1
|
Ganguly K, Adhikary K, Acharjee A, Acharjee P, Trigun SK, Mutlaq AS, Ashique S, Yasmin S, Alshahrani AM, Ansari MY. Biological significance and pathophysiological role of Matrix Metalloproteinases in the Central Nervous System. Int J Biol Macromol 2024; 280:135967. [PMID: 39322129 DOI: 10.1016/j.ijbiomac.2024.135967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 09/21/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
Matrix Metalloproteinases (MMPs), which are endopeptidase reliant on zinc, are low in embryonic tissues but increases in response to a variety of physiological stimulus and pathological stresses. Neuro-glial cells, endothelial cells, fibroblasts, and leucocytes secrete MMPs, which cleave extracellular matrix proteins in a time-dependent manner. MMPs affect synaptic plasticity and the development of short-term memory by controlling the size, shape, and excitatory synapses' function through the lateral diffusion of receptors. In addition, MMPs influence the Extracellular Matrix proteins in the Peri-Neuronal Net at the Neuro-glial interface, which aids in the establishment of long-term memory. Through modulating neuronal, and glial cells migration, differentiation, Neurogenesis, and survival, MMPs impact brain development in mammals. In adult brains, MMPs play a beneficial role in physiological plasticity, which includes learning, memory consolidation, social interaction, and complex behaviors, by proteolytically altering a wide variety of factors, including growth factors, cytokines, receptors, DNA repair enzymes, and matrix proteins. Additionally, stress, depression, addiction, hepatic encephalopathy, and stroke may all have negative effects on MMPs. In addition to their role in glioblastoma development, MMPs influence neurological diseases such as epilepsy, schizophrenia, autism spectrum disorder, brain damage, pain, neurodegeneration, and Alzheimer's and Parkinson's. To help shed light on the potential of MMPs as a therapeutic target for neurodegenerative diseases, this review summarizes their regulation, mode of action, and participation in brain physiological plasticity and pathological damage. Finally, by employing different MMP-based nanotools and inhibitors, MMPs may also be utilized to map the anatomical and functional connectome of the brain, analyze its secretome, and treat neurodegenerative illnesses.
Collapse
Affiliation(s)
- Krishnendu Ganguly
- Department of Medical Lab Technology, Paramedical College Durgapur, Helen Keller Sarani, Durgapur 713212, West Bengal, India.
| | - Krishnendu Adhikary
- Department of Medical Lab Technology, Paramedical College Durgapur, Helen Keller Sarani, Durgapur 713212, West Bengal, India.
| | - Arup Acharjee
- Molecular Omics Laboratory, Department of Zoology, University of Allahabad, Allahabad, Uttar Pradesh, India.
| | - Papia Acharjee
- Biochemistry Section, Department of Zoology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | - Surendra Kumar Trigun
- Biochemistry Section, Department of Zoology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | | | - Sumel Ashique
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur 713212, West Bengal, India.
| | - Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia.
| | - Asma M Alshahrani
- Department of Clinical Pharmacy, Faculty of Pharmacy, King Khalid University, Abha, Saudi Arabia; Department of Clinical Pharmacy, Shaqra University, Saudi Arabia.
| | - Mohammad Yousuf Ansari
- MM college of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India.
| |
Collapse
|
2
|
Itoh Y. Vesicle transport of matrix metalloproteinases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:361-380. [PMID: 38960480 DOI: 10.1016/bs.apcsb.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Multicellular organisms consist of cells and extracellular matrix (ECM). ECM creates a cellular microenvironment, and cells locally degrade the ECM according to their cellular activity. A major group of enzymes that modify ECM belongs to matrix metalloproteinases (MMPs) and play major roles in various pathophysiological events. ECM degradation by MMPs does not occur in all cellular surroundings but only where it is necessary, and cells achieve this by directionally secreting these proteolytic enzymes. Recent studies have indicated that such enzyme secretion is achieved by targeted vesicle transport along the microtubules, and several kinesin superfamily proteins (KIFs) have been identified as responsible motor proteins involved in the processes. This chapter discusses recent findings of the vesicle transport of MMPs and their roles.
Collapse
Affiliation(s)
- Yoshifumi Itoh
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
3
|
Wilkie IC. Basement Membranes, Brittlestar Tendons, and Their Mechanical Adaptability. BIOLOGY 2024; 13:375. [PMID: 38927255 PMCID: PMC11200632 DOI: 10.3390/biology13060375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024]
Abstract
Basement membranes (BMs) are thin layers of extracellular matrix that separate epithelia, endothelia, muscle cells, and nerve cells from adjacent interstitial connective tissue. BMs are ubiquitous in almost all multicellular animals, and their composition is highly conserved across the Metazoa. There is increasing interest in the mechanical functioning of BMs, including the involvement of altered BM stiffness in development and pathology, particularly cancer metastasis, which can be facilitated by BM destabilization. Such BM weakening has been assumed to occur primarily through enzymatic degradation by matrix metalloproteinases. However, emerging evidence indicates that non-enzymatic mechanisms may also contribute. In brittlestars (Echinodermata, Ophiuroidea), the tendons linking the musculature to the endoskeleton consist of extensions of muscle cell BMs. During the process of brittlestar autotomy, in which arms are detached for the purpose of self-defense, muscles break away from the endoskeleton as a consequence of the rapid destabilization and rupture of their BM-derived tendons. This contribution provides a broad overview of current knowledge of the structural organization and biomechanics of non-echinoderm BMs, compares this with the equivalent information on brittlestar tendons, and discusses the possible relationship between the weakening phenomena exhibited by BMs and brittlestar tendons, and the potential translational value of the latter as a model system of BM destabilization.
