1), a candidate therapeutic target in human malignancies. We have tested a battery of melanoma and colon carcinoma cell lines that carry mutations in BRAF, NRAS and KRAS genes and have observed that those with NRAS and KRAS mutations are more sensitive to killing by IPA3. Genetic manipulations suggest that the differential response depends not just on these oncogenes, but also on additional events that were co-selected during tumor evolution. Furthermore, sublethal doses of IPA3 or ectopic expression of dominant-negative PAK1 sensitized Ras-mutated cells to GDC-0897 and AZD6244, which otherwise have reduced efficiency against cells with activated Ras. Dominant-negative PAK1 also reduced the growth of NRAS-mutated cells in confluent cultures, but, unlike IPA3, caused no significant toxicity. Although it remains to be proven that all the effects of IPA3 are exclusively due to inhibition of PAK1, our findings point to the existence of selective vulnerabilities, which are associated with Ras mutations and could be useful for better understanding and treatment of a large subset of tumors.
Collapse
Affiliation(s)
- Ruchi Singhal
- Roswell Park Cancer Institute, Department of Cell Stress Biology, Elm and Carlton St., Buffalo, NY 142263, USA
| | | |
Collapse
|
39
|
Gu S, Kounenidakis M, Schmidt EM, Deshpande D, Alkahtani S, Alarifi S, Föller M, Alevizopoulos K, Lang F, Stournaras C. Rapid activation of FAK/mTOR/p70S6K/PAK1-signaling controls the early testosterone-induced actin reorganization in colon cancer cells. Cell Signal 2013; 25:66-73. [DOI: 10.1016/j.cellsig.2012.08.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
40
|
Itoh T, Hasegawa J. Mechanistic insights into the regulation of circular dorsal ruffle formation. J Biochem 2012; 153:21-9. [PMID: 23175656 DOI: 10.1093/jb/mvs138] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Growth factor stimulations induce dynamic changes in the cytoskeleton beneath the plasma membrane. Among them is the formation of membrane ruffles organized in a circular array, called 'circular dorsal ruffles' (CDRs). Physiological functions of CDRs include downregulation of cell growth by desensitizing the signalling from growth factor receptors as well as rearrangement of adhesion sites at the onset of cell migration. For the formation of CDRs, not only the activators of actin polymerization, such as N-WASP and the Arp2/3-complex, but also membrane deforming proteins with BAR/F-BAR domains are necessary. Small GTPases are also involved in the formation of CDRs by controlling intracellular trafficking through endosomes. Moreover, recent analyses of another circular cytoskeletal structure, podosome rosettes, have revealed common molecular features shared with CDRs. Among them, the roles of PI3-kinase and phosphoinositide 5-phosphatase may hold the key to the induction of these circular structures.
Collapse
Affiliation(s)
- Toshiki Itoh
- Division of Membrane Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Kobe 650-0017, Japan.
| | | |
Collapse
|
41
|
Tseng P, Judy JW, Di Carlo D. Magnetic nanoparticle-mediated massively parallel mechanical modulation of single-cell behavior. Nat Methods 2012; 9:1113-9. [PMID: 23064517 PMCID: PMC3501759 DOI: 10.1038/nmeth.2210] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 09/06/2012] [Indexed: 12/31/2022]
Abstract
We report a technique for generating controllable, time-varying and localizable forces on arrays of cells in a massively parallel fashion. To achieve this, we grow magnetic nanoparticle-dosed cells in defined patterns on micromagnetic substrates. By manipulating and coalescing nanoparticles within cells, we apply localized nanoparticle-mediated forces approaching cellular yield tensions on the cortex of HeLa cells. We observed highly coordinated responses in cellular behavior, including the p21-activated kinase-dependent generation of active, leading edge-type filopodia and biasing of the metaphase plate during mitosis. The large sample size and rapid sample generation inherent to this approach allow the analysis of cells at an unprecedented rate: in a single experiment, potentially tens of thousands of cells can be stimulated for high statistical accuracy in measurements. This technique shows promise as a tool for both cell analysis and control.
