1
|
Funakoshi M, Araki T. Mechanism of initiation and regulation of axonal degeneration with special reference to NMNATs and Sarm1. Neurosci Res 2023; 197:3-8. [PMID: 34767875 DOI: 10.1016/j.neures.2021.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 10/19/2022]
Abstract
Axonal degeneration is observed in a variety of contexts in both the central and peripheral nervous systems. Pathological signaling to regulate the progression of axonal degeneration has long been studied using Wallerian degeneration, the prototypical axonal degradation observed after injury, as a representative model. Understanding metabolism of nicotinamide adenine dinucleotide (NAD+) and the functional regulation of Sarm1 has generated great progress in this field, but there are a number of remaining questions. Here, in this short review, we describe our current understanding of the axonal degeneration mechanism, with special reference to the biology related to wlds mice and Sarm1. Furthermore, variations of axonal degeneration initiation are discussed in order to address the remaining questions needed for mechanistic clarification.
Collapse
Affiliation(s)
- Masabumi Funakoshi
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Toshiyuki Araki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan.
| |
Collapse
|
2
|
Atkins M, Nicol X, Fassier C. Microtubule remodelling as a driving force of axon guidance and pruning. Semin Cell Dev Biol 2023; 140:35-53. [PMID: 35710759 DOI: 10.1016/j.semcdb.2022.05.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/26/2022] [Accepted: 05/31/2022] [Indexed: 01/28/2023]
Abstract
The establishment of neuronal connectivity relies on the microtubule (MT) cytoskeleton, which provides mechanical support, roads for axonal transport and mediates signalling events. Fine-tuned spatiotemporal regulation of MT functions by tubulin post-translational modifications and MT-associated proteins is critical for the coarse wiring and subsequent refinement of neuronal connectivity. The defective regulation of these processes causes a wide range of neurodevelopmental disorders associated with connectivity defects. This review focuses on recent studies unravelling how MT composition, post-translational modifications and associated proteins influence MT functions in axon guidance and/or pruning to build functional neuronal circuits. We here summarise experimental evidence supporting the key role of this network as a driving force for growth cone steering and branch-specific axon elimination. We further provide a global overview of the MT-interactors that tune developing axon behaviours, with a special emphasis on their emerging versatility in the regulation of MT dynamics/structure. Recent studies establishing the key and highly selective role of the tubulin code in the regulation of MT functions in axon pathfinding are also reported. Finally, our review highlights the emerging molecular links between these MT regulation processes and guidance signals that wire the nervous system.
Collapse
Affiliation(s)
- Melody Atkins
- INSERM, UMR-S 1270, Institut du Fer à Moulin, Sorbonne Université, F-75005 Paris, France
| | - Xavier Nicol
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France
| | - Coralie Fassier
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France.
| |
Collapse
|
3
|
Chang YH, Chang SW, Hsu WT, Yang CP, Lo YL, Chen CJ, Tsai HF, Shiau MY. Implication of Adipogenesis-Coupled CRMP2 Functional Profile in Metabolic Homeostasis and Imbalance. Biomedicines 2022; 10:biomedicines10102603. [PMID: 36289868 PMCID: PMC9599587 DOI: 10.3390/biomedicines10102603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Our previous studies demonstrated that collapsin response mediator protein 2 (CRMP2) is associated with obesity and, in addition, that hyperglycemia-suppressed CRMP2 augments malignant traits of colorectal cancer and is associated with advanced tumor stage. Regulation of CRMP2 profile was further explored in this study using 3T3-L1 pre-adipocyte adipogenesis as a study model for illustrating the roles of CRMP2 in metabolic homeostasis. Hyperglycemia inhibited expression of CRMP2, adipogenic machinery and adipocyte markers. CRMP2 displayed f-CRMP2 (62~66 kDa) and s-CMRP2 (58 kDa) isoforms at the growth arrest phase. Expression of s-CRMP2 was coupled with the mitotic clonal expansion (MCE) phase to direct cell proliferation and rapidly down-regulated in post-mitotic cells. In the late differentiation phase, f-CRMP2 was co-localized with tubulin in the cortical area. Insulin-enhanced CRMP2-glucose transporter 4 (GLUT4) co-localization and CRMP2 puncta on lipid droplets (LDs) suggested participation of CRMP2 in GLUT4 translocation and LD fusion. Collectively, the CRMP2 functional profile must be finely controlled to adjust cytoskeletal stability for meeting dynamic cellular needs. Manipulating the s-CRMP2/f-CRMP2 ratio and thus the cytoskeleton dynamics is anticipated to improve glucose uptake and insulin sensitivity. In summary, our data provide molecular evidence explaining the functions of CRMP2 in physiological, pathological and disease progression in metabolic homeostasis and disorders related to metabolic abnormalities, including cancer.
Collapse
Affiliation(s)
- Yih-Hsin Chang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Shu-Wen Chang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Nursing, College of Nursing, Hungkuang University, Taichung 433, Taiwan
| | - Wei-Ting Hsu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ching-Ping Yang
- Department of Medical Technology, Jenteh Junior College of Medicine, Nursing and Management, Miaoli 356, Taiwan
| | - Yu-Li Lo
- Department and Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan
| | - Hui-Fang Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 402, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Ming-Yuh Shiau
- Department of Nursing, College of Nursing, Hungkuang University, Taichung 433, Taiwan
- Correspondence: ; Tel.: +886-4-26318652 (ext. 7090); Fax: +886-4-26331198
| |
Collapse
|
4
|
Kawamoto Y, Tada M, Asano T, Nakamura H, Jitsuki-Takahashi A, Makihara H, Kubota S, Hashiguchi S, Kunii M, Ohshima T, Goshima Y, Takeuchi H, Doi H, Nakamura F, Tanaka F. Phosphorylated CRMP1, axon guidance protein, is a component of spheroids and is involved in axonal pathology in amyotrophic lateral sclerosis. Front Neurol 2022; 13:994676. [PMID: 36237616 PMCID: PMC9552802 DOI: 10.3389/fneur.2022.994676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/06/2022] [Indexed: 11/19/2022] Open
Abstract
In amyotrophic lateral sclerosis (ALS), neurodegeneration is characterized by distal axonopathy that begins at the distal axons, including the neuromuscular junctions, and progresses proximally in a “dying back” manner prior to the degeneration of cell bodies. However, the molecular mechanism for distal axonopathy in ALS has not been fully addressed. Semaphorin 3A (Sema3A), a repulsive axon guidance molecule that phosphorylates collapsin response mediator proteins (CRMPs), is known to be highly expressed in Schwann cells near distal axons in a mouse model of ALS. To clarify the involvement of Sema3A–CRMP signaling in the axonal pathogenesis of ALS, we investigated the expression of phosphorylated CRMP1 (pCRMP1) in the spinal cords of 35 patients with sporadic ALS and seven disease controls. In ALS patients, we found that pCRMP1 accumulated in the proximal axons and co-localized with phosphorylated neurofilaments (pNFs), which are a major protein constituent of spheroids. Interestingly, the pCRMP1:pNF ratio of the fluorescence signal in spheroid immunostaining was inversely correlated with disease duration in 18 evaluable ALS patients, indicating that the accumulation of pCRMP1 may precede that of pNFs in spheroids or promote ALS progression. In addition, overexpression of a phospho-mimicking CRMP1 mutant inhibited axonal outgrowth in Neuro2A cells. Taken together, these results indicate that pCRMP1 may be involved in the pathogenesis of axonopathy in ALS, leading to spheroid formation through the proximal progression of axonopathy.
Collapse
Affiliation(s)
- Yuko Kawamoto
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Mikiko Tada
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tetsuya Asano
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Haruko Nakamura
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Aoi Jitsuki-Takahashi
- Department of Biochemistry, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Hiroko Makihara
- Department of Nursing Course Biological Science and Nursing, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shun Kubota
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shunta Hashiguchi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Misako Kunii
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Toshio Ohshima
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hideyuki Takeuchi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroshi Doi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Fumio Nakamura
- Department of Biochemistry, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- *Correspondence: Fumiaki Tanaka
| |
Collapse
|
5
|
Chang YH, Yang HJ, Chen HW, Hsiao CW, Hsieh YC, Chan YW, Chang SW, Hwang WL, Chen WS, Cheng HH, Chou TY, Chang FP, Ho HL, Chu FY, Lo YL, Chen CJ, Tsai HF, Shiau MY. Characterization of Collapsin Response Mediator Protein 2 in Colorectal Cancer Progression in Subjects with Diabetic Comorbidity. Cells 2022; 11:cells11040727. [PMID: 35203376 PMCID: PMC8869905 DOI: 10.3390/cells11040727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Common demographic risk factors are identified in colorectal cancer (CRC) and type 2 diabetes mellitus (DM), nevertheless, the molecular link and mechanism for CRC-DM comorbidity remain elusive. Dysregulated glycogen synthase kinase-3 beta under metabolic imbalance is suggested to accelerate CRC pathogenesis/progression via regulating collpasin response mediator protein-2 (CRMP2). Accordingly, roles of CRMP2 in CRC and CRC-DM patients were investigated for elucidating the molecular convergence of CRC and DM. METHODS CRMP2 profile in tumor tissues from CRC and CRC-DM patients was investigated to explore the link between CRC and DM etiology. Meanwhile, molecular mechanism of glucose to regulate CRMP2 profile and CRC characteristics was examined in vitro and in vivo. RESULTS CRMP2 was significantly lower in tumor lesions and associated with advanced tumor stage in CRC-DM patients. Physiological hyperglycemia suppressed CRMP2 expression/activity and augmented malignant characteristics of CRC cells. Hyperglycemia promotes actin de-polymerization, cytoskeleton flexibility and cell proliferation/metastasis by downregulating CRMP2 profile and thus contributes to CRC disease progression. CONCLUSIONS This study uncovers molecular evidence to substantiate and elucidate the link between CRC and T2DM, as well as characterizing the roles of CRMP2 in CRC-DM. Accordingly, altered metabolic adaptations are promising targets for anti-diabetic and cancer strategies.
Collapse
Affiliation(s)
- Yih-Hsin Chang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.C.); (H.-J.Y.); (H.-W.C.); (C.-W.H.); (Y.-C.H.); (Y.-W.C.); (S.-W.C.); (W.-L.H.)
| | - Hui-Ju Yang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.C.); (H.-J.Y.); (H.-W.C.); (C.-W.H.); (Y.-C.H.); (Y.-W.C.); (S.-W.C.); (W.-L.H.)
- Department of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (W.-S.C.); (H.-H.C.)
| | - Huan-Wen Chen
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.C.); (H.-J.Y.); (H.-W.C.); (C.-W.H.); (Y.-C.H.); (Y.-W.C.); (S.-W.C.); (W.-L.H.)
| | - Chiao-Wan Hsiao
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.C.); (H.-J.Y.); (H.-W.C.); (C.-W.H.); (Y.-C.H.); (Y.-W.C.); (S.-W.C.); (W.-L.H.)
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 115, Taiwan
| | - Yi-Chen Hsieh
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.C.); (H.-J.Y.); (H.-W.C.); (C.-W.H.); (Y.-C.H.); (Y.-W.C.); (S.-W.C.); (W.-L.H.)
| | - Yu-Wei Chan
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.C.); (H.-J.Y.); (H.-W.C.); (C.-W.H.); (Y.-C.H.); (Y.-W.C.); (S.-W.C.); (W.-L.H.)
| | - Shu-Wen Chang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.C.); (H.-J.Y.); (H.-W.C.); (C.-W.H.); (Y.-C.H.); (Y.-W.C.); (S.-W.C.); (W.-L.H.)
- Department of Nursing, College of Nursing, Hungkuang University, Taichung 433, Taiwan
| | - Wei-Lun Hwang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.C.); (H.-J.Y.); (H.-W.C.); (C.-W.H.); (Y.-C.H.); (Y.-W.C.); (S.-W.C.); (W.-L.H.)
| | - Wei-Shone Chen
- Department of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (W.-S.C.); (H.-H.C.)
- Division of Colon & Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Hou-Hsuan Cheng
- Department of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (W.-S.C.); (H.-H.C.)
- Division of Colon & Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Teh-Ying Chou
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan; (T.-Y.C.); (F.-P.C.); (H.-L.H.)
| | - Fu-Pang Chang
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan; (T.-Y.C.); (F.-P.C.); (H.-L.H.)
| | - Hsiang-Ling Ho
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan; (T.-Y.C.); (F.-P.C.); (H.-L.H.)
| | - Fang-Yeh Chu
- Department of Clinical Pathology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan;
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University, Hsinchu 300, Taiwan
- School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Li Lo
- Department and Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan
| | - Hui-Fang Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 402, Taiwan;
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Ming-Yuh Shiau
- Department of Nursing, College of Nursing, Hungkuang University, Taichung 433, Taiwan
- Correspondence: ; Tel.: +886-4-26318652 (ext. 7090)
| |
Collapse
|
6
|
Sánchez-Huertas C, Herrera E. With the Permission of Microtubules: An Updated Overview on Microtubule Function During Axon Pathfinding. Front Mol Neurosci 2021; 14:759404. [PMID: 34924953 PMCID: PMC8675249 DOI: 10.3389/fnmol.2021.759404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/01/2021] [Indexed: 01/27/2023] Open
Abstract
During the establishment of neural circuitry axons often need to cover long distances to reach remote targets. The stereotyped navigation of these axons defines the connectivity between brain regions and cellular subtypes. This chemotrophic guidance process mostly relies on the spatio-temporal expression patterns of extracellular proteins and the selective expression of their receptors in projection neurons. Axon guidance is stimulated by guidance proteins and implemented by neuronal traction forces at the growth cones, which engage local cytoskeleton regulators and cell adhesion proteins. Different layers of guidance signaling regulation, such as the cleavage and processing of receptors, the expression of co-receptors and a wide variety of intracellular cascades downstream of receptors activation, have been progressively unveiled. Also, in the last decades, the regulation of microtubule (MT) assembly, stability and interactions with the submembranous actin network in the growth cone have emerged as crucial effector mechanisms in axon pathfinding. In this review, we will delve into the intracellular signaling cascades downstream of guidance receptors that converge on the MT cytoskeleton of the growing axon. In particular, we will focus on the microtubule-associated proteins (MAPs) network responsible of MT dynamics in the axon and growth cone. Complementarily, we will discuss new evidences that connect defects in MT scaffold proteins, MAPs or MT-based motors and axon misrouting during brain development.