Collapse
Affiliation(s)
- Iain C Wilkie
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
4
|
Bajaj R, Warner AN, Fradette JF, Gibbons DL. Dance of The Golgi: Understanding Golgi Dynamics in Cancer Metastasis. Cells 2022; 11:1484. [PMID: 35563790 PMCID: PMC9102947 DOI: 10.3390/cells11091484] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 12/17/2022] Open
Abstract
The Golgi apparatus is at the center of protein processing and trafficking in normal cells. Under pathological conditions, such as in cancer, aberrant Golgi dynamics alter the tumor microenvironment and the immune landscape, which enhances the invasive and metastatic potential of cancer cells. Among these changes in the Golgi in cancer include altered Golgi orientation and morphology that contribute to atypical Golgi function in protein trafficking, post-translational modification, and exocytosis. Golgi-associated gene mutations are ubiquitous across most cancers and are responsible for modifying Golgi function to become pro-metastatic. The pharmacological targeting of the Golgi or its associated genes has been difficult in the clinic; thus, studying the Golgi and its role in cancer is critical to developing novel therapeutic agents that limit cancer progression and metastasis. In this review, we aim to discuss how disrupted Golgi function in cancer cells promotes invasion and metastasis.
Collapse
Affiliation(s)
- Rakhee Bajaj
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Amanda N. Warner
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Jared F. Fradette
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
| | - Don L. Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
5
|
Extracellular Metalloproteinases in the Plasticity of Excitatory and Inhibitory Synapses. Cells 2021; 10:cells10082055. [PMID: 34440823 PMCID: PMC8391609 DOI: 10.3390/cells10082055] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023] Open
Abstract
Long-term synaptic plasticity is shaped by the controlled reorganization of the synaptic proteome. A key component of this process is local proteolysis performed by the family of extracellular matrix metalloproteinases (MMPs). In recent years, considerable progress was achieved in identifying extracellular proteases involved in neuroplasticity phenomena and their protein substrates. Perisynaptic metalloproteinases regulate plastic changes at synapses through the processing of extracellular and membrane proteins. MMP9 was found to play a crucial role in excitatory synapses by controlling the NMDA-dependent LTP component. In addition, MMP3 regulates the L-type calcium channel-dependent form of LTP as well as the plasticity of neuronal excitability. Both MMP9 and MMP3 were implicated in memory and learning. Moreover, altered expression or mutations of different MMPs are associated with learning deficits and psychiatric disorders, including schizophrenia, addiction, or stress response. Contrary to excitatory drive, the investigation into the role of extracellular proteolysis in inhibitory synapses is only just beginning. Herein, we review the principal mechanisms of MMP involvement in the plasticity of excitatory transmission and the recently discovered role of proteolysis in inhibitory synapses. We discuss how different matrix metalloproteinases shape dynamics and turnover of synaptic adhesome and signal transduction pathways in neurons. Finally, we discuss future challenges in exploring synapse- and plasticity-specific functions of different metalloproteinases.
Collapse
|
6
|
Lu Y, Almeida S, Gao FB. TBK1 haploinsufficiency in ALS and FTD compromises membrane trafficking. Acta Neuropathol 2021; 142:217-221. [PMID: 34081168 PMCID: PMC8500533 DOI: 10.1007/s00401-021-02331-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 12/31/2022]
Affiliation(s)
- Yubing Lu
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | - Sandra Almeida
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
7
|
Dumitru CA, Brouwer E, Stelzer T, Nocerino S, Rading S, Wilkens L, Sandalcioglu IE, Karsak M. Dynein Light Chain Protein Tctex1: A Novel Prognostic Marker and Molecular Mediator in Glioblastoma. Cancers (Basel) 2021; 13:cancers13112624. [PMID: 34071761 PMCID: PMC8199143 DOI: 10.3390/cancers13112624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Glioblastoma (GBM) remains one of the deadliest solid cancers, with only a dismal proportion of GBM patients achieving 5-year survival. Thus, it is critical to identify molecular mechanisms that could be targeted by novel therapeutic approaches in this tumor type. Our study identified Tctex1/DYNLT1 as an independent prognostic marker for the overall survival of GBM patients. Importantly, Tctex1 promoted the aggressiveness of GBM cells by enhancing tumor proliferation and invasion. These effects of Tctex1 appeared to be modulated via phosphorylation of retinoblastoma protein (RB) and the release of matrix metalloprotease 2 (MMP2), respectively. As Tctex1 can potentially be inhibited in vivo, our study provides a rationale for novel, individualized therapeutic strategies in GBM patients. Abstract The purpose of this study was to determine the role of Tctex1 (DYNLT1, dynein light chain-1) in the pathophysiology of glioblastoma (GBM). To this end, we performed immunohistochemical analyses on tissues from GBM patients (n = 202). Tctex1 was additionally overexpressed in two different GBM cell lines, which were then evaluated in regard to their proliferative and invasive properties. We found that Tctex1 levels were significantly higher in GBM compared to healthy adjacent brain tissues. Furthermore, high Tctex1 expression was significantly associated with the short overall- (p = 0.002, log-rank) and progression-free (p = 0.028, log-rank) survival of GBM patients and was an independent predictor of poor overall survival in multivariate Cox-regression models. In vitro, Tctex1 promoted the metabolic activity, anchorage-independent growth and proliferation of GBM cells. This phenomenon was previously shown to occur via the phosphorylation of retinoblastoma protein (phospho-RB). Here, we found a direct and significant correlation between the levels of Tctex1 and phospho-RB (Ser807/801) in tissues from GBM patients (p = 0.007, Rho = 0.284, Spearman’s rank). Finally, Tctex1 enhanced the invasiveness of GBM cells and the release of pro-invasive matrix metalloprotease 2 (MMP2). These findings indicate that Tctex1 promotes GBM progression and therefore might be a useful therapeutic target in this type of cancer.