Collapse
Affiliation(s)
- Peter Tseng
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
- Department of Electrical Engineering, University of California, Los Angeles, Los Angeles, California
| | - Jack W. Judy
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
- Department of Electrical Engineering, University of California, Los Angeles, Los Angeles, California
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
42
|
Staser K, Shew MA, Michels EG, Mwanthi MM, Yang FC, Clapp DW, Park SJ. A Pak1-PP2A-ERM signaling axis mediates F-actin rearrangement and degranulation in mast cells. Exp Hematol 2012; 41:56-66.e2. [PMID: 23063725 DOI: 10.1016/j.exphem.2012.10.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 10/01/2012] [Accepted: 10/06/2012] [Indexed: 01/12/2023]
Abstract
Mast cells coordinate allergy and allergic asthma and are crucial cellular targets in therapeutic approaches to inflammatory disease. Allergens cross-link immunoglobulin E bound at high-affinity receptors on the mast cell's surface, causing release of preformed cytoplasmic granules containing inflammatory molecules, including histamine, a principal effector of fatal septic shock. Both p21 activated kinase 1 (Pak1) and protein phosphatase 2A (PP2A) modulate mast cell degranulation, but the molecular mechanisms underpinning these observations and their potential interactions in common or disparate pathways are unknown. In this study, we use genetic and other approaches to show that Pak1's kinase-dependent interaction with PP2A potentiates PP2A's subunit assembly and activation. PP2A then dephosphorylates threonine 567 of Ezrin/Radixin/Moesin (ERM) molecules that have been shown to couple F-actin to the plasma membrane in other cell systems. In our study, the activity of this Pak1-PP2A-ERM axis correlates with impaired systemic histamine release in Pak1(-/-) mice and defective F-actin rearrangement and impaired degranulation in Ezrin disrupted (Mx1Cre(+)Ezrin(flox/flox)) primary mast cells. This heretofore unknown mechanism of mast cell degranulation provides novel therapeutic targets in allergy and asthma and may inform studies of kinase regulation of cytoskeletal dynamics in other cell lineages.
Collapse
Affiliation(s)
- Karl Staser
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Regulation of adherens junction dynamics by phosphorylation switches. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:125295. [PMID: 22848810 PMCID: PMC3403498 DOI: 10.1155/2012/125295] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 05/21/2012] [Accepted: 05/22/2012] [Indexed: 12/15/2022]
Abstract
Adherens junctions connect the actin cytoskeleton of neighboring cells through transmembrane cadherin receptors and a network of adaptor proteins. The interactions between these adaptors and cadherin as well as the activity of actin regulators localized to adherens junctions are tightly controlled to facilitate cell junction assembly or disassembly in response to changes in external or internal forces and/or signaling. Phosphorylation of tyrosine, serine, or threonine residues acts as a switch on the majority of adherens junction proteins, turning "on" or "off" their interactions with other proteins and/or their enzymatic activity. Here, we provide an overview of the kinases and phosphatases regulating phosphorylation of adherens junction proteins and bring examples of phosphorylation events leading to the assembly or disassembly of adherens junctions, highlighting the important role of phosphorylation switches in regulating their dynamics.
Collapse
|
44
|
Jung Y, Oh SH, Witek RP, Petersen BE. Somatostatin stimulates the migration of hepatic oval cells in the injured rat liver. Liver Int 2012; 32:312-320. [PMID: 22098068 PMCID: PMC3253984 DOI: 10.1111/j.1478-3231.2011.02642.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 08/16/2011] [Indexed: 12/12/2022]
Abstract
BACKGROUND Somatostatin is a pleiotropic peptide, exerting a variety of effects through its receptor subtypes. Recently, somatostatin has been shown to act as a chemoattractant for haematopoietic progenitor cells and hepatic oval cells (HOC) via receptor subtype 2 and subtype 4 (SSTR4) respectively. AIMS We investigated the in vivo effect of somatostatin/SSTR4 on HOC migration in the injured liver model of rats and the type of signalling molecules associated with the chemotactic function. METHODS Migration assay, HOC transplantation and phosphatidylinositol-3-kinase (PI3K) signalling were assessed with or without somatostatin and an analogue of somatostatin (TT232) that specifically binds to SSTR4. RESULTS TT232 was shown to have an antimigratory action on HOC induced by somatostatin in vitro. In HOC transplantation experiments, a lower number of donor-derived cells were detected in TT232-treated animals, as compared with control animals. Activation of PI3K was observed in HOC exposed to somatostatin, and this activation was suppressed by either SSTR4 antibody or TT232-pretreatment. In addition, a PI3K inhibitor abrogated the motility of HOC. CONCLUSION Together, these data suggest that somatostatin stimulates the migration of HOC within injured liver through SSTR4, and this action appears to be mediated by the PI3K pathway.