Collapse
Affiliation(s)
- Carlos Sánchez-Huertas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Alicante, Spain
| | | |
Collapse
|
7
|
Cai S, Moutal A, Yu J, Chew LA, Isensee J, Chawla R, Gomez K, Luo S, Zhou Y, Chefdeville A, Madura C, Perez-Miller S, Bellampalli SS, Dorame A, Scott DD, François-Moutal L, Shan Z, Woodward T, Gokhale V, Hohmann AG, Vanderah TW, Patek M, Khanna M, Hucho T, Khanna R. Selective targeting of NaV1.7 via inhibition of the CRMP2-Ubc9 interaction reduces pain in rodents. Sci Transl Med 2021; 13:eabh1314. [PMID: 34757807 DOI: 10.1126/scitranslmed.abh1314] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The voltage-gated sodium NaV1.7 channel, critical for sensing pain, has been actively targeted by drug developers; however, there are currently no effective and safe therapies targeting NaV1.7. Here, we tested whether a different approach, indirect NaV1.7 regulation, could have antinociceptive effects in preclinical models. We found that preventing addition of small ubiquitin-like modifier (SUMO) on the NaV1.7-interacting cytosolic collapsin response mediator protein 2 (CRMP2) blocked NaV1.7 functions and had antinociceptive effects in rodents. In silico targeting of the SUMOylation site in CRMP2 (Lys374) identified >200 hits, of which compound 194 exhibited selective in vitro and ex vivo NaV1.7 engagement. Orally administered 194 was not only antinociceptive in preclinical models of acute and chronic pain but also demonstrated synergy alongside other analgesics—without eliciting addiction, rewarding properties, or neurotoxicity. Analgesia conferred by 194 was opioid receptor dependent. Our results demonstrate that 194 is a first-in-class protein-protein inhibitor that capitalizes on CRMP2-NaV1.7 regulation to deliver safe analgesia in rodents.
Collapse
Affiliation(s)
- Song Cai
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Jie Yu
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Lindsey A Chew
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Jörg Isensee
- Department of Anesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital of Cologne, University Cologne, Joseph-Stelzmann-Str 9, Cologne D-50931, Germany
| | - Reena Chawla
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
| | - Kimberly Gomez
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Shizhen Luo
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Yuan Zhou
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Aude Chefdeville
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Cynthia Madura
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Samantha Perez-Miller
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Shreya Sai Bellampalli
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Angie Dorame
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - David D Scott
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Liberty François-Moutal
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Zhiming Shan
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Taylor Woodward
- Department of Psychological and Brain Sciences, Program in Neuroscience and Gill Center for Biomolecular Science, Indiana University, Bloomington, IN 47405-2204, USA
| | - Vijay Gokhale
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
- College of Pharmacy, University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, USA
| | - Andrea G Hohmann
- Department of Psychological and Brain Sciences, Program in Neuroscience and Gill Center for Biomolecular Science, Indiana University, Bloomington, IN 47405-2204, USA
| | - Todd W Vanderah
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- Comprehensive Pain and Addiction Center, The University of Arizona, Tucson, AZ 85724, USA
| | - Marcel Patek
- Regulonix LLC, 1555 E. Entrada Segunda, Tucson, AZ 85718, USA
- Bright Rock Path LLC, Tucson, AZ 85724, USA
| | - May Khanna
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
- Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ 85721, USA
- Regulonix LLC, 1555 E. Entrada Segunda, Tucson, AZ 85718, USA
| | - Tim Hucho
- Department of Anesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital of Cologne, University Cologne, Joseph-Stelzmann-Str 9, Cologne D-50931, Germany
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
- Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ 85721, USA
- Comprehensive Pain and Addiction Center, The University of Arizona, Tucson, AZ 85724, USA
- Regulonix LLC, 1555 E. Entrada Segunda, Tucson, AZ 85718, USA
| |
Collapse
|
8
|
Muraoka S, Lin W, Takamatsu-Yukawa K, Hu J, Ikezu S, DeTure MA, Dickson DW, Emili A, Ikezu T. Enrichment of Phosphorylated Tau (Thr181) and Functionally Interacting Molecules in Chronic Traumatic Encephalopathy Brain-derived Extracellular Vesicles. Aging Dis 2021; 12:1376-1388. [PMID: 34527416 PMCID: PMC8407888 DOI: 10.14336/ad.2020.1007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/07/2020] [Indexed: 12/21/2022] Open
Abstract
Chronic Traumatic Encephalopathy (CTE) is a tauopathy that affects individuals with a history of mild repetitive brain injury. The initial neuropathologic changes of CTE include perivascular deposition of phosphorylated microtubule-associated protein tau (p-tau). Extracellular vesicles (EVs) are known to carry pathogenic molecules, such as tau in Alzheimer's disease and CTE suggesting their contribution in pathogenesis. We therefore examined the protein composition of EVs separated from CTE and an age-matched control brain tissues by tandem mass tag -mass spectrometry. The reporter ion intensity was used to quantify the identified molecules. A total of 516 common proteins were identified among three sets of experiments. Weighted protein co-expression network analysis identified 18 unique modules of co-expressed proteins. Two modules were significantly correlated with total tau (t-tau) and p-tau protein in the isolated EVs and enriched in cellular components and biological processes for synaptic vesicle secretion and multivesicular body-plasma membrane fusion. The p-tau (Thr181) level is significantly higher in CTE EVs compared to control EVs and can distinguish the two groups with 73.6% accuracy. A combination of t-tau or p-tau (Thr181) with SNAP-25, PLXNA4 or UBA1, enhanced the accuracy to 96.3, 93.8 and 93.8%, respectively. Bioinformatic protein-protein interaction analysis revealed the functional interaction of SNAP-25 and PLXNA4 with tau, suggesting their interaction in CTE EVs. These data indicate the future application of identified EV proteins for monitoring the CTE risk assessments and understanding the EV-mediated disease progression mechanism.
Collapse
Affiliation(s)
- Satoshi Muraoka
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | - Weiwei Lin
- Department of Biochemistry, Boston University, Boston, MA, USA.
- Center for Network Systems Biology, Boston University, Boston, MA, USA.
| | - Kayo Takamatsu-Yukawa
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | - Jianqiao Hu
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | - Seiko Ikezu
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | | | | | - Andrew Emili
- Department of Biochemistry, Boston University, Boston, MA, USA.
- Center for Network Systems Biology, Boston University, Boston, MA, USA.
| | - Tsuneya Ikezu
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
- Center for Systems Neuroscience, Boston University, Boston, MA, USA.
- Department of Neurology and Alzheimer’s Disease Center, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
9
|
Molecular Mechanism for PACAP 38-Induced Neurite Outgrowth in PC12 Cells. Neural Plast 2021; 2021:2522454. [PMID: 34422037 PMCID: PMC8371652 DOI: 10.1155/2021/2522454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/08/2021] [Accepted: 07/17/2021] [Indexed: 11/17/2022] Open
Abstract
The present research investigates the molecular mechanism of neurite outgrowth (protrusion elongation) under pituitary adenylate cyclase-activating polypeptide (PACAP) 38 treatments using a rat adrenal-derived pheochromocytoma cell line-PC12. This study specifically looks into the regulation of PACAP38-induced collapsing response mediator protein 2 (CRMP2) previously identified in a mouse brain ischemia model and which could be recovered by PACAP38 treatment. Previously, DNA microarray analysis revealed that PACAP 38-mediated neuroprotection involved not only CRMP2 but also pathways related to glycogen synthase kinase-3β (GSK-3β) and other signaling components. Thus, to clarify whether CRMP2 acts directly on PACAP38 or through GSK-3β as part of the mechanism of PACAP38-induced neurite outgrowth, we observed neurite outgrowth in the presence of GSK-3β inhibitors and activators. PC12 cells were treated with PACAP38 being added to the cell culture medium at concentrations of 10-7 M, 10-8 M, and 10-9 M. Post PACAP38 treatment, immunostaining was used to confirm protrusion elongation of the PC12 cells, while RT-PCR, two-dimensional gel electrophoresis in conjunction with Western blotting, and inhibition experiments were performed to confirm the expression of the PACAP gene, its receptors, and downstream signaling components. Our data show that neurite protrusion elongation by PACAP38 (10-7 M) in PC12 cells is mediated through the PAC1-R receptor as demonstrated by its suppression by a specific inhibitor PA-8. Inhibitor experiments suggested that PACAP38-triggered neurite protrusion follows a GSK-3β-regulated pathway, where the AKT and cAMP/ERK pathways are involved and where the inhibition of Rho/Roc could enhance neurite protrusion under PACAP38 stimulation. Although we could not yet confirm the exact role and position of CRMP2 in PACAP38-mediated PC12 cell elongation, it appears that its phosphorylation and dephosphorylation have a correlation with the neurite protrusion elongation through the interplay of CDK5, which needs to be investigated further.
Collapse
|
10
|
Carretero-Rodriguez L, Guðjónsdóttir R, Poparic I, Reilly ML, Chol M, Bianco IH, Chiapello M, Feret R, Deery MJ, Guthrie S. The Rac-GAP alpha2-Chimaerin Signals via CRMP2 and Stathmins in the Development of the Ocular Motor System. J Neurosci 2021; 41:6652-6672. [PMID: 34168008 PMCID: PMC8336708 DOI: 10.1523/jneurosci.0983-19.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 04/02/2021] [Accepted: 04/05/2021] [Indexed: 11/21/2022] Open
Abstract
A precise sequence of axon guidance events is required for the development of the ocular motor system. Three cranial nerves grow toward, and connect with, six extraocular muscles in a stereotyped pattern, to control eye movements. The signaling protein alpha2-chimaerin (α2-CHN) plays a pivotal role in the formation of the ocular motor system; mutations in CHN1, encoding α2-CHN, cause the human eye movement disorder Duane Retraction Syndrome (DRS). Our research has demonstrated that the manipulation of α2-chn signaling in the zebrafish embryo leads to ocular motor axon wiring defects, although the signaling cascades regulated by α2-chn remain poorly understood. Here, we demonstrate that several cytoskeletal regulatory proteins-collapsin response mediator protein 2 (CRMP2; encoded by the gene dpysl2), stathmin1, and stathmin 2-bind to α2-CHN. dpysl2, stathmin1, and especially stathmin2 are expressed by ocular motor neurons. We find that the manipulation of dpysl2 and of stathmins in zebrafish larvae leads to defects in both the axon wiring of the ocular motor system and the optokinetic reflex, impairing horizontal eye movements. Knockdowns of these molecules in zebrafish larvae of either sex caused axon guidance phenotypes that included defasciculation and ectopic branching; in some cases, these phenotypes were reminiscent of DRS. chn1 knock-down phenotypes were rescued by the overexpression of CRMP2 and STMN1, suggesting that these proteins act in the same signaling pathway. These findings suggest that CRMP2 and stathmins signal downstream of α2-CHN to orchestrate ocular motor axon guidance and to control eye movements.SIGNIFICANCE STATEMENT The precise control of eye movements is crucial for the life of vertebrate animals, including humans. In humans, this control depends on the arrangement of nerve wiring of the ocular motor system, composed of three nerves and six muscles, a system that is conserved across vertebrate phyla. Mutations in the protein alpha2-chimaerin have previously been shown to cause eye movement disorders (squint) and axon wiring defects in humans. Our recent work has unraveled how alpha2-chimaerin coordinates axon guidance of the ocular motor system in animal models. In this article, we demonstrate key roles for the proteins CRMP2 and stathmin 1/2 in the signaling pathway orchestrated by alpha2-chimaerin, potentially giving insight into the etiology of eye movement disorders in humans.
Collapse
Affiliation(s)
| | | | - Ivana Poparic
- Department of Developmental Neurobiology, King's College London, London SE1 1UL, United Kingdom
| | | | - Mary Chol
- Department of Developmental Neurobiology, King's College London, London SE1 1UL, United Kingdom
| | - Isaac H Bianco
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Marco Chiapello
- Cambridge Centre for Proteomics, Cambridge Systems Biology Centre, Department of Biochemistry, University of Cambridge, Cambridge CB2 1QR, United Kingdom
| | - Renata Feret
- Institute for Sustainable Plant Protection, National Research Council, 10135 Torino, Italy
| | - Michael J Deery
- Institute for Sustainable Plant Protection, National Research Council, 10135 Torino, Italy
| | - Sarah Guthrie
- School of Life Sciences, University of Sussex, Brighton BN7 9QG, United Kingdom
| |
Collapse
|
11
|
Zhao H, Wang L, Wang S, Chen X, Liang M, Zhang X, Wang J, Xu X. CHN1 promotes epithelial-mesenchymal transition via the Akt/GSK-3β/Snail pathway in cervical carcinoma. J Transl Med 2021; 19:295. [PMID: 34238315 PMCID: PMC8264971 DOI: 10.1186/s12967-021-02963-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/25/2021] [Indexed: 12/02/2022] Open
Abstract
Background Metastasis and invasion are crucial in determining the mortality of cervical carcinoma (CC) patients. The epithelial–mesenchymal transition (EMT) is now a universal explanation for the mechanisms of tumor metastasis. Α-chimeric protein (α-chimaerin, CHN1) plays an important role in the regulation of signal transduction and development. However, the molecular regulatory relationships between CHN1 and CC progression in relation to EMT have not yet been identified. Methods The expression of CHN1 in CC tissues, adjacent tissues, and lymph node metastases from CC patients was detected by immunohistochemistry. Upregulation and knockdown of CHN1 were achieved by transfection of CC cells. The effect of CHN1 on cell proliferation was determined by CCK-8 and plate clone formation assays. Changes in migration and invasion capabilities were evaluated using scratch migration and transwell invasion assays. The effect of CHN1 overexpression and interference on xenograft tumor growth was determined by tumor weight and pathological analyses. The expression of EMT-related mRNAs was measured by qRT-PCR in transfected CC cells. EMT-related proteins and Akt/GSK-3β/Snail signaling pathway-related proteins were also evaluated by western blotting. Results CHN1 was overexpressed in CC tissues and was associated with lymph node metastasis and low survival in CC patients. Overexpression of CHN1 promoted cell proliferation, migration, and invasion in CC cells. In contrast, silencing of CHN1 inhibited these phenomena. Overexpression of CHN1 promoted tumor formation in an in vivo xenograft tumor mouse model, with increased tumor volumes and weights. In addition, CHN1 induced the expression of EMT-related transcription factors, accompanied by the decreased expression of epithelial markers and increased expression of mesenchymal markers. The Akt/GSK-3β/Snail signaling pathway was activated by overexpression of CHN1 in vitro, and activation of this pathway was inhibited by the signaling pathway inhibitor LY294002. Conclusion These results suggest that CHN1 promotes the development and progression of cervical carcinoma via the Akt/GSK-3β/Snail pathway by inducing EMT. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02963-7.