Collapse
Affiliation(s)
- Claudia Alexandra Dumitru
- Department of Neurosurgery, Otto-von-Guericke University, 39120 Magdeburg, Germany;
- Correspondence: (C.A.D.); (M.K.)
| | - Eileen Brouwer
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (E.B.); (T.S.); (S.N.); (S.R.)
| | - Tamina Stelzer
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (E.B.); (T.S.); (S.N.); (S.R.)
| | - Salvatore Nocerino
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (E.B.); (T.S.); (S.N.); (S.R.)
| | - Sebastian Rading
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (E.B.); (T.S.); (S.N.); (S.R.)
| | - Ludwig Wilkens
- Department of Pathology, Nordstadt Hospital Hannover, 30167 Hannover, Germany;
| | | | - Meliha Karsak
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (E.B.); (T.S.); (S.N.); (S.R.)
- Correspondence: (C.A.D.); (M.K.)
| |
Collapse
|
8
|
Hu G, Xia ZS, Guo X. Differential expression of serum GBP-28, NBP-Cyc 3 and TIMP-1 complicates pregnancy in hypertensive disorder pregnancy. J Reprod Immunol 2021; 144:103288. [PMID: 33601303 DOI: 10.1016/j.jri.2021.103288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/24/2021] [Accepted: 02/06/2021] [Indexed: 10/22/2022]
Abstract
The current study is aimed at analyzing the correlation and differential expression of three entities namely, TIMP metallopeptidase inhibitor-1 (TIMP-1), a glycoprotein, serum adipokine (GBP-28), an amino acid protein and neuroendocrine basic polypeptide NBP-cystatin3 (NBP-Cyc 3) in HDP (Hypertensive Disorders complicating Pregnancy). A total of 63 patients, diagnosed with HDP at the study hospital during the study period, was placed under treatment (HDP) group. While healthy group had a total of 50 women with normal pregnancy during the same period. Both these groups were compared in terms of GBP-28, TIMP-1 and NBP-Cyc 3 levels. Further, the author also checked the correlation, diagnostic value and prognosis for the three factors and HDP. There was a significant increase observed in the expression levels of serum TIMP-1 and NBP-Cyc 3 in HDP during ELISA compared to GB. However, HDP group recorded low value of serum GBP-28 than healthy group (all P < 0.001). There is a relationship between the expressions of GBP-28, TIMP-1 and NBP-Cyc 3 and the abnormalities in lipid and glucose metabolisms, resulting in severe clinical conditions among HDP patients. The inference from spearman correlation analysis is that serum GBP-28 and the severity of HDP are negatively correlated. While Serum TIMP-1 and NBP-Cyc 3 had a positive correlation with the severity of HDP (all P < 0.001). When diagnosing HDP, the AUC values of both GBP-28 and NBP-Cyc 3 single diagnosis were above 0.8. Multivariate conditional logistic regression was deployed to assess the risk factors associated with HDP. The results listed the independent risk factors such as GBP-28, TIMP-1 and NBP-Cyc 3 and disease severity for the prognosis of HDP. Among HDP patients, upregulated expressions of serum TIMP-1and NBP-Cyc 3 were observed while in case of GBP-28, it was vice versa. The significant role, played by GBP-28, TIMP-1 and NBP-Cyc 3 in the progression of HDP, makes these entities potential serum biomarkers in diagnosis and assessment of HDP.
Collapse
Affiliation(s)
- Guantong Hu
- Institute of Cardiovascular & Medical Sciences, The University of Glasgow, Scotland, UK
| | - Zhong-Su Xia
- Second Hospital of Shandong University, Shangdong University, Jinan, Shandong, China.
| | - Xuan Guo
- Second Hospital of Shandong University, Shangdong University, Jinan, Shandong, China
| |
Collapse
|
9
|
Bajaj R, Kundu ST, Grzeskowiak CL, Fradette JJ, Scott KL, Creighton CJ, Gibbons DL. IMPAD1 and KDELR2 drive invasion and metastasis by enhancing Golgi-mediated secretion. Oncogene 2020; 39:5979-5994. [PMID: 32753652 PMCID: PMC7539228 DOI: 10.1038/s41388-020-01410-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/14/2020] [Accepted: 07/23/2020] [Indexed: 01/12/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the deadliest form of cancer worldwide, due in part to its proclivity to metastasize. Identifying novel drivers of invasion and metastasis holds therapeutic potential for the disease. We conducted a gain-of-function invasion screen, which identified two separate hits, IMPAD1 and KDELR2, as robust, independent drivers of lung cancer invasion and metastasis. Given that IMPAD1 and KDELR2 are known to be localized to the ER-Golgi pathway, we studied their common mechanism of driving in vitro invasion and in vivo metastasis and demonstrated that they enhance Golgi-mediated function and secretion. Therapeutically inhibiting matrix metalloproteases (MMPs) suppressed both IMPAD1- and KDELR2-mediated invasion. The hits from this unbiased screen and the mechanistic validation highlight Golgi function as one of the key cellular features altered during invasion and metastasis.