Collapse
Affiliation(s)
- Youngmi Jung
- Department of Biological Sciences, Pusan National University, Pusan, Korea.
| | | | | | | |
Collapse
|
45
|
Föller M, Hermann A, Gu S, Alesutan I, Qadri SM, Borst O, Schmidt E, Schiele F, Hagen JMV, Saft C, Schöls L, Lerche H, Stournaras C, Storch A, Lang F. Chorein‐sensitive polymerization of cortical actin and suicidal cell death in chorea‐acanthocytosis. FASEB J 2012; 26:1526-34. [DOI: 10.1096/fj.11-198317] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Michael Föller
- Department of PhysiologyUniversity of TübingenTübingenGermany
| | - Andreas Hermann
- Division of Neurodegenerative DiseasesDepartment of NeurologyUniversity of TechnologyDresdenGermany
| | - Shuchen Gu
- Department of PhysiologyUniversity of TübingenTübingenGermany
| | - Ioana Alesutan
- Department of PhysiologyUniversity of TübingenTübingenGermany
| | - Syed M. Qadri
- Department of PhysiologyUniversity of TübingenTübingenGermany
| | - Oliver Borst
- Department of PhysiologyUniversity of TübingenTübingenGermany
| | | | - Franziska Schiele
- Department of NeurologyUniversity of TübingenTübingenGermany
- Hertie‐Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Jennifer Müller vom Hagen
- Department of NeurologyUniversity of TübingenTübingenGermany
- Hertie‐Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Carsten Saft
- Department of NeurologyRuhr UniversityBochumGermany
| | - Ludger Schöls
- Department of NeurologyUniversity of TübingenTübingenGermany
- Hertie‐Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
- DZNEResearch Site TübingenTübingenGermany
| | - Holger Lerche
- Department of NeurologyUniversity of TübingenTübingenGermany
- Hertie‐Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | | | - Alexander Storch
- Division of Neurodegenerative DiseasesDepartment of NeurologyUniversity of TechnologyDresdenGermany
- German Center for Neurodegenerative Diseases (DZNE)Research Site DresdenDresdenGermany
| | - Florian Lang
- Department of PhysiologyUniversity of TübingenTübingenGermany
| |
Collapse
|
46
|
Naik MU, Naik UP. Contra-regulation of calcium- and integrin-binding protein 1-induced cell migration on fibronectin by PAK1 and MAP kinase signaling. J Cell Biochem 2011; 112:3289-99. [PMID: 21748785 PMCID: PMC3196778 DOI: 10.1002/jcb.23255] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Calcium- and integrin-binding protein 1 (CIB1) has been shown to be involved in cell spreading and migration. The signaling events regulated by CIB1 during cell migration are poorly understood. Here we found that accumulation of CIB1 at the tip of the filopodia requires an intact cytoskeleton. Depletion of CIB1 using shRNA affects formation of FAK- and phosphotyrosine-rich focal adhesions without affecting stress fiber formation. Overexpression of CIB1 results in cell migration on fibronectin and Erk1/2 MAP kinase activation. CIB1-induced cell migration is dependent upon Erk1/2 activation, since it is inhibited by the MEK-specific inhibitor PD98059. Furthermore, CIB1-induced cell migration, as well as Erk1/2 activation, is dependent on PKC, Src family kinases as well as PI-3 kinase as it is inhibited by bisindolylmaleimide 1, PP2, and wortmannin, respectively, in a dose-dependent manner. Co-expression of dominant-negative Cdc42 completely abolished CIB1-induced cell migration. Additionally, co-expression of constitutively active, but not dominant negative PAK1, a CIB1 binding protein, inhibited CIB1-induced cell migration. These results suggest that CIB1 positively regulates cell migration and is necessary for the recruitment of FAK to the focal adhesions. Furthermore, CIB1-induced cell migration is dependent on MAP kinase signaling and its function is attenuated by PAK1.