Collapse
Affiliation(s)
- Haoqi Zhao
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.,Reproductive Physiology Laboratory, National Research Institute for Family Planning, Beijing, 100081, China.,National Engineering and Research Center of Continuous Casting Technology, China Iron and Steel Research Institute Group, Beijing, 100081, China
| | - Lan Wang
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215126, Jiangsu, China
| | - Shufang Wang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.,Reproductive Physiology Laboratory, National Research Institute for Family Planning, Beijing, 100081, China.,Department of Forensic Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xihua Chen
- Reproductive Physiology Laboratory, National Research Institute for Family Planning, Beijing, 100081, China
| | - Min Liang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.,Reproductive Physiology Laboratory, National Research Institute for Family Planning, Beijing, 100081, China
| | - Xin Zhang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.,Reproductive Physiology Laboratory, National Research Institute for Family Planning, Beijing, 100081, China
| | - Jiedong Wang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.,Reproductive Physiology Laboratory, National Research Institute for Family Planning, Beijing, 100081, China
| | - Xiangbo Xu
- Reproductive Physiology Laboratory, National Research Institute for Family Planning, Beijing, 100081, China.
| |
Collapse
|
12
|
Abstract
The chronification of pain can be attributed to changes in membrane receptors and channels underlying neuronal plasticity and signal transduction largely within nociceptive neurons that initiate and maintain pathological pain states. These proteins are subject to dynamic modification by posttranslational modifications, creating a code that controls protein function in time and space. Phosphorylation is an important posttranslational modification that affects ∼30% of proteins in vivo. Increased phosphorylation of various nociceptive ion channels and of their modulators underlies sensitization of different pain states. Cyclin-dependent kinases are proline-directed serine/threonine kinases that impact various biological and cellular systems. Cyclin-dependent kinase 5 (Cdk5), one member of this kinase family, and its activators p35 and p39 are expressed in spinal nerves, dorsal root ganglia, and the dorsal horn of the spinal cord. In neuropathic pain conditions, expression and/or activity of Cdk5 is increased, implicating Cdk5 in nociception. Experimental evidence suggests that Cdk5 is regulated through its own phosphorylation, through increasing p35's interaction with Cdk5, and through cleavage of p35 into p25. This narrative review discusses the molecular mechanisms of Cdk5-mediated regulation of target proteins involved in neuropathic pain. We focus on Cdk5 substrates that have been linked to nociceptive pathways, including channels (eg, transient receptor potential cation channel and voltage-gated calcium channel), proteins involved in neurotransmitter release (eg, synaptophysin and collapsin response mediator protein 2), and receptors (eg, glutamate, purinergic, and opioid). By altering the phosphoregulatory "set point" of proteins involved in pain signaling, Cdk5 thus appears to be an attractive target for treating neuropathic pain conditions.
Collapse
|
13
|
Khayachi A, Schorova L, Alda M, Rouleau GA, Milnerwood AJ. Posttranslational modifications & lithium's therapeutic effect-Potential biomarkers for clinical responses in psychiatric & neurodegenerative disorders. Neurosci Biobehav Rev 2021; 127:424-445. [PMID: 33971223 DOI: 10.1016/j.neubiorev.2021.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/14/2021] [Accepted: 05/03/2021] [Indexed: 01/03/2023]
Abstract
Several neurodegenerative diseases and neuropsychiatric disorders display aberrant posttranslational modifications (PTMs) of one, or many, proteins. Lithium treatment has been used for mood stabilization for many decades, and is highly effective for large subsets of patients with diverse neurological conditions. However, the differential effectiveness and mode of action are not fully understood. In recent years, studies have shown that lithium alters several protein PTMs, altering their function, and consequently neuronal physiology. The impetus for this review is to outline the links between lithium's therapeutic mode of action and PTM homeostasis. We first provide an overview of the principal PTMs affected by lithium. We then describe several neuropsychiatric disorders in which PTMs have been implicated as pathogenic. For each of these conditions, we discuss lithium's clinical use and explore the putative mechanism of how it restores PTM homeostasis, and thereby cellular physiology. Evidence suggests that determining specific PTM patterns could be a promising strategy to develop biomarkers for disease and lithium responsiveness.
Collapse
Affiliation(s)
- A Khayachi
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada.
| | - L Schorova
- McGill University Health Center Research Institute, Montréal, Quebec, Canada
| | - M Alda
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - G A Rouleau
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada; Department of Human Genetics, McGill University, Montréal, Quebec, Canada.
| | - A J Milnerwood
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada.
| |
Collapse
|
14
|
Cuveillier C, Boulan B, Ravanello C, Denarier E, Deloulme JC, Gory-Fauré S, Delphin C, Bosc C, Arnal I, Andrieux A. Beyond Neuronal Microtubule Stabilization: MAP6 and CRMPS, Two Converging Stories. Front Mol Neurosci 2021; 14:665693. [PMID: 34025352 PMCID: PMC8131560 DOI: 10.3389/fnmol.2021.665693] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/09/2021] [Indexed: 12/21/2022] Open
Abstract
The development and function of the central nervous system rely on the microtubule (MT) and actin cytoskeletons and their respective effectors. Although the structural role of the cytoskeleton has long been acknowledged in neuronal morphology and activity, it was recently recognized to play the role of a signaling platform. Following this recognition, research into Microtubule Associated Proteins (MAPs) diversified. Indeed, historically, structural MAPs—including MAP1B, MAP2, Tau, and MAP6 (also known as STOP);—were identified and described as MT-binding and -stabilizing proteins. Extensive data obtained over the last 20 years indicated that these structural MAPs could also contribute to a variety of other molecular roles. Among multi-role MAPs, MAP6 provides a striking example illustrating the diverse molecular and cellular properties of MAPs and showing how their functional versatility contributes to the central nervous system. In this review, in addition to MAP6’s effect on microtubules, we describe its impact on the actin cytoskeleton, on neuroreceptor homeostasis, and its involvement in signaling pathways governing neuron development and maturation. We also discuss its roles in synaptic plasticity, brain connectivity, and cognitive abilities, as well as the potential relationships between the integrated brain functions of MAP6 and its molecular activities. In parallel, the Collapsin Response Mediator Proteins (CRMPs) are presented as examples of how other proteins, not initially identified as MAPs, fall into the broader MAP family. These proteins bind MTs as well as exhibiting molecular and cellular properties very similar to MAP6. Finally, we briefly summarize the multiple similarities between other classical structural MAPs and MAP6 or CRMPs.In summary, this review revisits the molecular properties and the cellular and neuronal roles of the classical MAPs, broadening our definition of what constitutes a MAP.
Collapse
|
15
|
Łukasik P, Załuski M, Gutowska I. Cyclin-Dependent Kinases (CDK) and Their Role in Diseases Development-Review. Int J Mol Sci 2021; 22:ijms22062935. [PMID: 33805800 PMCID: PMC7998717 DOI: 10.3390/ijms22062935] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/07/2021] [Accepted: 03/09/2021] [Indexed: 12/13/2022] Open
Abstract
Cyclin-dependent kinases (CDKs) are involved in many crucial processes, such as cell cycle and transcription, as well as communication, metabolism, and apoptosis. The kinases are organized in a pathway to ensure that, during cell division, each cell accurately replicates its DNA, and ensure its segregation equally between the two daughter cells. Deregulation of any of the stages of the cell cycle or transcription leads to apoptosis but, if uncorrected, can result in a series of diseases, such as cancer, neurodegenerative diseases (Alzheimer’s or Parkinson’s disease), and stroke. This review presents the current state of knowledge about the characteristics of cyclin-dependent kinases as potential pharmacological targets.
Collapse
Affiliation(s)
- Paweł Łukasik
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstancow Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Michał Załuski
- Department of Pharmaceutical Chemistry, Pomeranian Medical University in Szczecin, Powstancow Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstancow Wlkp. 72 Av., 70-111 Szczecin, Poland;
- Correspondence:
| |
Collapse
|
16
|
A Review on Potential Footprints of Ferulic Acid for Treatment of Neurological Disorders. Neurochem Res 2021; 46:1043-1057. [PMID: 33547615 DOI: 10.1007/s11064-021-03257-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 01/16/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023]
Abstract
Ferulic acid is being screened in preclinical settings to combat various neurological disorders. It is a naturally occurring dietary flavonoid commonly found in grains, fruits, and vegetables such as rice, wheat, oats, tomatoes, sweet corn etc., which exhibits protective effects against a number of neurological diseases such as epilepsy, depression, ischemia-reperfusion injury, Alzheimer's disease, and Parkinson's disease. Ferulic acid prevents and treats different neurological diseases pertaining to its potent anti-oxidative and anti-inflammatory effects, beside modulating unique neuro-signaling pathways. It stays in the bloodstream for longer periods than other dietary polyphenols and antioxidants and easily crosses blood brain barrier. The use of novel drug delivery systems such as solid-lipid nanoparticles (SLNs) or its salt forms (sodium ferulate, ethyl ferulate, and isopentyl ferulate) further enhance its bioavailability and cerebral penetration. Based on reported studies, ferulic acid appears to be a promising molecule for treatment of neurological disorders; however, more preclinical (in vitro and in vivo) mechanism-based studies should be planned and conceived followed by its testing in clinical settings.
Collapse
|
17
|
Paik S, Somvanshi RK, Oliveira HA, Zou S, Kumar U. Somatostatin Ameliorates β-Amyloid-Induced Cytotoxicity via the Regulation of CRMP2 Phosphorylation and Calcium Homeostasis in SH-SY5Y Cells. Biomedicines 2021; 9:biomedicines9010027. [PMID: 33401710 PMCID: PMC7823260 DOI: 10.3390/biomedicines9010027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/24/2020] [Accepted: 12/25/2020] [Indexed: 11/17/2022] Open
Abstract
Somatostatin is involved in the regulation of multiple signaling pathways and affords neuroprotection in response to neurotoxins. In the present study, we investigated the role of Somatostatin-14 (SST) in cell viability and the regulation of phosphorylation of Collapsin Response Mediator Protein 2 (CRMP2) (Ser522) via the blockade of Ca2+ accumulation, along with the inhibition of cyclin-dependent kinase 5 (CDK5) and Calpain activation in differentiated SH-SY5Y cells. Cell Viability and Caspase 3/7 assays suggest that the presence of SST ameliorates mitochondrial stability and cell survival pathways while augmenting pro-apoptotic pathways activated by Aβ. SST inhibits the phosphorylation of CRMP2 at Ser522 site, which is primarily activated by CDK5. Furthermore, SST effectively regulates Ca2+ influx in the presence of Aβ, directly affecting the activity of calpain in differentiated SH-SY5Y cells. We also demonstrated that SSTR2 mediates the protective effects of SST. In conclusion, our results highlight the regulatory role of SST in intracellular Ca2+ homeostasis. The neuroprotective role of SST via axonal regeneration and synaptic integrity is corroborated by regulating changes in CRMP2; however, SST-mediated changes in the blockade of Ca2+ influx, calpain expression, and toxicity did not correlate with CDK5 expression and p35/25 accumulation. To summarize, our findings suggest two independent mechanisms by which SST mediates neuroprotection and confirms the therapeutic implications of SST in AD as well as in other neurodegenerative diseases where the effective regulation of calcium homeostasis is required for a better prognosis.
Collapse
Affiliation(s)
| | | | | | | | - Ujendra Kumar
- Correspondence: ; Tel.: +1-604-827-3660; Fax: +1-604-822-3035
| |
Collapse
|
18
|
Ouyang L, Chen Y, Wang Y, Chen Y, Fu AKY, Fu WY, Ip NY. p39-associated Cdk5 activity regulates dendritic morphogenesis. Sci Rep 2020; 10:18746. [PMID: 33127972 PMCID: PMC7603351 DOI: 10.1038/s41598-020-75264-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Dendrites, branched structures extending from neuronal cell soma, are specialized for processing information from other neurons. The morphogenesis of dendritic structures is spatiotemporally regulated by well-orchestrated signaling cascades. Dysregulation of these processes impacts the wiring of neuronal circuit and efficacy of neurotransmission, which contribute to the pathogeneses of neurological disorders. While Cdk5 (cyclin-dependent kinase 5) plays a critical role in neuronal dendritic development, its underlying molecular control is not fully understood. In this study, we show that p39, one of the two neuronal Cdk5 activators, is a key regulator of dendritic morphogenesis. Pyramidal neurons deficient in p39 exhibit aberrant dendritic morphology characterized by shorter length and reduced arborization, which is comparable to dendrites in Cdk5-deficient neurons. RNA sequencing analysis shows that the adaptor protein, WDFY1 (WD repeat and FYVE domain-containing 1), acts downstream of Cdk5/p39 to regulate dendritic morphogenesis. While WDFY1 is elevated in p39-deficient neurons, suppressing its expression rescues the impaired dendritic arborization. Further phosphoproteomic analysis suggests that Cdk5/p39 mediates dendritic morphogenesis by modulating various downstream signaling pathways, including PI3K/Akt-, cAMP-, or small GTPase-mediated signaling transduction pathways, thereby regulating cytoskeletal organization, protein synthesis, and protein trafficking.