Collapse
Affiliation(s)
- Rakhee Bajaj
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Samrat T Kundu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Caitlin L Grzeskowiak
- Department of Molecular and Human Genetics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Medicine, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jared J Fradette
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Kenneth L Scott
- Department of Medicine, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chad J Creighton
- Department of Medicine, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
10
|
Pacheco-Fernandez N, Pakdel M, Blank B, Sanchez-Gonzalez I, Weber K, Tran ML, Hecht TKH, Gautsch R, Beck G, Perez F, Hausser A, Linder S, von Blume J. Nucleobindin-1 regulates ECM degradation by promoting intra-Golgi trafficking of MMPs. J Cell Biol 2020; 219:e201907058. [PMID: 32479594 PMCID: PMC7401813 DOI: 10.1083/jcb.201907058] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 12/29/2019] [Accepted: 05/04/2020] [Indexed: 12/14/2022] Open
Abstract
Matrix metalloproteinases (MMPs) degrade several ECM components and are crucial modulators of cell invasion and tissue organization. Although much has been reported about their function in remodeling ECM in health and disease, their trafficking across the Golgi apparatus remains poorly understood. Here we report that the cis-Golgi protein nucleobindin-1 (NUCB1) is critical for MMP2 and MT1-MMP trafficking along the Golgi apparatus. This process is Ca2+-dependent and is required for invasive MDA-MB-231 cell migration as well as for gelatin degradation in primary human macrophages. Our findings emphasize the importance of NUCB1 as an essential component of MMP transport and its overall impact on ECM remodeling.
Collapse
Affiliation(s)
| | | | - Birgit Blank
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | | | - Kathrin Weber
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg, Hamburg, Germany
| | - Mai Ly Tran
- Max Planck Institute of Biochemistry, Martinsried, Germany
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Tobias Karl-Heinz Hecht
- Max Planck Institute of Biochemistry, Martinsried, Germany
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Renate Gautsch
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Gisela Beck
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Franck Perez
- Institute Curie, PSL Research University, Centre National de la Recherche Scientifique, UMR 144, Paris, France
| | - Angelika Hausser
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Stefan Linder
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg, Hamburg, Germany
| | - Julia von Blume
- Max Planck Institute of Biochemistry, Martinsried, Germany
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
11
|
Murase S, Winkowski D, Liu J, Kanold PO, Quinlan EM. Homeostatic regulation of perisynaptic matrix metalloproteinase 9 (MMP9) activity in the amblyopic visual cortex. eLife 2019; 8:52503. [PMID: 31868167 PMCID: PMC6961978 DOI: 10.7554/elife.52503] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/19/2019] [Indexed: 01/07/2023] Open
Abstract
Dark exposure (DE) followed by light reintroduction (LRx) reactivates robust synaptic plasticity in adult mouse primary visual cortex (V1), which allows subsequent recovery from amblyopia. Previously we showed that perisynaptic proteolysis by MMP9 mediates the enhancement of plasticity by LRx in binocular adult mice (Murase et al., 2017). However, it was unknown if a visual system compromised by amblyopia could engage this pathway. Here we show that LRx to adult amblyopic mice induces perisynaptic MMP2/9 activity and extracellular matrix (ECM) degradation in deprived and non-deprived V1. Indeed, LRx restricted to the amblyopic eye is sufficient to induce robust MMP2/9 activity at thalamo-cortical synapses and ECM degradation in deprived V1. Two-photon live imaging demonstrates that the history of visual experience regulates MMP2/9 activity in V1, and that DE lowers the threshold for the proteinase activation. The homeostatic reduction of the MMP2/9 activation threshold by DE enables visual input from the amblyopic pathway to trigger robust perisynaptic proteolysis.