Collapse
Affiliation(s)
- Meghna U. Naik
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Ulhas P. Naik
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
- Department of Biochemistry and Chemistry, University of Delaware, Newark, DE, USA
- Department of Chemical Engineering, University of Delaware, Newark, DE, USA
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, USA
| |
Collapse
|
47
|
Zaichick SV, Bohannon KP, Smith GA. Alphaherpesviruses and the cytoskeleton in neuronal infections. Viruses 2011; 3:941-81. [PMID: 21994765 PMCID: PMC3185784 DOI: 10.3390/v3070941] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 06/03/2011] [Accepted: 06/17/2011] [Indexed: 12/13/2022] Open
Abstract
Following infection of exposed peripheral tissues, neurotropic alphaherpesviruses invade nerve endings and deposit their DNA genomes into the nuclei of neurons resident in ganglia of the peripheral nervous system. The end result of these events is the establishment of a life-long latent infection. Neuroinvasion typically requires efficient viral transmission through a polarized epithelium followed by long-distance transport through the viscous axoplasm. These events are mediated by the recruitment of the cellular microtubule motor proteins to the intracellular viral particle and by alterations to the cytoskeletal architecture. The focus of this review is the interplay between neurotropic herpesviruses and the cytoskeleton.
Collapse
Affiliation(s)
- Sofia V Zaichick
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | | | | |
Collapse
|
48
|
El-Osta MA, Idkowiak-Baldys J, Hannun YA. Delayed phosphorylation of classical protein kinase C (PKC) substrates requires PKC internalization and formation of the pericentrion in a phospholipase D (PLD)-dependent manner. J Biol Chem 2011; 286:19340-53. [PMID: 21478146 DOI: 10.1074/jbc.m110.152330] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
It was previously demonstrated that sustained activation (30-60 min) of protein kinase C (PKC) results in translocation of PKC α and βII to the pericentrion, a dynamic subset of the recycling compartment whose formation is dependent on PKC and phospholipase D (PLD). Here we investigated whether the formation of the pericentrion modulates the ability of PKC to phosphorylate substrates, especially if it reduces substrate phosphorylation by sequestering PKC. Surprisingly, using an antibody that detects phosphosubstrates of classical PKCs, the results showed that the majority of PKC phosphosubstrates are phosphorylated with delayed kinetics, correlating with the time frame of PKC translocation to the pericentrion. Substrate phosphorylation was blocked by PLD inhibitors and was not observed in response to activation of a PKC βII mutant (F663D) that is defective in interaction with PLD and in internalization. Phosphorylation was also inhibited by blocking clathrin-dependent endocytosis, demonstrating a requirement for endocytosis for the PKC-dependent major phosphorylation effects. Serotonin receptor activation by serotonin showed a similar response to phorbol 12-myristate 13-acetate, implicating a potential role of delayed kinetics in G protein-coupled receptor signaling. Evaluation of candidate substrates revealed that the phosphorylation of the PKC substrate p70S6K kinase behaved in a similar manner. Gradient-based fractionation revealed that the majority of these PKC substrates reside within the pericentrion-enriched fractions and not in the plasma membrane. Finally, proteomic analysis of the pericentrion-enriched fractions revealed several proteins as known PKC substrates and/or proteins involved in endocytic trafficking. These results reveal an important role for PKC internalization and for the pericentrion as key determinants/amplifiers of PKC action.