Collapse
Affiliation(s)
- Li Ouyang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Yu Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China.,The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, Guangdong, China.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, 518057, Guangdong, China
| | - Ye Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Yuewen Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China.,The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, Guangdong, China.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, 518057, Guangdong, China
| | - Amy K Y Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, 518057, Guangdong, China
| | - Wing-Yu Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Nancy Y Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China. .,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China. .,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, 518057, Guangdong, China.
| |
Collapse
|
19
|
Nakamura F, Ohshima T, Goshima Y. Collapsin Response Mediator Proteins: Their Biological Functions and Pathophysiology in Neuronal Development and Regeneration. Front Cell Neurosci 2020; 14:188. [PMID: 32655376 PMCID: PMC7325199 DOI: 10.3389/fncel.2020.00188] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/29/2020] [Indexed: 12/19/2022] Open
Abstract
Collapsin response mediator proteins (CRMPs), which consist of five homologous cytosolic proteins, are one of the major phosphoproteins in the developing nervous system. The prominent feature of the CRMP family proteins is a new class of microtubule-associated proteins that play important roles in the whole process of developing the nervous system, such as axon guidance, synapse maturation, cell migration, and even in adult brain function. The CRMP C-terminal region is subjected to posttranslational modifications such as phosphorylation, which, in turn, regulates the interaction between the CRMPs and various kinds of proteins including receptors, ion channels, cytoskeletal proteins, and motor proteins. The gene-knockout of the CRMP family proteins produces different phenotypes, thereby showing distinct roles of all CRMP family proteins. Also, the phenotypic analysis of a non-phosphorylated form of CRMP2-knockin mouse model, and studies of pharmacological responses to CRMP-related drugs suggest that the phosphorylation/dephosphorylation process plays a pivotal role in pathophysiology in neuronal development, regeneration, and neurodegenerative disorders, thus showing CRMPs as promising target molecules for therapeutic intervention.
Collapse
Affiliation(s)
- Fumio Nakamura
- Department of Biochemistry, Tokyo Women's Medical University, Tokyo, Japan
| | - Toshio Ohshima
- Department of Life Science and Medical Bio-Science, Waseda University, Tokyo, Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
20
|
Fyn Tyrosine Kinase as Harmonizing Factor in Neuronal Functions and Dysfunctions. Int J Mol Sci 2020; 21:ijms21124444. [PMID: 32580508 PMCID: PMC7352836 DOI: 10.3390/ijms21124444] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 12/25/2022] Open
Abstract
Fyn is a non-receptor or cytoplasmatic tyrosine kinase (TK) belonging to the Src family kinases (SFKs) involved in multiple transduction pathways in the central nervous system (CNS) including synaptic transmission, myelination, axon guidance, and oligodendrocyte formation. Almost one hundred years after the original description of Fyn, this protein continues to attract extreme interest because of its multiplicity of actions in the molecular signaling pathways underlying neurodevelopmental as well as neuropathologic events. This review highlights and summarizes the most relevant recent findings pertinent to the role that Fyn exerts in the brain, emphasizing aspects related to neurodevelopment and synaptic plasticity. Fyn is a common factor in healthy and diseased brains that targets different proteins and shapes different transduction signals according to the neurological conditions. We will primarily focus on Fyn-mediated signaling pathways involved in neuronal differentiation and plasticity that have been subjected to considerable attention lately, opening the fascinating scenario to target Fyn TK for the development of potential therapeutic interventions for the treatment of CNS injuries and certain neurodegenerative disorders like Alzheimer’s disease.
Collapse
|
21
|
Allnutt AB, Waters AK, Kesari S, Yenugonda VM. Physiological and Pathological Roles of Cdk5: Potential Directions for Therapeutic Targeting in Neurodegenerative Disease. ACS Chem Neurosci 2020; 11:1218-1230. [PMID: 32286796 DOI: 10.1021/acschemneuro.0c00096] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cyclin-dependent kinase 5 (Cdk5) is a proline-directed serine (ser)/threonine (Thr) kinase that has been demonstrated to be one of the most functionally diverse kinases within neurons. Cdk5 is regulated via binding with its neuron-specific regulatory subunits, p35 or p39. Cdk5-p35 activity is critical for a variety of developmental and cellular processes in the brain, including neuron migration, memory formation, microtubule regulation, and cell cycle suppression. Aberrant activation of Cdk5 via the truncated p35 byproduct, p25, is implicated in the pathogenesis of several neurodegenerative diseases. The present review highlights the importance of Cdk5 activity and function in the brain and demonstrates how deregulation of Cdk5 can contribute to the development of neurodegenerative conditions such as Alzheimer's and Parkinson's disease. Additionally, we cover past drug discovery attempts at inhibiting Cdk5-p25 activity and discuss which types of targeting strategies may prove to be the most successful moving forward.
Collapse
|
22
|
Zhao WN, Tobe BTD, Udeshi ND, Xuan LL, Pernia CD, Zolg DP, Roberts AJ, Mani D, Blumenthal SR, Kurtser I, Patnaik D, Gaisina I, Bishop J, Sheridan SD, Lalonde J, Carr SA, Snyder EY, Haggarty SJ. Discovery of suppressors of CRMP2 phosphorylation reveals compounds that mimic the behavioral effects of lithium on amphetamine-induced hyperlocomotion. Transl Psychiatry 2020; 10:76. [PMID: 32094324 PMCID: PMC7039883 DOI: 10.1038/s41398-020-0753-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 11/08/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022] Open
Abstract
The effective treatment of bipolar disorder (BD) represents a significant unmet medical need. Although lithium remains a mainstay of treatment for BD, limited knowledge regarding how it modulates affective behavior has proven an obstacle to discovering more effective mood stabilizers with fewer adverse side effects. One potential mechanism of action of lithium is through inhibition of the serine/threonine protein kinase GSK3β, however, relevant substrates whose change in phosphorylation may mediate downstream changes in neuroplasticity remain poorly understood. Here, we used human induced pluripotent stem cell (hiPSC)-derived neuronal cells and stable isotope labeling by amino acids in cell culture (SILAC) along with quantitative mass spectrometry to identify global changes in the phosphoproteome upon inhibition of GSK3α/β with the highly selective, ATP-competitive inhibitor CHIR-99021. Comparison of phosphorylation changes to those induced by therapeutically relevant doses of lithium treatment led to the identification of collapsin response mediator protein 2 (CRMP2) as being highly sensitive to both treatments as well as an extended panel of structurally distinct GSK3α/β inhibitors. On this basis, a high-content image-based assay in hiPSC-derived neurons was developed to screen diverse compounds, including FDA-approved drugs, for their ability to mimic lithium's suppression of CRMP2 phosphorylation without directly inhibiting GSK3β kinase activity. Systemic administration of a subset of these CRMP2-phosphorylation suppressors were found to mimic lithium's attenuation of amphetamine-induced hyperlocomotion in mice. Taken together, these studies not only provide insights into the neural substrates regulated by lithium, but also provide novel human neuronal assays for supporting the development of mechanism-based therapeutics for BD and related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Wen-Ning Zhao
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA
| | - Brian T. D. Tobe
- grid.479509.60000 0001 0163 8573Center for Stem Cells & Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA ,grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA 92037 USA ,grid.468218.1Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037 USA ,Present Address: Kaiser Health, San Diego, CA USA
| | - Namrata D. Udeshi
- grid.38142.3c000000041936754XProteomics Platform, Broad Institute of MIT and Harvard University, Cambridge, MA 02142 USA
| | - Lucius L. Xuan
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA
| | - Cameron D. Pernia
- grid.479509.60000 0001 0163 8573Center for Stem Cells & Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA ,grid.468218.1Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037 USA
| | - Daniel P. Zolg
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA ,grid.6936.a0000000123222966Present Address: TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Amanda J. Roberts
- grid.468218.1Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037 USA
| | - Deepak Mani
- grid.38142.3c000000041936754XProteomics Platform, Broad Institute of MIT and Harvard University, Cambridge, MA 02142 USA
| | - Sarah R. Blumenthal
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA
| | - Iren Kurtser
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA
| | - Debasis Patnaik
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA
| | - Irina Gaisina
- grid.185648.60000 0001 2175 0319Department of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Joshua Bishop
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA ,grid.417993.10000 0001 2260 0793Present Address: Merck, Boston, MA USA
| | - Steven D. Sheridan
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA
| | - Jasmin Lalonde
- grid.34429.380000 0004 1936 8198Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Road, East, Guelph, ON Canada N1G 2W1
| | - Steven A. Carr
- grid.38142.3c000000041936754XProteomics Platform, Broad Institute of MIT and Harvard University, Cambridge, MA 02142 USA
| | - Evan Y. Snyder
- grid.479509.60000 0001 0163 8573Center for Stem Cells & Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA ,grid.468218.1Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037 USA ,grid.266100.30000 0001 2107 4242Department of Pediatrics, University of California San Diego, La Jolla, CA 92037 USA
| | - Stephen J. Haggarty
- grid.32224.350000 0004 0386 9924Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA ,grid.32224.350000 0004 0386 9924Departments of Psychiatry & Neurology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114 USA
| |
Collapse
|
23
|
Low Incidence of High-Grade Pancreatic Intraepithelial Neoplasia Lesions in a Crmp4 Gene-Deficient Mouse Model of Pancreatic Cancer. Transl Oncol 2020; 13:100746. [PMID: 32105991 PMCID: PMC7044544 DOI: 10.1016/j.tranon.2020.100746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 12/24/2022] Open
Abstract
Pancreatic intraepithelial neoplasia (PanIN), the most common premalignant lesion of the pancreas, is a histologically well-defined precursor to invasive pancreatic ductal adenocarcinoma (PDAC). However, the molecular mechanisms underlying the progression of PanINs have not been fully elucidated. Previously, we demonstrated that the expression of collapsin response mediator protein 4 (CRMP4) in PDAC was associated with poor prognosis. The expression of CRMP4 was also augmented in a pancreatitis mouse model. However, the role of CRMP4 in the progression of PanIN lesions remains uncertain. In the present study, we examined the relationship between CRMP4 expression and progression of PanIN lesions using genetically engineered mouse models. PanIN lesions were induced by peritoneal injection of the cholecystokinin analog caerulein in LSL-KRASG12D; Pdx1-Cre (KC-Crmp4 wild-type, WT) mice and LSL-KRASG12D; Pdx1-Cre; Crmp4−/− (KC-Crmp4 knockout, KO) mice. We analyzed pancreatic tissue sections from these mice and evaluated PanIN grade by hematoxylin and eosin staining. CRMP4 expression was examined and the cellular components assessed by immunohistochemistry using antibodies against CRMP4, CD3, and α-smooth muscle actin (SMA). The incidence of high-grade PanIN in KC-Crmp4 WT mice was higher than that in KC-Crmp4 KO animals. CRMP4 was expressed not only in epithelial cells but also in αSMA-positive cells in stromal areas of PanIN lesions. The CRMP4 expression in stromal areas correlated with PanIN grade in WT mice. These results suggested that the expression of CRMP4 in stromal cells may underlie the incidence or progression of PanIN.
Collapse
|
24
|
Ziak J, Weissova R, Jeřábková K, Janikova M, Maimon R, Petrasek T, Pukajova B, Kleisnerova M, Wang M, Brill MS, Kasparek P, Zhou X, Alvarez-Bolado G, Sedlacek R, Misgeld T, Stuchlik A, Perlson E, Balastik M. CRMP2 mediates Sema3F-dependent axon pruning and dendritic spine remodeling. EMBO Rep 2020; 21:e48512. [PMID: 31919978 DOI: 10.15252/embr.201948512] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 11/09/2022] Open
Abstract
Regulation of axon guidance and pruning of inappropriate synapses by class 3 semaphorins are key to the development of neural circuits. Collapsin response mediator protein 2 (CRMP2) has been shown to regulate axon guidance by mediating semaphorin 3A (Sema3A) signaling; however, nothing is known about its role in synapse pruning. Here, using newly generated crmp2-/- mice we demonstrate that CRMP2 has a moderate effect on Sema3A-dependent axon guidance in vivo, and its deficiency leads to a mild defect in axon guidance in peripheral nerves and the corpus callosum. Surprisingly, crmp2-/- mice display prominent defects in stereotyped axon pruning in hippocampus and visual cortex and altered dendritic spine remodeling, which is consistent with impaired Sema3F signaling and with models of autism spectrum disorder (ASD). We demonstrate that CRMP2 mediates Sema3F signaling in primary neurons and that crmp2-/- mice display ASD-related social behavior changes in the early postnatal period as well as in adults. Together, we demonstrate that CRMP2 mediates Sema3F-dependent synapse pruning and its dysfunction shares histological and behavioral features of ASD.
Collapse
Affiliation(s)
- Jakub Ziak
- Department of Molecular Neurobiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.,Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Romana Weissova
- Department of Molecular Neurobiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.,Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Kateřina Jeřábková
- Department of Transgenic Models of Diseases and Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martina Janikova
- Department of Neurophysiology of the Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Roy Maimon
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Tomas Petrasek
- Department of Neurophysiology of the Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Barbora Pukajova
- Department of Molecular Neurobiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.,Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Marie Kleisnerova
- Department of Molecular Neurobiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Mengzhe Wang
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Monika S Brill
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Petr Kasparek
- Department of Transgenic Models of Diseases and Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Xunlei Zhou
- Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | | | - Radislav Sedlacek
- Department of Transgenic Models of Diseases and Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.,German Center for Neurodegenerative Diseases and Munich Cluster for Systems Neurology, Munich, Germany
| | - Ales Stuchlik
- Department of Neurophysiology of the Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Eran Perlson
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Martin Balastik
- Department of Molecular Neurobiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
25
|
Moutal A, White KA, Chefdeville A, Laufmann RN, Vitiello PF, Feinstein D, Weimer JM, Khanna R. Dysregulation of CRMP2 Post-Translational Modifications Drive Its Pathological Functions. Mol Neurobiol 2019; 56:6736-6755. [PMID: 30915713 PMCID: PMC6728212 DOI: 10.1007/s12035-019-1568-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/15/2019] [Indexed: 12/13/2022]
Abstract
Collapsin response mediator proteins (CRMPs) are a family of ubiquitously expressed, homologous phosphoproteins best known for coordinating cytoskeletal formation and regulating cellular division, migration, polarity, and synaptic connection. CRMP2, the most studied of the five family members, is best known for its affinity for tubulin heterodimers and function in regulating the microtubule network. These functions are tightly regulated by post-translational modifications including phosphorylation, SUMOylation, oxidation, and O-GlcNAcylation. While CRMP2's physiological functions rely mostly on its non-phosphorylated state, dysregulation of CRMP2 phosphorylation and SUMOylation has been reported to be involved in the pathophysiology of multiple diseases including cancer, chronic pain, spinal cord injury, neurofibromatosis type 1, and others. Here, we provide a consolidated update on what is known about CRMP2 signaling and function, first focusing on axonal growth and neuronal polarity, then illustrating the link between dysregulated CRMP2 post-translational modifications and diseases. We additionally discuss the roles of CRMP2 in non-neuronal cells, both in the CNS and regions of the periphery. Finally, we offer thoughts on the therapeutic implications of modulating CRMP2 function in a variety of diseases.