Collapse
Affiliation(s)
- Sachiko Murase
- Department of Biology, University of Maryland, College Park, United States.,Neuroscience Cognitive Sciences Program, University of Maryland, College Park, United States
| | - Dan Winkowski
- Department of Biology, University of Maryland, College Park, United States.,Neuroscience Cognitive Sciences Program, University of Maryland, College Park, United States
| | - Ji Liu
- Department of Biology, University of Maryland, College Park, United States.,Neuroscience Cognitive Sciences Program, University of Maryland, College Park, United States
| | - Patrick O Kanold
- Department of Biology, University of Maryland, College Park, United States.,Neuroscience Cognitive Sciences Program, University of Maryland, College Park, United States
| | - Elizabeth M Quinlan
- Department of Biology, University of Maryland, College Park, United States.,Neuroscience Cognitive Sciences Program, University of Maryland, College Park, United States
| |
Collapse
|
12
|
Nichols EL, Smith CJ. Synaptic-like Vesicles Facilitate Pioneer Axon Invasion. Curr Biol 2019; 29:2652-2664.e4. [DOI: 10.1016/j.cub.2019.06.078] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/24/2019] [Accepted: 06/26/2019] [Indexed: 12/19/2022]
|
13
|
Krishnaswamy VR, Benbenishty A, Blinder P, Sagi I. Demystifying the extracellular matrix and its proteolytic remodeling in the brain: structural and functional insights. Cell Mol Life Sci 2019; 76:3229-3248. [PMID: 31197404 PMCID: PMC11105229 DOI: 10.1007/s00018-019-03182-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 12/29/2022]
Abstract
The extracellular matrix (ECM) plays diverse roles in several physiological and pathological conditions. In the brain, the ECM is unique both in its composition and in functions. Furthermore, almost all the cells in the central nervous system contribute to different aspects of this intricate structure. Brain ECM, enriched with proteoglycans and other small proteins, aggregate into distinct structures around neurons and oligodendrocytes. These special structures have cardinal functions in the normal functioning of the brain, such as learning, memory, and synapse regulation. In this review, we have compiled the current knowledge about the structure and function of important ECM molecules in the brain and their proteolytic remodeling by matrix metalloproteinases and other enzymes, highlighting the special structures they form. In particular, the proteoglycans in brain ECM, which are essential for several vital functions, are emphasized in detail.
Collapse
Affiliation(s)
| | - Amit Benbenishty
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Pablo Blinder
- Neurobiology, Biochemistry and Biophysics School, Tel Aviv University, Tel Aviv, Israel
- Sagol School for Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
14
|
Beroun A, Mitra S, Michaluk P, Pijet B, Stefaniuk M, Kaczmarek L. MMPs in learning and memory and neuropsychiatric disorders. Cell Mol Life Sci 2019; 76:3207-3228. [PMID: 31172215 PMCID: PMC6647627 DOI: 10.1007/s00018-019-03180-8] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 05/27/2019] [Accepted: 05/29/2019] [Indexed: 12/20/2022]
Abstract
Matrix metalloproteinases (MMPs) are a group of over twenty proteases, operating chiefly extracellularly to cleave components of the extracellular matrix, cell adhesion molecules as well as cytokines and growth factors. By virtue of their expression and activity patterns in animal models and clinical investigations, as well as functional studies with gene knockouts and enzyme inhibitors, MMPs have been demonstrated to play a paramount role in many physiological and pathological processes in the brain. In particular, they have been shown to influence learning and memory processes, as well as major neuropsychiatric disorders such as schizophrenia, various kinds of addiction, epilepsy, fragile X syndrome, and depression. A possible link connecting all those conditions is either physiological or aberrant synaptic plasticity where some MMPs, e.g., MMP-9, have been demonstrated to contribute to the structural and functional reorganization of excitatory synapses that are located on dendritic spines. Another common theme linking the aforementioned pathological conditions is neuroinflammation and MMPs have also been shown to be important mediators of immune responses.
Collapse
Affiliation(s)
- Anna Beroun
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland
| | | | - Piotr Michaluk
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland
| | - Barbara Pijet
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland
| | | | - Leszek Kaczmarek
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland.
| |
Collapse
|
15
|
Alaiyed S, Conant K. A Role for Matrix Metalloproteases in Antidepressant Efficacy. Front Mol Neurosci 2019; 12:117. [PMID: 31133801 PMCID: PMC6517485 DOI: 10.3389/fnmol.2019.00117] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 04/24/2019] [Indexed: 01/10/2023] Open
Abstract
Major depressive disorder is a debilitating condition that affects approximately 15% of the United States population. Though the neurophysiological mechanisms that underlie this disorder are not completely understood, both human and rodent studies suggest that excitatory/inhibitory (E/I) balance is reduced with the depressive phenotype. In contrast, antidepressant efficacy in responsive individuals correlates with increased excitatory neurotransmission in select brain regions, suggesting that the restoration of E/I balance may improve mood. Enhanced excitatory transmission can occur through mechanisms including increased dendritic arborization and synapse formation in pyramidal neurons. Reduced activity of inhibitory neurons may also contribute to antidepressant efficacy. Consistent with this possibility, the fast-acting antidepressant ketamine may act by selective inhibition of glutamatergic input to GABA releasing parvalbumin (PV)-expressing interneurons. Recent work has also shown that a negative allosteric modulator of the GABA-A receptor α subunit can improve depression-related behavior. PV-expressing interneurons are thought to represent critical pacemakers for synchronous network events. These neurons also represent the predominant GABAergic neuronal population that is enveloped by the perineuronal net (PNN), a lattice-like structure that is thought to stabilize glutamatergic input to this cell type. Disruption of the PNN reduces PV excitability and increases pyramidal cell excitability. Various antidepressant medications increase the expression of matrix metalloproteinases (MMPs), enzymes that can increase pyramidal cell dendritic arborization and spine formation. MMPs can also cleave PNN proteins to reduce PV neuron-mediated inhibition. The present review will focus on mechanisms that may underlie antidepressant efficacy, with a focus on monoamines as facilitators of increased matrix metalloprotease (MMP) expression and activation. Discussion will include MMP-dependent effects on pyramidal cell structure and function, as well as MMP-dependent effects on PV expressing interneurons. We conclude with discussion of antidepressant use for those at risk for Alzheimer’s disease, and we also highlight areas for further study.