Collapse
Affiliation(s)
- Mohamad A El-Osta
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | |
Collapse
|
49
|
Du J, Sun C, Hu Z, Yang Y, Zhu Y, Zheng D, Gu L, Lu X. Lysophosphatidic acid induces MDA-MB-231 breast cancer cells migration through activation of PI3K/PAK1/ERK signaling. PLoS One 2010; 5:e15940. [PMID: 21209852 PMCID: PMC3012724 DOI: 10.1371/journal.pone.0015940] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 11/30/2010] [Indexed: 01/28/2023] Open
Abstract
Background Enhanced motility of cancer cells is a critical step in promoting tumor metastasis. Lysophosphatidic acid (LPA), representing the major mitogenic activity in serum, stimulates migration in various types of cancer cells. However, the underlying signaling mechanisms for LPA-induced motility of cancer cells remain to be elucidated. Methodology/Principal Findings In this study, we found that LPA dose-dependently stimulated migration of MDA-MB-231 breast cancer cells, with 10 µM being the most effective. LPA also increased ERK activity and the MEK inhibitor U0126 could block LPA-induced ERK activity and cell migration. In addition, LPA induced PAK1 activation while ERK activation and cell migration were inhibited by ectopic expression of an inactive mutant form of PAK1 in MDA-MB-231 cells. Furthermore, LPA increased PI3K activity, and the PI3K inhibitor LY294002 inhibited both LPA-induced PAK1/ERK activation and cell migration. Moreover, in the breast cancer cell, LPA treatment resulted in remarkable production of reactive oxygen species (ROS), while LPA-induced ROS generation, PI3K/PAK1/ERK activation and cell migration could be inhibited by N-acetyl-L-Cysteine, a scavenger of ROS. Conclusions/Significance Taken together, this study identifies a PI3K/PAK1/ERK signaling pathway for LPA-stimulated breast cancer cell migration. These data also suggest that ROS generation plays an essential role in the activation of LPA-stimulated PI3K/PAK1/ERK signaling and breast cancer cell migration. These findings may provide a basis for designing future therapeutic strategy for blocking breast cancer metastasis.
Collapse
Affiliation(s)
- Jun Du
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Chongqi Sun
- Kangda College, Nanjing Medical University, Nanjing, China
| | - Zhenzhen Hu
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Yu Yang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Yichao Zhu
- Cancer Center, Nanjing Medical University, Nanjing, China
| | - Datong Zheng
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Luo Gu
- Cancer Center, Nanjing Medical University, Nanjing, China
- * E-mail: (LG); (XL)
| | - Xiang Lu
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- * E-mail: (LG); (XL)
| |
Collapse
|
50
|
Tornillo G, Bisaro B, Camacho-Leal MDP, Galiè M, Provero P, Di Stefano P, Turco E, Defilippi P, Cabodi S. p130Cas promotes invasiveness of three-dimensional ErbB2-transformed mammary acinar structures by enhanced activation of mTOR/p70S6K and Rac1. Eur J Cell Biol 2010; 90:237-48. [PMID: 20961652 DOI: 10.1016/j.ejcb.2010.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 07/22/2010] [Accepted: 09/01/2010] [Indexed: 11/29/2022] Open
Abstract
ErbB2 over-expression is detected in approximately 25% of invasive breast cancers and is strongly associated with poor patient survival. We have previously demonstrated that p130Cas adaptor is a crucial mediator of ErbB2 transformation. Here, we analysed the molecular mechanisms through which p130Cas controls ErbB2-dependent invasion in three-dimensional cultures of mammary epithelial cells. Concomitant p130Cas over-expression and ErbB2 activation enhance PI3K/Akt and Erk1/2 MAPK signalling pathways and promote invasion of mammary acini. By using pharmacological inhibitors, we demonstrate that both signalling cascades are required for the invasive behaviour of p130Cas over-expressing and ErbB2 activated acini. Erk1/2 MAPK and PI3K/Akt signalling triggers invasion through distinct downstream effectors involving mTOR/p70S6K and Rac1 activation, respectively. Moreover, in silico analyses indicate that p130Cas expression in ErbB2 positive human breast cancers significantly correlates with higher risk to develop distant metastasis, thus underlying the value of the p130Cas/ErbB2 synergism in regulating breast cancer invasion. In conclusion, high levels of p130Cas favour progression of ErbB2-transformed cells towards an invasive phenotype.
Collapse
Affiliation(s)
- Giusy Tornillo
- Molecular Biology Center, Department of Genetics, Biology and Biochemistry, University of Torino, Via Nizza 52, Torino, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|