Collapse
Affiliation(s)
- Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Katherine A White
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 E 60th St N, Sioux Falls, SD, 57104, USA
| | - Aude Chefdeville
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Rachel N Laufmann
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 E 60th St N, Sioux Falls, SD, 57104, USA
| | - Peter F Vitiello
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| | - Douglas Feinstein
- Department of Veterans Affairs, Jesse Brown VA Medical Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Jill M Weimer
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA.
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA.
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA.
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 E 60th St N, Sioux Falls, SD, 57104, USA.
- Department of Anesthesiology, University of Arizona, Tucson, AZ, USA.
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ, USA.
| |
Collapse
|
26
|
Migita K, Matsumoto T, Terada K, Ono K, Hara S. Effects of geldanamycin on neurite outgrowth-related proteins and kinases in nerve growth factor-differentiated pheochromocytoma 12 cells. J Pharmacol Sci 2019; 140:255-262. [PMID: 31402210 DOI: 10.1016/j.jphs.2019.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/19/2019] [Accepted: 07/23/2019] [Indexed: 01/14/2023] Open
Abstract
Heat shock protein 90 (HSP90) antagonists are currently being evaluated as potential anticancer drugs. However, adverse effects related to these drugs, such as fatigue and pain, suggest that they affect neurons. Therefore, to understand the influence of HSP90 inhibitors on neurons, we investigated the effects of geldanamycin, an HSP90 antagonist, on nerve growth factor (NGF)-differentiated pheochromocytoma 12 (PC12) cells, particularly, on the expression and phosphorylation of proteins and kinases in the NGF pathway. Geldanamycin significantly inhibited NGF-induced neurite outgrowth and phosphorylation of Akt and extracellular signal-related kinase 1/2 in PC12 cells. Furthermore, geldanamycin inhibited the phosphorylation of collapsin response mediator protein 2 and the expression of cyclin-dependent kinase 5 in the presence of NGF, but did not significantly affect the expression of glycogen synthase kinase 3β. These results suggest that geldanamycin influences microtubule-binding proteins and kinases relating to neurite outgrowth, thereby inducing neuronal impairment.
Collapse
Affiliation(s)
- Keisuke Migita
- Department of Drug Informatics and Translational Research, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan.
| | - Taichi Matsumoto
- Department of Drug Informatics and Translational Research, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Kazuki Terada
- Laboratory of Drug Design and Drug Delivery, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan
| | - Kazuhiko Ono
- Department of Drug Informatics and Translational Research, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Shuji Hara
- Department of Drug Informatics and Translational Research, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
27
|
Kim Y, Jeon S, Jeong HJ, Lee SM, Peña ID, Kim HJ, Han DH, Kim BN, Cheong JH. Restoration of Cdk5, TrkB and Soluble N-ethylmaleimide-Sensitive Factor Attachment Protein Receptor Proteins after Chronic Methylphenidate Treatment in Spontaneous Hypertensive Rats, a Model for Attention-Deficit Hyperactivity Disorder. Psychiatry Investig 2019; 16:558-564. [PMID: 31352739 PMCID: PMC6664221 DOI: 10.30773/pi.2019.04.22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/22/2019] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE Synaptic vesicle mobilization and neurite outgrowth regulation molecules were examined in modulation of effects of methylphenidate (MPH) in Spontaneous Hypertensive Rats (SHRs), a model for attention-deficit hyperactivity disorder (ADHD). METHODS We compared the changes in the protein expression level of Cyclin dependent kinase 5 (Cdk5) and molecular substrates of Cdk5; tropomyosin receptor kinase B (TrkB), syntaxin 1A (STX1A) and synaptosomal-associated protein 25 (SNAP25). Comparisons were made in prefrontal cortex of vehicle (distilled water i.p. for 7 days)-treated SHRs, vehicle-treated Wistar Kyoto Rats (WKYs) and MPH (2 mg/kg i.p. for 7 days) treated SHRs. RESULTS The Cdk5 level of vehicle-treated SHRs was significantly decreased compared to the Cdk5 level of vehicle-treated WKY rats, but was restored to the expression level of vehicle-treated WKYs in MPH-treated SHR. The ratio of p25/p35 was significantly decreased in MPH-treated SHR compared to vehicle-treated SHR. Moreover, TrkB, STX1A and SNAP25 of vehicle-treated SHRs were significantly decreased compared to vehicle-treated WKY rats, but were restored to the expression level of vehicle-treated WKYs in MPH-treated SHR. CONCLUSION The results show that Cdk5, TrkB, STX1A, and SNAP25 were involved in the modulation of MPH effects in prefrontal cortex of SHRs and play important role in treatment of ADHD.
Collapse
Affiliation(s)
- Yeni Kim
- Department of Child and Adolescent Psychiatry, National Center for Mental Health, Seoul, Republic of Korea
| | - Songhee Jeon
- Department of Biomedical Sciences, Center for Creative Biomedical Scientists at Chonnam National University, Gwangju, Republic of Korea
| | - Ha Jin Jeong
- Department of Biomedical Sciences, Center for Creative Biomedical Scientists at Chonnam National University, Gwangju, Republic of Korea
| | - Seong Mi Lee
- Department of Child and Adolescent Psychiatry, National Center for Mental Health, Seoul, Republic of Korea
| | - Ike Dela Peña
- Department of Pharmacy, Sahmyook University, Uimyung Research Institute for Neuroscience, Seoul, Republic of Korea.,Department of Pharmaceutical and Administrative Sciences, Loma Linda University Health, Loma Linda, CA, USA
| | - Hee Jin Kim
- Department of Pharmacy, Sahmyook University, Uimyung Research Institute for Neuroscience, Seoul, Republic of Korea
| | - Doug Hyun Han
- Department of Psychiatry, Chung Ang University Hospital, Seoul, Republic of Korea
| | - Bung-Nyun Kim
- Division of Child & Adolescent Psychiatry, Department of Psychiatry and Institute of Human Behavioral Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jae Hoon Cheong
- Department of Pharmacy, Sahmyook University, Uimyung Research Institute for Neuroscience, Seoul, Republic of Korea
| |
Collapse
|
28
|
Wang X, Zhang W, Li J, Yu M, Dong M, Meng H. Collapsin Response Mediator Protein 2, a Potential Therapeutic Target in Temporal Lobe Epilepsy. NEUROCHEM J+ 2019. [DOI: 10.1134/s1819712419020144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
29
|
Bott CJ, Johnson CG, Yap CC, Dwyer ND, Litwa KA, Winckler B. Nestin in immature embryonic neurons affects axon growth cone morphology and Semaphorin3a sensitivity. Mol Biol Cell 2019; 30:1214-1229. [PMID: 30840538 PMCID: PMC6724523 DOI: 10.1091/mbc.e18-06-0361] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 02/21/2019] [Accepted: 02/26/2019] [Indexed: 12/14/2022] Open
Abstract
Correct wiring in the neocortex requires that responses to an individual guidance cue vary among neurons in the same location, and within the same neuron over time. Nestin is an atypical intermediate filament expressed strongly in neural progenitors and is thus used widely as a progenitor marker. Here we show a subpopulation of embryonic cortical neurons that transiently express nestin in their axons. Nestin expression is thus not restricted to neural progenitors, but persists for 2-3 d at lower levels in newborn neurons. We found that nestin-expressing neurons have smaller growth cones, suggesting that nestin affects cytoskeletal dynamics. Nestin, unlike other intermediate filament subtypes, regulates cdk5 kinase by binding the cdk5 activator p35. Cdk5 activity is induced by the repulsive guidance cue Semaphorin3a (Sema3a), leading to axonal growth cone collapse in vitro. Therefore, we tested whether nestin-expressing neurons showed altered responses to Sema3a. We find that nestin-expressing newborn neurons are more sensitive to Sema3a in a roscovitine-sensitive manner, whereas nestin knockdown results in lowered sensitivity to Sema3a. We propose that nestin functions in immature neurons to modulate cdk5 downstream of the Sema3a response. Thus, the transient expression of nestin could allow temporal and/or spatial modulation of a neuron's response to Sema3a, particularly during early axon guidance.
Collapse
Affiliation(s)
- C. J. Bott
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908
| | - C. G. Johnson
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834
| | - C. C. Yap
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908
| | - N. D. Dwyer
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908
| | - K. A. Litwa
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834
| | - B. Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
30
|
Numata-Uematsu Y, Wakatsuki S, Nagano S, Shibata M, Sakai K, Ichinohe N, Mikoshiba K, Ohshima T, Yamashita N, Goshima Y, Araki T. Inhibition of collapsin response mediator protein-2 phosphorylation ameliorates motor phenotype of ALS model mice expressing SOD1G93A. Neurosci Res 2018; 139:63-68. [PMID: 30194029 DOI: 10.1016/j.neures.2018.08.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/22/2018] [Accepted: 08/30/2018] [Indexed: 10/28/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset neurological disease characterized by the selective degeneration of motor neurons leading to paralysis and immobility. Missense mutations in the gene coding for the Cu2+/Zn2+ superoxide dismutase 1 (SOD1) accounts for 15-20% of familial ALS, and mice overexpressing ALS-linked SOD1 mutants have been frequently used as an animal model for ALS. Degeneration of motor neurons in ALS progresses in a manner called "dying back", in which the degeneration of synapses and axons precedes the loss of cell bodies. Phosphorylation of collapsin response mediator protein 2 (CRMP2) is implicated in the progression of neuronal/axonal degeneration of different etiologies. To evaluate the role of CRMP2 phosphorylation in ALS pathogenesis, we utilized CRMP2 S522A knock-in (CRMP2ki/ki) mice, in which the serine residue 522 was homozygously replaced with alanine and thereby making CRMP2 no longer phosphorylatable by CDK5 or GSK3B. We found that the CRMP2ki/ki/SOD1G93A mice showed delay in the progression of the motor phenotype compared to their SOD1G93-Tg littermates. Histological analysis revealed that the CRMP2ki/ki/SOD1G93A mice retained more intact axons and NMJs than their SOD1G93A-Tg littermates. These results suggest that the phosphorylation of CRMP2 may contribute to the axonal degeneration of motor neurons in ALS.
Collapse
Affiliation(s)
- Yurika Numata-Uematsu
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Japan
| | - Shuji Wakatsuki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Japan
| | - Seiichi Nagano
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Japan; Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Megumi Shibata
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Japan
| | - Kazuhisa Sakai
- Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Japan
| | - Noritaka Ichinohe
- Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Japan
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Center for Brain Science, Japan
| | - Toshio Ohshima
- Laboratory for Molecular Brain Science, Department of Life Science and Medical Bioscience, Waseda University, Japan
| | - Naoya Yamashita
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Japan; Department of Pharmacology, Juntendo University School of Medicine, Japan
| | - Yoshiro Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Japan
| | - Toshiyuki Araki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Japan.
| |
Collapse
|
31
|
Li XB, Ding MX, Ding CL, Li LL, Feng J, Yu XJ. Toll‑Like receptor 4 promotes the phosphorylation of CRMP2 via the activation of Rho‑kinase in MCAO rats. Mol Med Rep 2018; 18:342-348. [PMID: 29749502 PMCID: PMC6059689 DOI: 10.3892/mmr.2018.8968] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 04/20/2018] [Indexed: 02/05/2023] Open
Abstract
The mechanism associated with Toll‑like receptor 4 (TLR4) in neurological injury remains unclear. The aim of the present study was to investigate the pathology of TLR4 in middle cerebral artery occlusion (MCAO)/reperfusion rat models via the regulation of collapsin response mediator protein 2 (CRMP2) phosphorylation. The modified neurological severity score (mNSS) was applied to assess neurological recovery. Immunofluorescence and western blotting were used to detect the protein expressions of TLR4, Rho‑associated protein kinase 2 (ROCK‑II) and CRMP2 following the intracerebroventricular administration of TLR4‑specific agonist, lipopolysaccharide (LPS) and TLR4‑neutralizing antibody, the ROCK‑II specific inhibitor Y‑27632 or LPS+Y‑27632 30 min prior to MCAO. The expression levels of TLR4 and the phosphorylation of CRMP2 significantly increased in response to LPS‑mediated induction and/or MCAO; however, they were reversed by treatment with LPS+TLR4‑neutralizing antibody. Y‑27632 decreased the expression of ROCK‑II and phosphorylated (p)‑CRMP2, and suppressed the increased ROCK‑II and p‑CRMP2 induced by LPS; however, no effect on the levels of TLR4 expression was observed. The neurological function as measured by mNSS score was reduced in the LPS group when compared with the MCAO group, whereas the LPS+Y‑27632 group reversed the reduced neurological function at 7 and 14 days post‑MCAO. The results of the present study suggested that TLR4 may promote the phosphorylation of CRMP2 via the activation of ROCK‑II in MCAO rats, which further characterizes the pathological mechanism of TLR4 in stroke, and that modulation of TLR4 could be a potential target to limit secondary post‑stroke brain damage.