Collapse
Affiliation(s)
- Seham Alaiyed
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC, United States
| | - Katherine Conant
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
16
|
Semelakova M, Grauzam S, Betadthunga P, Tiedeken J, Coaxum S, Neskey DM, Rosenzweig SA. Vimentin and Non-Muscle Myosin IIA are Members of the Neural Precursor Cell Expressed Developmentally Down-Regulated 9 (NEDD9) Interactome in Head and Neck Squamous Cell Carcinoma Cells. Transl Oncol 2019; 12:49-61. [PMID: 30267961 PMCID: PMC6160858 DOI: 10.1016/j.tranon.2018.09.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 09/08/2018] [Accepted: 09/08/2018] [Indexed: 12/11/2022] Open
Abstract
Here we demonstrate an interaction between neural precursor cell expressed, developmentally-downregulated 9 (NEDD9) and the cytoskeletal proteins vimentin and non-muscle myosin IIA (NMIIA), based on co-immunoprecipitation and mass spectrometric sequence identification. Vimentin was constitutively phosphorylated at Ser56 but vimentin associated with NEDD9-was not phosphorylated at Ser56. In contrast, NMIIA bound to NEDD9 was phosphorylated on S1943 consistent with its function in invasion and secretion. Treatment of cells with the vimentin-targeting steroidal lactone withaferin A had no effect on vimentin turnover as previously reported, instead causing NEDD9 cleavage and cell death. The NMIIA-selective inhibitor blebbistatin induced cells to form long extensions and attenuated secretion of matrix metalloproteinases (MMPs) 2 and 9. While the site of vimentin interaction on NEDD9 was not defined, NMIIA was found to interact with NEDD9 at its substrate domain. NEDD9 interactions with vimentin and NMIIA are consistent with these proteins having roles in MMP secretion and cell invasion. These findings suggest that a better understanding of NEDD9 signaling is likely to reveal novel therapeutic targets for the prevention of invasion and metastasis.
Collapse
Affiliation(s)
- Martina Semelakova
- Institute of Biology and Ecology, Department of Cell Biology, Faculty of Science, Pavol Jozef Šafárik University, Košice, Slovakia; Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue MSC 509, Charleston, SC 29425-5050
| | - Stèphane Grauzam
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue MSC 509, Charleston, SC 29425-5050
| | - Prabhakar Betadthunga
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue MSC 509, Charleston, SC 29425-5050; Department of Post Graduate-Studies and Research in Biotechnology, Sahydri Science College, Kuvempu University, Shimoga, Karnataka, India, 577203
| | - Jessica Tiedeken
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue MSC 509, Charleston, SC 29425-5050
| | - Sonya Coaxum
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue MSC 509, Charleston, SC 29425-5050; Department of Otolaryngology, Head and Neck Surgery, Medical University of South Carolina
| | - David M Neskey
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue MSC 509, Charleston, SC 29425-5050; Department of Otolaryngology, Head and Neck Surgery, Medical University of South Carolina; Hollings Cancer Center, Medical University of South Carolina, 173 Ashley Avenue MSC 550, Charleston, SC 29425-5050
| | - Steven A Rosenzweig
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue MSC 509, Charleston, SC 29425-5050; Hollings Cancer Center, Medical University of South Carolina, 173 Ashley Avenue MSC 550, Charleston, SC 29425-5050.
| |
Collapse
|
17
|
Paumier JM, Py NA, García-González L, Bernard A, Stephan D, Louis L, Checler F, Khrestchatisky M, Baranger K, Rivera S. Proamyloidogenic effects of membrane type 1 matrix metalloproteinase involve MMP‐2 and BACE‐1 activities, and the modulation of APP trafficking. FASEB J 2018; 33:2910-2927. [DOI: 10.1096/fj.201801076r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
| | - Nathalie A. Py
- Aix-Marseille UnivCNRSINPInst NeurophysiopatholMarseilleFrance
| | | | - Anne Bernard
- Aix-Marseille UnivCNRSINPInst NeurophysiopatholMarseilleFrance
| | | | - Laurence Louis
- Aix-Marseille UnivCNRSINPInst NeurophysiopatholMarseilleFrance
| | - Frédéric Checler
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC)Unité Mixte de Recherche (UMR) 7275 CNRS–Université Nice Sophia (UNS)Excellence Laboratory (Labex) Development of Innovaive Strategies for a Transdisciplinary Approach to Alzheimer's Disease (DistAlz)ValbonneFrance
| | | | - Kévin Baranger
- Aix-Marseille UnivCNRSINPInst NeurophysiopatholMarseilleFrance
| | - Santiago Rivera
- Aix-Marseille UnivCNRSINPInst NeurophysiopatholMarseilleFrance
| |
Collapse
|
18
|
The passive avoidance memory improving effect of curcumin in young adult mice: Considering hippocampal MMP-2, MMP-9 and Akt/GSK3β. PHARMANUTRITION 2018. [DOI: 10.1016/j.phanu.2018.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
19
|
NEDD9 stimulated MMP9 secretion is required for invadopodia formation in oral squamous cell carcinoma. Oncotarget 2018; 9:25503-25516. [PMID: 29876004 PMCID: PMC5986644 DOI: 10.18632/oncotarget.25347] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/24/2018] [Indexed: 12/19/2022] Open
Abstract