Collapse
Affiliation(s)
- Xue-Bo Li
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Key Laboratory of Evidence Identification in Universities of Shandong, Shandong University of Political Science and Law, Jinan, Shandong 250014, P.R. China
| | - Ming-Xia Ding
- Department of Obstetrics and Gynecology, The Second Hospital of Shandong University, Jinan, Shandong 250014, P.R. China
| | - Chun-Li Ding
- Key Laboratory of Evidence Identification in Universities of Shandong, Shandong University of Political Science and Law, Jinan, Shandong 250014, P.R. China
| | - Liang-Liang Li
- Key Laboratory of Evidence Identification in Universities of Shandong, Shandong University of Political Science and Law, Jinan, Shandong 250014, P.R. China
| | - Jinzhou Feng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiao-Jun Yu
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Correspondence to: Dr Xiao-Jun Yu, Department of Forensic Medicine, Shantou University Medical College, 22 Xinling Road, Shantou, Guangdong 515041, P.R. China, E-mail:
| |
Collapse
|
32
|
Abstract
SIGNIFICANCE Numerous studies have demonstrated the actions of reactive oxygen species (ROS) as regulators of several physiological processes. In this study, we discuss how redox signaling mechanisms operate to control different processes such as neuronal differentiation, oligodendrocyte differentiation, dendritic growth, and axonal growth. Recent Advances: Redox homeostasis regulates the physiology of neural stem cells (NSCs). Notably, the neuronal differentiation process of NSCs is determined by a change toward oxidative metabolism, increased levels of mitochondrial ROS, increased activity of NADPH oxidase (NOX) enzymes, decreased levels of Nrf2, and differential regulation of different redoxins. Furthermore, during the neuronal maturation processes, NOX and MICAL produce ROS to regulate cytoskeletal dynamics, which control the dendritic and axonal growth, as well as the axonal guidance. CRITICAL ISSUES The redox homeostasis changes are, in part, attributed to cell metabolism and compartmentalized production of ROS, which is regulated, sensed, and transduced by different molecules such as thioredoxins, glutaredoxins, peroxiredoxins, and nucleoredoxin to control different signaling pathways in different subcellular regions. The study of how these elements cooperatively act is essential for the understanding of nervous system development, as well as the application of regenerative therapies that recapitulate these processes. FUTURE DIRECTIONS The information about these topics in the last two decades leads us to the conclusion that the role of ROS signaling in development of the nervous system is more important than it was previously believed and makes clear the importance of exploring in more detail the mechanisms of redox signaling. Antioxid. Redox Signal. 28, 1603-1625.
Collapse
Affiliation(s)
- Mauricio Olguín-Albuerne
- División de Neurociencias, Instituto de Fisiología Celular , Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Julio Morán
- División de Neurociencias, Instituto de Fisiología Celular , Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
33
|
Nirwan N, Siraj F, Vohora D. Inverted-U response of lacosamide on pilocarpine-induced status epilepticus and oxidative stress in C57BL/6 mice is independent of hippocampal collapsin response mediator protein-2. Epilepsy Res 2018; 145:93-101. [PMID: 29935443 DOI: 10.1016/j.eplepsyres.2018.06.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 06/09/2018] [Accepted: 06/15/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Currently, lacosamide (LCM) is not approved for use in status epilepticus (SE) but several shreds of evidence are available to support its use. The present study was, therefore, undertaken to evaluate the effect of LCM on pilocarpine (PILO) induced SE and neurodegeneration in C57BL/6 mice and to ascertain the involvement of CRMP-2 in mediating above effect. METHODS Pilocarpine-induced SE model was developed to explore the effect of LCM 20, 40 and 80 mg/kg in mice. We assessed the seizure severity, seizure latency, spontaneous alternation behavior (SAB) and motor coordination by behavioral observation. Histopathological evaluation and measurement of the levels of CRMP-2, reduced glutathione (GSH) and malondialdehyde (MDA) were carried out in mice hippocampus. RESULTS LCM exhibited a biphasic effect i.e., protection against SE at 20 mg/kg and 40 mg/kg dose whilst aggravated seizure-like behavior and mortality at 80 mg/kg. Further, it increased percentage alternation (i.e., restored spatial memory) in SAB and elevated motor impairment with increasing dose. Histologically, LCM 20 mg/kg and 40 mg/kg (but not 80 mg/kg) reduced neurodegeneration. LCM 20 mg/kg and 40 mg/kg reversed the elevated MDA and GSH levels while 80 mg/kg showed a tendency to increase oxidative stress. In contrast, LCM (at all doses) reversed the pilocarpine-induced elevation of collapsin response mediator protein-2 (CRMP-2). CONCLUSION LCM protected against pilocarpine-induced SE, associated neurodegeneration and improved pilocarpine-associated impairment of spatial memory. The study reveals that CRMP-2 may not be mediating the inverted-U-response of LCM at least in pilocarpine model. Therefore, the anti-oxidant effect of LCM (and not its ability to modulate CRMP-2) was anticipated as the mechanism underlying neuroprotection.
Collapse
Affiliation(s)
- Nikita Nirwan
- Neurobehavioral Pharmacology Laboratory, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Fouzia Siraj
- National Institute of Pathology (ICMR), Safdarjang Hospital Campus, New Delhi 110029, India
| | - Divya Vohora
- Neurobehavioral Pharmacology Laboratory, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
34
|
Dixit AB, Banerjee J, Tripathi M, Sarkar C, Chandra PS. Synaptic roles of cyclin-dependent kinase 5 & its implications in epilepsy. Indian J Med Res 2018. [PMID: 28639593 PMCID: PMC5501049 DOI: 10.4103/ijmr.ijmr_1249_14] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
There is an urgent need to understand the molecular mechanisms underlying epilepsy to find novel prognostic/diagnostic biomarkers to prevent epilepsy patients at risk. Cyclin-dependent kinase 5 (CDK5) is involved in multiple neuronal functions and plays a crucial role in maintaining homeostatic synaptic plasticity by regulating intracellular signalling cascades at synapses. CDK5 deregulation is shown to be associated with various neurodegenerative diseases such as Alzheimer's disease. The association between chronic loss of CDK5 and seizures has been reported in animal models of epilepsy. Genetic expression of CDK5 at transcriptome level has been shown to be abnormal in intractable epilepsy. In this review various possible mechanisms by which deregulated CDK5 may alter synaptic transmission and possibly lead to epileptogenesis have been discussed. Further, CDK5 has been proposed as a potential biomarker as well as a pharmacological target for developing treatments for epilepsy.
Collapse
Affiliation(s)
- Aparna Banerjee Dixit
- Center for Excellence in Epilepsy, A Joint National Brain Research Centre (NBRC)- All India Institute of Medical Sciences (AIIMS) Collaboration, NBRC, Gurugram, India
| | - Jyotirmoy Banerjee
- Center for Excellence in Epilepsy, A Joint National Brain Research Centre (NBRC)- All India Institute of Medical Sciences (AIIMS) Collaboration, NBRC, Gurugram, India
| | | | | | | |
Collapse
|
35
|
Zheng Y, Sethi R, Mangala LS, Taylor C, Goldsmith J, Wang M, Masuda K, Karaminejadranjbar M, Mannion D, Miranda F, Herrero-Gonzalez S, Hellner K, Chen F, Alsaadi A, Albukhari A, Fotso DC, Yau C, Jiang D, Pradeep S, Rodriguez-Aguayo C, Lopez-Berestein G, Knapp S, Gray NS, Campo L, Myers KA, Dhar S, Ferguson D, Bast RC, Sood AK, von Delft F, Ahmed AA. Tuning microtubule dynamics to enhance cancer therapy by modulating FER-mediated CRMP2 phosphorylation. Nat Commun 2018; 9:476. [PMID: 29396402 PMCID: PMC5797184 DOI: 10.1038/s41467-017-02811-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 12/27/2017] [Indexed: 12/13/2022] Open
Abstract
Though used widely in cancer therapy, paclitaxel only elicits a response in a fraction of patients. A strong determinant of paclitaxel tumor response is the state of microtubule dynamic instability. However, whether the manipulation of this physiological process can be controlled to enhance paclitaxel response has not been tested. Here, we show a previously unrecognized role of the microtubule-associated protein CRMP2 in inducing microtubule bundling through its carboxy terminus. This activity is significantly decreased when the FER tyrosine kinase phosphorylates CRMP2 at Y479 and Y499. The crystal structures of wild-type CRMP2 and CRMP2-Y479E reveal how mimicking phosphorylation prevents tetramerization of CRMP2. Depletion of FER or reducing its catalytic activity using sub-therapeutic doses of inhibitors increases paclitaxel-induced microtubule stability and cytotoxicity in ovarian cancer cells and in vivo. This work provides a rationale for inhibiting FER-mediated CRMP2 phosphorylation to enhance paclitaxel on-target activity for cancer therapy. Some anticancer drugs target cell microtubules inhibiting mitosis and cell division. Here, the authors show that CRMP2 induces microtubule bundling and that this activity is regulated by the FER kinase, thus providing a rationale for targeting FER in combination with microtubule-targeting drugs.
Collapse
Affiliation(s)
- Yiyan Zheng
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Ritika Sethi
- Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Lingegowda S Mangala
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Charlotte Taylor
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Juliet Goldsmith
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Ming Wang
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Kenta Masuda
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Mohammad Karaminejadranjbar
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - David Mannion
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Fabrizio Miranda
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Sandra Herrero-Gonzalez
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Karin Hellner
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Fiona Chen
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Abdulkhaliq Alsaadi
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Ashwag Albukhari
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK.,Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, 21551, Saudi Arabia
| | - Donatien Chedom Fotso
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Christopher Yau
- Wellcome Trust Centre for Human Genetics and NIHR Biomedical Research Centre, Roosevelt Drive, Oxford, OX3 7BN, UK.,Department of Statistics, 1 South Parks Road, Oxford, OX1 3TG, UK
| | - Dahai Jiang
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Sunila Pradeep
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Cristian Rodriguez-Aguayo
- Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel Lopez-Berestein
- Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stefan Knapp
- Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK.,Goethe-University Frankfurt, Institute for Pharmaceutical Chemistry and Buchmann Institute for Life Sciences, Riedberg Campus, Frankfurt am Main, 60438, Germany
| | - Nathanael S Gray
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA.,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Leticia Campo
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Kevin A Myers
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Sunanda Dhar
- Department of Histopathology, Oxford University Hospitals, Oxford, OX3 9DU, UK
| | - David Ferguson
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine University of Oxford, Oxford University Hospitals, Oxford, OX3 9DU, UK
| | - Robert C Bast
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Center for RNAi and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Frank von Delft
- Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK.,Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, OX11 0QX, UK.,Department of Biochemistry, University of Johannesburg, Auckland Park, 2006, South Africa
| | - Ahmed Ashour Ahmed
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK. .,Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| |
Collapse
|
36
|
Mokhtar SH, Kim MJ, Magee KA, Aui PM, Thomas S, Bakhuraysah MM, Alrehaili AA, Lee JY, Steer DL, Kenny R, McLean C, Azari MF, Birpanagos A, Lipiec E, Heraud P, Wood B, Petratos S. Amyloid-beta-dependent phosphorylation of collapsin response mediator protein-2 dissociates kinesin in Alzheimer's disease. Neural Regen Res 2018; 13:1066-1080. [PMID: 29926835 PMCID: PMC6022475 DOI: 10.4103/1673-5374.233451] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by accumulation of amyloid plaques and neurofibrillary tangles. Prior to the development of these characteristic pathological hallmarks of AD, anterograde axonal transport is impaired. However, the key proteins that initiate these intracellular impairments remain elusive. The collapsin response mediator protein-2 (CRMP-2) plays an integral role in kinesin-1-dependent axonal transport and there is evidence that phosphorylation of CRMP-2 releases kinesin-1. Here, we tested the hypothesis that amyloid-beta (Aβ)-dependent phosphorylation of CRMP-2 disrupts its association with the kinesin-1 (an anterograde axonal motor transport protein) in AD. We found that brain sections and lysates from AD patients demonstrated elevated phosphorylation of CRMP-2 at the T555 site. Additionally, in the transgenic Tg2576 mouse model of familial AD (FAD) that exhibits Aβ accumulation in the brain with age, we found substantial co-localization of pT555CRMP-2 and dystrophic neurites. In SH-SY5Y differentiated neuronal cultures, Aβ-dependent phosphorylation of CRMP-2 at the T555 site was also elevated and this reduced the CRMP-2 association with kinesin-1. The overexpression of an unphosphorylatable form of CRMP-2 in neurons promoted the re-establishment of CRMP-2-kinesin association and axon elongation. These data suggest that Aβ-dependent phosphorylation of CRMP-2 at the T555 site may directly impair anterograde axonal transport protein function, leading to neuronal defects.
Collapse
Affiliation(s)
- Sara H Mokhtar
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria, Australia
| | - Min Joung Kim
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria, Australia
| | - Kylie A Magee
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria, Australia
| | - Pei Mun Aui
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria, Australia
| | - Speros Thomas
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria, Australia
| | - Maha M Bakhuraysah
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria, Australia
| | - Amani A Alrehaili
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria, Australia
| | - Jae Young Lee
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria, Australia
| | - David L Steer
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Rachel Kenny
- Department of Anatomy & Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Catriona McLean
- Department of Anatomical Pathology, Alfred Hospital, Prahran, Victoria, Australia
| | - Michael F Azari
- Department of Anatomy & Developmental Biology, Monash University, Clayton; School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Antonis Birpanagos
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Ilisia, Athens, Greece
| | - Ewlina Lipiec
- The Henryk Niewodniczanski Institute of Nuclear Physics, Polish Academy of Sciences, Department of Applied Spectroscopy, Radzikowskiego, Krakow, Poland
| | - Philip Heraud
- Centre for Biospectroscopy and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Bayden Wood
- Centre for Biospectroscopy and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria, Australia
| |
Collapse
|
37
|
Whitman MC, Engle EC. Ocular congenital cranial dysinnervation disorders (CCDDs): insights into axon growth and guidance. Hum Mol Genet 2017; 26:R37-R44. [PMID: 28459979 DOI: 10.1093/hmg/ddx168] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 04/27/2017] [Indexed: 12/11/2022] Open
Abstract
Unraveling the genetics of the paralytic strabismus syndromes known as congenital cranial dysinnervation disorders (CCDDs) is both informing physicians and their patients and broadening our understanding of development of the ocular motor system. Genetic mutations underlying ocular CCDDs alter either motor neuron specification or motor nerve development, and highlight the importance of modulations of cell signaling, cytoskeletal transport, and microtubule dynamics for axon growth and guidance. Here we review recent advances in our understanding of two CCDDs, congenital fibrosis of the extraocular muscles (CFEOM) and Duane retraction syndrome (DRS), and discuss what they have taught us about mechanisms of axon guidance and selective vulnerability. CFEOM presents with congenital ptosis and restricted eye movements, and can be caused by heterozygous missense mutations in the kinesin motor protein KIF21A or in the β-tubulin isotypes TUBB3 or TUBB2B. CFEOM-causing mutations in these genes alter protein function and result in axon growth and guidance defects. DRS presents with inability to abduct one or both eyes. It can be caused by decreased function of several transcription factors critical for abducens motor neuron identity, including MAFB, or by heterozygous missense mutations in CHN1, which encodes α2-chimaerin, a Rac-GAP GTPase that affects cytoskeletal dynamics. Examination of the orbital innervation in mice lacking Mafb has established that the stereotypical misinnervation of the lateral rectus by fibers of the oculomotor nerve in DRS is secondary to absence of the abducens nerve. Studies of a CHN1 mouse model have begun to elucidate mechanisms of selective vulnerability in the nervous system.
Collapse
Affiliation(s)
- Mary C Whitman
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA.,Department of Ophthalmology, Boston Children's Hospital, Boston, MA 02115, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Elizabeth C Engle
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA.,Department of Ophthalmology, Boston Children's Hospital, Boston, MA 02115, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA.,Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA.,Department of Neurology, Harvard Medical School, Boston, MA 02115, USA.,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
38
|
Biological functions of CDK5 and potential CDK5 targeted clinical treatments. Oncotarget 2017; 8:17373-17382. [PMID: 28077789 PMCID: PMC5370047 DOI: 10.18632/oncotarget.14538] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 12/17/2016] [Indexed: 12/11/2022] Open
Abstract
Cyclin dependent kinases are proline-directed serine/threonine protein kinases that are traditionally activated upon association with a regulatory subunit. For most CDKs, activation by a cyclin occurs through association and phosphorylation of the CDK’s T-loop. CDK5 is unusual because it is not typically activated upon binding with a cyclin and does not require T-loop phosphorylation for activation, even though it has high amino acid sequence homology with other CDKs. While it was previously thought that CDK5 only interacted with p35 or p39 and their cleaved counterparts, Recent evidence suggests that CDK5 can interact with certain cylins, amongst other proteins, which modulate CDK5 activity levels. This review discusses recent findings of molecular interactions that regulate CDK5 activity and CDK5 associated pathways that are implicated in various diseases. Also covered herein is the growing body of evidence for CDK5 in contributing to the onset and progression of tumorigenesis.
Collapse
|
39
|
Protein Tyrosine Phosphatase δ Mediates the Sema3A-Induced Cortical Basal Dendritic Arborization through the Activation of Fyn Tyrosine Kinase. J Neurosci 2017. [PMID: 28637841 DOI: 10.1523/jneurosci.2519-16.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Leukocyte common antigen-related (LAR) class protein tyrosine phosphatases (PTPs) are critical for axonal guidance; however, their relation to specific guidance cues is poorly defined. We here show that PTP-3, a LAR homolog in Caenorhabditis elegans, is involved in axon guidance regulated by Semaphorin-2A-signaling. PTPδ, one of the vertebrate LAR class PTPs, participates in the Semaphorin-3A (Sema3A)-induced growth cone collapse response of primary cultured dorsal root ganglion neurons from Mus musculus embryos. In vivo, however, the contribution of PTPδ in Sema3A-regualted axon guidance was minimal. Instead, PTPδ played a major role in Sema3A-dependent cortical dendritic growth. Ptpδ-/- and Sema3a-/- mutant mice exhibited poor arborization of basal dendrites of cortical layer V neurons. This phenotype was observed in both male and female mutants. The double-heterozygous mutants, Ptpδ+/-; Sema3a+/-, also showed a similar phenotype, indicating the genetic interaction. In Ptpδ-/- brains, Fyn and Src kinases were hyperphosphorylated at their C-terminal Tyr527 residues. Sema3A-stimulation induced dephosphorylation of Tyr527 in the dendrites of wild-type cortical neurons but not of Ptpδ-/- Arborization of cortical basal dendrites was reduced in Fyn-/- as well as in Ptpδ+/-; Fyn+/- double-heterozygous mutants. Collectively, PTPδ mediates Sema3A-signaling through the activation of Fyn by C-terminal dephosphorylation.SIGNIFICANCE STATEMENT The relation of leukocyte common antigen-related (LAR) class protein tyrosine phosphatases (PTPs) and specific axon guidance cues is poorly defined. We show that PTP-3, a LAR homolog in Caenorhabditis elegans, participates in Sema2A-regulated axon guidance. PTPδ, a member of vertebrate LAR class PTPs, is involved in Sema3A-regulated cortical dendritic growth. In Sema3A signaling, PTPδ activates Fyn and Src kinases by dephosphorylating their C-terminal Tyr residues. This is the first evidence showing that LAR class PTPs participate in Semaphorin signaling in vivo.
Collapse
|
40
|
Chilton JK, Guthrie S. Axons get ahead: Insights into axon guidance and congenital cranial dysinnervation disorders. Dev Neurobiol 2017; 77:861-875. [DOI: 10.1002/dneu.22477] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 12/07/2016] [Accepted: 12/07/2016] [Indexed: 11/12/2022]
Affiliation(s)
- John K. Chilton
- Wellcome Wolfson Centre for Medical Research; University of Exeter Medical School, Wellcome-Wolfson Centre for Medical Research; Exeter EX2 5DW United Kingdom
| | - Sarah Guthrie
- School of Life Sciences; University of Sussex; Falmer Brighton, BN1 9QG
| |
Collapse
|
41
|
Wang L, Liu W, Tang H, Xie X, Zou C, Wang Y, Gao Z, Yin J. DRP5 is involved in cancer cell growth and predicts poor prognosis in human osteosarcoma. Cancer Med 2017; 6:982-993. [PMID: 28374915 PMCID: PMC5430105 DOI: 10.1002/cam4.1009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/13/2016] [Accepted: 12/15/2016] [Indexed: 12/11/2022] Open
Abstract
Osteosarcoma is an extremely aggressive primary malignant bone tumor of childhood. Collapsin response mediator proteins (CRMPs), which are highly expressed in the developing nervous system, were recently shown to be associated with cancer development. However, the relationship between DRP5 (CRMP5) and osteosarcoma has not been evaluated. In this study, we investigated the role of DRP5 in the regulation of osteosarcoma growth. DRP5 mRNA and protein levels were significantly upregulated in human osteosarcoma cell lines and associated with increased migration and invasion. Genetic knockdown of DRP5 markedly suppressed the expression of matrix metalloproteinase (MMP)‐2 and MMP‐9. DRP5 silencing significantly inhibited osteosarcoma cell growth in vitro and in a xenograft mouse model in vivo. Microarray immunohistochemical analysis of osteosarcoma specimens and Kaplan–Meier analysis showed that patients with high DRP5 protein expression had shorter overall survival than those with low DRP5 levels. Taken together, these results suggest that DRP5 plays a critical role in the regulation of osteosarcoma and could be a potential therapeutic target and prognostic factor in osteosarcoma.
Collapse
Affiliation(s)
- Lin Wang
- Department of Oncology, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Weihai Liu
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hengtao Tang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xianbiao Xie
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Changye Zou
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yongqian Wang
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhenhua Gao
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Junqiang Yin
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
42
|
Nugent AA, Park JG, Wei Y, Tenney AP, Gilette NM, DeLisle MM, Chan WM, Cheng L, Engle EC. Mutant α2-chimaerin signals via bidirectional ephrin pathways in Duane retraction syndrome. J Clin Invest 2017; 127:1664-1682. [PMID: 28346224 DOI: 10.1172/jci88502] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 02/02/2017] [Indexed: 01/18/2023] Open
Abstract
Duane retraction syndrome (DRS) is the most common form of congenital paralytic strabismus in humans and can result from α2-chimaerin (CHN1) missense mutations. We report a knockin α2-chimaerin mouse (Chn1KI/KI) that models DRS. Whole embryo imaging of Chn1KI/KI mice revealed stalled abducens nerve growth and selective trochlear and first cervical spinal nerve guidance abnormalities. Stalled abducens nerve bundles did not reach the orbit, resulting in secondary aberrant misinnervation of the lateral rectus muscle by the oculomotor nerve. By contrast, Chn1KO/KO mice did not have DRS, and embryos displayed abducens nerve wandering distinct from the Chn1KI/KI phenotype. Murine embryos lacking EPH receptor A4 (Epha4KO/KO), which is upstream of α2-chimaerin in corticospinal neurons, exhibited similar abducens wandering that paralleled previously reported gait alterations in Chn1KO/KO and Epha4KO/KO adult mice. Findings from Chn1KI/KI Epha4KO/KO mice demonstrated that mutant α2-chimaerin and EphA4 have different genetic interactions in distinct motor neuron pools: abducens neurons use bidirectional ephrin signaling via mutant α2-chimaerin to direct growth, while cervical spinal neurons use only ephrin forward signaling, and trochlear neurons do not use ephrin signaling. These findings reveal a role for ephrin bidirectional signaling upstream of mutant α2-chimaerin in DRS, which may contribute to the selective vulnerability of abducens motor neurons in this disorder.
Collapse
|
43
|
Huang GH, Sun ZL, Li HJ, Feng DF. Rho GTPase-activating proteins: Regulators of Rho GTPase activity in neuronal development and CNS diseases. Mol Cell Neurosci 2017; 80:18-31. [PMID: 28163190 DOI: 10.1016/j.mcn.2017.01.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 01/06/2017] [Accepted: 01/29/2017] [Indexed: 12/22/2022] Open
Abstract
The Rho family of small GTPases was considered as molecular switches in regulating multiple cellular events, including cytoskeleton reorganization. The Rho GTPase-activating proteins (RhoGAPs) are one of the major families of Rho GTPase regulators. RhoGAPs were initially considered negative mediators of Rho signaling pathways via their GAP domain. Recent studies have demonstrated that RhoGAPs also regulate numerous aspects of neuronal development and are related to various neurodegenerative diseases in GAP-dependent and GAP-independent manners. Moreover, RhoGAPs are regulated through various mechanisms, such as phosphorylation. To date, approximately 70 RhoGAPs have been identified; however, only a small portion has been thoroughly investigated. Thus, the characterization of important RhoGAPs in the central nervous system is crucial to understand their spatiotemporal role during different stages of neuronal development. In this review, we summarize the current knowledge of RhoGAPs in the brain with an emphasis on their molecular function, regulation mechanism and disease implications in the central nervous system.
Collapse
Affiliation(s)
- Guo-Hui Huang
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201900, China
| | - Zhao-Liang Sun
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201900, China
| | - Hong-Jiang Li
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201900, China
| | - Dong-Fu Feng
- Department of Neurosurgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201900, China; Institute of Traumatic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 201900, China.
| |
Collapse
|
44
|
Sato S, Nakamura F, Hiroshima Y, Nagashima Y, Kato I, Yamashita N, Goshima Y, Endo I. Caerulein-induced pancreatitis augments the expression and phosphorylation of collapsin response mediator protein 4. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2016; 23:422-31. [PMID: 27207309 DOI: 10.1002/jhbp.361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/19/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Chronic pancreatitis is a significant risk factor for pancreatic cancer. Previously, we demonstrated that the pancreatic cancer cells show enhanced expression of collapsin response mediator protein 4 (CRMP4) that strongly correlates with severe venous invasion, liver metastasis, and poor prognosis. However, involvement of CRMP4 in acute or chronic pancreatitis remains unknown. METHODS Acute and chronic pancreatitis mice models were developed by periodic injection of caerulein. The expression levels of CRMP4 and its phosphorylation were examined. RESULTS Elevated CRMP4 levels were observed in the infiltrated lymphocytes as well as in the pancreas parenchyma of both acute and chronic pancreatitis. The expression pattern of phosphorylated CRMP4 was similar to that of CRMP4. Cdk5 partially co-localized with the phosphorylated CRMP4. CONCLUSIONS Pancreatitis induces CRMP4 expression in the pancreas parenchyma and in the infiltrated lymphocytes. Overlapping expression of CRMP4 and Cdk5 may suggest that the Cdk5 is at least, in part, responsible for the phosphorylation of CRMP4. The results suggest that CRMP4 is involved in the inflammatory response in pancreatitis. Understanding the mechanisms of CRMP4 would help us to develop novel therapeutic strategies against acute or chronic pancreatitis, and pancreatic cancer.
Collapse
Affiliation(s)
- Sho Sato
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Fumio Nakamura
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa Ward, Yokohama, Kanagawa, 236-0004, Japan
| | - Yukihiko Hiroshima
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yoji Nagashima
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Ikuma Kato
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Naoya Yamashita
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa Ward, Yokohama, Kanagawa, 236-0004, Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa Ward, Yokohama, Kanagawa, 236-0004, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
45
|
Shah K, Lahiri DK. A Tale of the Good and Bad: Remodeling of the Microtubule Network in the Brain by Cdk5. Mol Neurobiol 2016; 54:2255-2268. [PMID: 26944284 DOI: 10.1007/s12035-016-9792-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/11/2016] [Indexed: 10/22/2022]
Abstract
Cdk5, a cyclin-dependent kinase family member, is a global orchestrator of neuronal cytoskeletal dynamics. During embryogenesis, Cdk5 is indispensable for brain development. In adults, it is essential for numerous neuronal processes, including higher cognitive functions such as learning and memory formation, drug addiction, pain signaling, and long-term behavior changes through long-term potentiation and long-term depression, all of which rely on rapid alterations in the cytoskeleton. Cdk5 activity becomes deregulated in various brain disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, attention-deficit hyperactivity disorder, epilepsy, schizophrenia, and ischemic stroke; these all result in profound remodeling of the neuronal cytoskeleton. This Commentary specifically focuses on the pleiotropic contribution of Cdk5 in regulating neuronal microtubule remodeling. Because the vast majority of the physiological substrates of Cdk5 are associated with the neuronal cytoskeleton, our emphasis is on the Cdk5 substrates, such as CRMP2, stathmin, drebrin, dixdc1, axin, MAP2, MAP1B, doublecortin, kinesin-5, and tau, that have allowed to unravel the molecular mechanisms through which Cdk5 exerts its divergent roles in regulating neuronal microtubule dynamics, both in healthy and disease states.