Neural precursor cell expressed developmentally downregulated 9 (NEDD9) is a component of the metastatic signatures of melanoma, breast cancer, glioblastoma, lung cancer and head and neck squamous cell carcinoma (HNSCC). Here we tested the efficacy of NEDD9's domains in stimulating matrix metalloproteinase (MMP) secretion and invadopodia formation in cells stably expressing various NEDD9 mutants. Replacement of the 13 YxxP motif substrate domain (SD) tyrosines and the C-terminal Y629 with phenylalanines (F14NEDD9) eliminated tyrosine phosphorylation, MMP9 secretion and loss of invadopodia formation. Mutation of the N-terminal SH3 domain Y12 to glutamic acid (Y12ENEDD9) or phenylalanine (Y12FNEDD9) reduced MMP9 secretion and inhibited invadopodia formation. SH3 domain deletion (∆SH3NEDD9) resulted in the loss of MMP9 secretion and a lack of invadopodia formation. The SH3–SD domain (SSNEDD9) construct exhibited tyrosine phosphorylation and stimulated MMP9 secretion, as did ∆CTNEDD9 which lacked the C-terminus (∆C-terminal; ∆CT). E13NEDD9 expression blocked MMP9 secretion and invadopodia formation. MICAL1 (molecule interacting with Cas-L1) silencing with a short hairpin RNA reduced MMP9 secretion, vimentin and E-cadherin levels while increasing N-cadherin and Rab6 levels, consistent with reduced invasive behavior. These findings indicate that NEDD9 SD phosphorylation and SH3 domain interactions are necessary for increasing MMP9 secretion and invadopodia formation.
Collapse
|
20
|
Aguayo FI, Pacheco AA, García-Rojo GJ, Pizarro-Bauerle JA, Doberti AV, Tejos M, García-Pérez MA, Rojas PS, Fiedler JL. Matrix Metalloproteinase 9 Displays a Particular Time Response to Acute Stress: Variation in Its Levels and Activity Distribution in Rat Hippocampus. ACS Chem Neurosci 2018; 9:945-956. [PMID: 29361213 DOI: 10.1021/acschemneuro.7b00387] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
A single stress exposure facilitates memory formation through neuroplastic processes that reshape excitatory synapses in the hippocampus, probably requiring changes in extracellular matrix components. We tested the hypothesis that matrix metalloproteinase 9 (MMP-9), an enzyme that degrades components of extracellular matrix and synaptic proteins such as β-dystroglycan (β-DG43), changes their activity and distribution in rat hippocampus during the acute stress response. After 2.5 h of restraint stress, we found (i) increased MMP-9 levels and potential activity in whole hippocampal extracts, accompanied by β-DG43 cleavage, and (ii) a significant enhancement of MMP-9 immunoreactivity in dendritic fields such as stratum radiatum and the molecular layer of hippocampus. After 24 h of stress, we found that (i) MMP-9 net activity rises at somatic field, i.e., stratum pyramidale and granule cell layers, and also at synaptic field, mainly stratum radiatum and the molecular layer of hippocampus, and (ii) hippocampal synaptoneurosome fractions are enriched with MMP-9, without variation of its potential enzymatic activity, in accordance with the constant level of cleaved β-DG43. These findings indicate that stress triggers a peculiar timing response in the MMP-9 levels, net activity, and subcellular distribution in the hippocampus, suggesting its involvement in the processing of substrates during the stress response.
Collapse
Affiliation(s)
- Felipe I. Aguayo
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Aníbal A. Pacheco
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Gonzalo J. García-Rojo
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Javier A. Pizarro-Bauerle
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Ana V. Doberti
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Macarena Tejos
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - María A. García-Pérez
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Paulina S. Rojas
- Escuela de Quı́mica y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Jenny L. Fiedler
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| |
Collapse
|
21
|
Endres M, Kneitz S, Orth MF, Perera RK, Zernecke A, Butt E. Regulation of matrix metalloproteinases (MMPs) expression and secretion in MDA-MB-231 breast cancer cells by LIM and SH3 protein 1 (LASP1). Oncotarget 2018; 7:64244-64259. [PMID: 27588391 PMCID: PMC5325439 DOI: 10.18632/oncotarget.11720] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 08/09/2016] [Indexed: 01/27/2023] Open
Abstract
The process of tumor invasion requires degradation of extracellular matrix by proteolytic enzymes. Cancer cells form protrusive invadopodia, which produce and release matrix metalloproteinases (MMPs) to degrade the basement membrane thereby enabling metastasis. We investigated the effect of LASP1, a newly identified protein in invadopodia, on expression, secretion and activation of MMPs in invasive breast tumor cell lines. By analyzing microarray data of in-house generated control and LASP1-depleted MDA-MB-231 breast cancer cells, we observed downregulation of MMP1, -3 and -9 upon LASP1 depletion. This was confirmed by Western blot analysis. Conversely, rescue experiments restored in part MMP expression and secretion. The regulatory effect of LASP1 on MMP expression was also observed in BT-20 breast cancer cells as well as in prostate and bladder cancer cell lines. In line with bioinformatic FunRich analysis of our data, which mapped a high regulation of transcription factors by LASP1, public microarray data analysis detected a correlation between high LASP1 expression and enhanced c-Fos levels, a protein that is part of the transcription factor AP-1 and known to regulate MMP expression. Compatibly, in luciferase reporter assays, AP-1 showed a decreased transcriptional activity after LASP1 knockdown. Zymography assays and Western blot analysis revealed an additional promotion of MMP secretion into the extracellular matrix by LASP1, thus, most likely, altering the microenvironment during cancer progression. The newly identified role of LASP1 in regulating matrix degradation by affecting MMP transcription and secretion elucidated the migratory potential of LASP1 overexpressing aggressive tumor cells in earlier studies.