Collapse
Affiliation(s)
- Kavita Shah
- Department of Chemistry and Purdue University Center for Cancer Research, 560 Oval Drive, West Lafayette, IN, 47907, USA.
| | - Debomoy K Lahiri
- Departments of Psychiatry and Medical & Molecular Genetics, Institute of Psychiatric Research, Neuroscience Research Center, Indiana University School of Medicine, 320 W. 15th Street, Indianapolis, IN, 46202-2266, USA
| |
Collapse
|
46
|
Rozés Salvador V, Heredia F, Berardo A, Palandri A, Wojnacki J, Vivinetto AL, Sheikh KA, Caceres A, Lopez PHH. Anti-glycan antibodies halt axon regeneration in a model of Guillain Barrè Syndrome axonal neuropathy by inducing microtubule disorganization via RhoA-ROCK-dependent inactivation of CRMP-2. Exp Neurol 2016; 278:42-53. [PMID: 26804001 DOI: 10.1016/j.expneurol.2016.01.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 01/19/2016] [Indexed: 10/22/2022]
Abstract
Several reports have linked the presence of high titers of anti-Gg Abs with delayed recovery/poor prognosis in GBS. In most cases, failure to recover is associated with halted/deficient axon regeneration. Previous work identified that monoclonal and patient-derived anti-Gg Abs can act as inhibitory factors in an animal model of axon regeneration. Further studies using primary dorsal root ganglion neuron (DRGn) cultures demonstrated that anti-Gg Abs can inhibit neurite outgrowth by targeting gangliosides via activation of the small GTPase RhoA and its associated kinase (ROCK), a signaling pathway common to other established inhibitors of axon regeneration. We aimed to study the molecular basis of the inhibitory effect of anti-Gg abs on neurite outgrowth by dissecting the molecular dynamics of growth cones (GC) cytoskeleton in relation to the spatial-temporal analysis of RhoA activity. We now report that axon growth inhibition in DRGn induced by a well characterized mAb targeting gangliosides GD1a/GT1b involves: i) an early RhoA/ROCK-independent collapse of lamellipodia; ii) a RhoA/ROCK-dependent shrinking of filopodia; and iii) alteration of GC microtubule organization/and presumably dynamics via RhoA/ROCK-dependent phosphorylation of CRMP-2 at threonine 555. Our results also show that mAb 1B7 inhibits peripheral axon regeneration in an animal model via phosphorylation/inactivation of CRMP-2 at threonine 555. Overall, our data may help to explain the molecular mechanisms underlying impaired nerve repair in GBS. Future work should define RhoA-independent pathway/s and effectors regulating actin cytoskeleton, thus providing an opportunity for the design of a successful therapy to guarantee an efficient target reinnervation.
Collapse
Affiliation(s)
- Victoria Rozés Salvador
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina
| | - Florencia Heredia
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina
| | - Andrés Berardo
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina
| | - Anabela Palandri
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina
| | - Jose Wojnacki
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina
| | - Ana L Vivinetto
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina
| | - Kazim A Sheikh
- Department of Neurology, University of Texas Medical School at Houston, Houston, USA
| | - Alfredo Caceres
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina
| | - Pablo H H Lopez
- Laboratory of Neurobiology, Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Argentina; Facultad de Psicología, Universidad Nacional de Córdoba, Argentina.
| |
Collapse
|
47
|
Developmental RacGAP α2-Chimaerin Signaling Is a Determinant of the Morphological Features of Dendritic Spines in Adulthood. J Neurosci 2016; 35:13728-44. [PMID: 26446225 DOI: 10.1523/jneurosci.0419-15.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Morphological characteristics of dendritic spines form the basis of cognitive ability. However, molecular mechanisms involved in fine-tuning of spine morphology during development are not fully understood. Moreover, it is unclear whether, and to what extent, these developmental mechanisms determine the normal adult spine morphological features. Here, we provide evidence that α2-isoform of Rac-specific GTPase-activating protein α-chimaerin (α2-chimaerin) is involved in spine morphological refinement during late postnatal period, and furthermore show that this developmental α2-chimaerin function affects adult spine morphologies. We used a series of mice with global and conditional knock-out of α-chimaerin isoforms (α1-chimaerin and α2-chimaerin). α2-Chimaerin disruption, but not α1-chimaerin disruption, in the mouse results in an increased size (and density) of spines in the hippocampus. In contrast, overexpression of α2-chimaerin in developing hippocampal neurons induces a decrease of spine size. Disruption of α2-chimaerin suppressed EphA-mediated spine morphogenesis in cultured developing hippocampal neurons. α2-Chimaerin disruption that begins during the juvenile stage results in an increased size of spines in the hippocampus. Meanwhile, spine morphologies are unaltered when α2-chimaerin is deleted only in adulthood. Consistent with these spine morphological results, disruption of α2-chimaerin beginning in the juvenile stage led to an increase in contextual fear learning in adulthood; whereas contextual learning was recently shown to be unaffected when α2-chimaerin was deleted only in adulthood. Together, these results suggest that α2-chimaerin signaling in developmental stages contributes to determination of the morphological features of adult spines and establishment of normal cognitive ability. SIGNIFICANCE STATEMENT Recent studies of neurodevelopmental disorders in humans and their animal models have led to an attractive hypothesis that spine morphogenesis during development forms the basis of adult cognition. In particular, the roles of Rac and its regulators, such as Rac-specific GTPase-activating proteins (RacGAPs) and Rac guanine nucleotide exchange factors, are a topic of focus in spine morphogenesis and cognitive ability. Using a series of mice with global and conditional knock-out (KO) of RacGAP α-chimaerin isoforms (α1-chimaerin and α2-chimaerin), we provide compelling evidence demonstrating that α2-chimaerin is involved in spine morphological refinement during late postnatal development and that this developmental α2-chimaerin function affects adult spine morphologies. Furthermore, our results clearly showed that α2-chimaerin signaling during late postnatal development contributes to normal cognitive ability in adult mice.
Collapse
|
48
|
Grant NJ, Coates PJ, Woods YL, Bray SE, Morrice NA, Hastie CJ, Lamont DJ, Carey FA, Sutherland C. Phosphorylation of a splice variant of collapsin response mediator protein 2 in the nucleus of tumour cells links cyclin dependent kinase-5 to oncogenesis. BMC Cancer 2015; 15:885. [PMID: 26555036 PMCID: PMC4640224 DOI: 10.1186/s12885-015-1691-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 10/07/2015] [Indexed: 12/17/2022] Open
Abstract
Background Cyclin-dependent protein kinase-5 (CDK5) is an unusual member of the CDK family as it is not cell cycle regulated. However many of its substrates have roles in cell growth and oncogenesis, raising the possibility that CDK5 modulation could have therapeutic benefit. In order to establish whether changes in CDK5 activity are associated with oncogenesis one could quantify phosphorylation of CDK5 targets in disease tissue in comparison to appropriate controls. However the identity of physiological and pathophysiological CDK5 substrates remains the subject of debate, making the choice of CDK5 activity biomarkers difficult. Methods Here we use in vitro and in cell phosphorylation assays to identify novel features of CDK5 target sequence determinants that confer enhanced CDK5 selectivity, providing means to select substrate biomarkers of CDK5 activity with more confidence. We then characterize tools for the best CDK5 substrate we identified to monitor its phosphorylation in human tissue and use these to interrogate human tumour arrays. Results The close proximity of Arg/Lys amino acids and a proline two residues N-terminal to the phosphorylated residue both improve recognition of the substrate by CDK5. In contrast the presence of a proline two residues C-terminal to the target residue dramatically reduces phosphorylation rate. Serine-522 of Collapsin Response Mediator-2 (CRMP2) is a validated CDK5 substrate with many of these structural criteria. We generate and characterise phosphospecific antibodies to Ser522 and show that phosphorylation appears in human tumours (lung, breast, and lymphoma) in stark contrast to surrounding non-neoplastic tissue. In lung cancer the anti-phospho-Ser522 signal is positive in squamous cell carcinoma more frequently than adenocarcinoma. Finally we demonstrate that it is a specific and unusual splice variant of CRMP2 (CRMP2A) that is phosphorylated in tumour cells. Conclusions For the first time this data associates altered CDK5 substrate phosphorylation with oncogenesis in some but not all tumour types, implicating altered CDK5 activity in aspects of pathogenesis. These data identify a novel oncogenic mechanism where CDK5 activation induces CRMP2A phosphorylation in the nuclei of tumour cells. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1691-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nicola J Grant
- Division of Cardiovascular and Diabetes Medicine, University of Dundee, Ninewells Medical School, DD1 9SY, Dundee, UK.
| | | | - Yvonne L Woods
- Department of Pathology, Ninewells Hospital, NHS Tayside, Dundee, UK.
| | - Susan E Bray
- Division of Cancer, University of Dundee, Dundee, UK.
| | | | - C James Hastie
- Division of Signal Transduction and Therapy, University of Dundee, Dundee, UK.
| | - Douglas J Lamont
- FingerPrints Proteomics Facility, University of Dundee, Dundee, UK.
| | - Francis A Carey
- Department of Pathology, Ninewells Hospital, NHS Tayside, Dundee, UK.
| | - Calum Sutherland
- Division of Cardiovascular and Diabetes Medicine, University of Dundee, Ninewells Medical School, DD1 9SY, Dundee, UK.
| |
Collapse
|
49
|
Laskowska-Macios K, Nys J, Hu TT, Zapasnik M, Van der Perren A, Kossut M, Burnat K, Arckens L. Binocular pattern deprivation interferes with the expression of proteins involved in primary visual cortex maturation in the cat. Mol Brain 2015; 8:48. [PMID: 26271461 PMCID: PMC4536594 DOI: 10.1186/s13041-015-0137-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/31/2015] [Indexed: 12/03/2022] Open
Abstract
Background Binocular pattern deprivation from eye opening (early BD) delays the maturation of the primary visual cortex. This delay is more pronounced for the peripheral than the central visual field representation within area 17, particularly between the age of 2 and 4 months [Laskowska-Macios, Cereb Cortex, 2014]. Results In this study, we probed for related dynamic changes in the cortical proteome. We introduced age, cortical region and BD as principal variables in a 2-D DIGE screen of area 17. In this way we explored the potential of BD-related protein expression changes between central and peripheral area 17 of 2- and 4-month-old BD (2BD, 4BD) kittens as a valid parameter towards the identification of brain maturation-related molecular processes. Consistent with the maturation delay, distinct developmental protein expression changes observed for normal kittens were postponed by BD, especially in the peripheral region. These BD-induced proteomic changes suggest a negative regulation of neurite outgrowth, synaptic transmission and clathrin-mediated endocytosis, thereby implicating these processes in normal experience-induced visual cortex maturation. Verification of the expression of proteins from each of the biological processes via Western analysis disclosed that some of the transient proteomic changes correlate to the distinct behavioral outcome in adult life, depending on timing and duration of the BD period [Neuroscience 2013;255:99-109]. Conclusions Taken together, the plasticity potential to recover from BD, in relation to ensuing restoration of normal visual input, appears to rely on specific protein expression changes and cellular processes induced by the loss of pattern vision in early life. Electronic supplementary material The online version of this article (doi:10.1186/s13041-015-0137-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Karolina Laskowska-Macios
- Laboratory of Neuroplasticity, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland. .,Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven - University of Leuven, 3000, Leuven, Belgium.
| | - Julie Nys
- Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven - University of Leuven, 3000, Leuven, Belgium.
| | - Tjing-Tjing Hu
- Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven - University of Leuven, 3000, Leuven, Belgium.
| | - Monika Zapasnik
- Laboratory of Neuroplasticity, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland.
| | - Anke Van der Perren
- Laboratory for Neurobiology and Gene Therapy, KU Leuven - University of Leuven, 3000, Leuven, Belgium.
| | - Malgorzata Kossut
- Laboratory of Neuroplasticity, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland.
| | - Kalina Burnat
- Laboratory of Neuroplasticity, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland.
| | - Lutgarde Arckens
- Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven - University of Leuven, 3000, Leuven, Belgium.
| |
Collapse
|
50
|
Kadoyama K, Matsuura K, Nakamura-Hirota T, Takano M, Otani M, Matsuyama S. Changes in the expression of collapsin response mediator protein-2 during synaptic plasticity in the mouse hippocampus. J Neurosci Res 2015; 93:1684-92. [DOI: 10.1002/jnr.23626] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/23/2015] [Accepted: 07/23/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Keiichi Kadoyama
- Department of Pharmaceutical Health Care; Faculty of Pharmaceutical Sciences; Himeji Dokkyo University; Himeji Japan
| | - Kenji Matsuura
- Department of Pharmaceutical Health Care; Faculty of Pharmaceutical Sciences; Himeji Dokkyo University; Himeji Japan
| | - Tooru Nakamura-Hirota
- Department of Pharmaceutical Health Care; Faculty of Pharmaceutical Sciences; Himeji Dokkyo University; Himeji Japan
| | - Masaoki Takano
- Department of Life Sciences Pharmacy; School of Pharmaceutical Sciences, Kobe Gakuin University; Kobe Japan
| | - Mieko Otani
- Department of Life Sciences Pharmacy; School of Pharmaceutical Sciences, Kobe Gakuin University; Kobe Japan
| | - Shogo Matsuyama
- Department of Pharmaceutical Health Care; Faculty of Pharmaceutical Sciences; Himeji Dokkyo University; Himeji Japan
| |
Collapse
|