Collapse
Affiliation(s)
- Marcel Endres
- Institute of Experimental Biomedicine II, University Medical Clinic of Wuerzburg, Wuerzburg, Germany
| | - Susanne Kneitz
- Physiological Chemistry, Biozentrum, University of Wuerzburg, Am Hubland, Wuerzburg, Germany
| | - Martin F Orth
- Institute of Experimental Biomedicine II, University Medical Clinic of Wuerzburg, Wuerzburg, Germany
| | - Ruwan K Perera
- Institute of Experimental Biomedicine II, University Medical Clinic of Wuerzburg, Wuerzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine II, University Medical Clinic of Wuerzburg, Wuerzburg, Germany
| | - Elke Butt
- Institute of Experimental Biomedicine II, University Medical Clinic of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
22
|
Cerebellum Susceptibility to Neonatal Asphyxia: Possible Protective Effects of N-Acetylcysteine Amide. DISEASE MARKERS 2018; 2018:5046372. [PMID: 29651324 PMCID: PMC5831588 DOI: 10.1155/2018/5046372] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 09/07/2017] [Accepted: 12/07/2017] [Indexed: 12/22/2022]
Abstract
Background After perinatal asphyxia, the cerebellum presents more damage than previously suggested. Objectives To explore if the antioxidant N-acetylcysteine amide (NACA) could reduce cerebellar injury after hypoxia-reoxygenation in a neonatal pig model. Methods Twenty-four newborn pigs in two intervention groups were exposed to 8% oxygen and hypercapnia, until base excess fell to -20 mmol/l or the mean arterial blood pressure declined to <20 mmHg. After hypoxia, they received either NACA (NACA group, n = 12) or saline (vehicle-treated group, n = 12). One sham-operated group (n = 5) served as a control and was not subjected to hypoxia. Observation time after the end of hypoxia was 9.5 hours. Results The intranuclear proteolytic activity in Purkinje cells of asphyxiated vehicle-treated pigs was significantly higher than that in sham controls (p = 0.03). Treatment with NACA was associated with a trend to decreased intranuclear proteolytic activity (p = 0.08), There were significantly less mutations in the mtDNA of the NACA group compared with the vehicle-treated group, 2.0 × 10-4 (±2.0 × 10-4) versus 4.8 × 10-5(±3.6 × 10-4, p < 0.05). Conclusion We found a trend to lower proteolytic activity in the core of Purkinje cells and significantly reduced mutation rate of mtDNA in the NACA group, which may indicate a positive effect of NACA after neonatal hypoxia. Measuring the proteolytic activity in the nucleus of Purkinje cells could be used to assess the effect of different neuroprotective substances after perinatal asphyxia.
Collapse
|
23
|
Pinto S, Cunha C, Barbosa M, Vaz AR, Brites D. Exosomes from NSC-34 Cells Transfected with hSOD1-G93A Are Enriched in miR-124 and Drive Alterations in Microglia Phenotype. Front Neurosci 2017; 11:273. [PMID: 28567000 PMCID: PMC5434170 DOI: 10.3389/fnins.2017.00273] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/28/2017] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal adult-onset neurodegenerative disorder affecting motor neurons (MNs). Evidences indicate that ALS is a non-cell autonomous disease in which glial cells participate in both disease onset and progression. Exosomal transfer of mutant copper-zinc superoxide dismutase 1 (mSOD1) from cell-to-cell was suggested to contribute to disease dissemination. Data from our group and others showed that exosomes from activated cells contain inflammatory-related microRNAs (inflamma-miRNAs) that recapitulate the donor cell. While glia-derived exosomes and their effects in neurons have been addressed by several studies, only a few investigated the influence of motor neuron (MN)-derived exosomes in other cell function, the aim of the present study. We assessed a set of inflamma-miRs in NSC-34 MN-like cells transfected with mutant SOD1(G93A) and extended the study into their derived exosomes (mSOD1 exosomes). Then, the effects produced by mSOD1 exosomes in the activation and polarization of the recipient N9 microglial cells were investigated. Exosomes in coculture with N9 microglia and NSC-34 cells [either transfected with either wild-type (wt) human SOD1 or mutant SOD1(G93A)] showed to be transferred into N9 cells. Increased miR-124 expression was found in mSOD1 NSC-34 cells and in their derived exosomes. Incubation of mSOD1 exosomes with N9 cells determined a sustained 50% reduction in the cell phagocytic ability. It also caused a persistent NF-kB activation and an acute generation of NO, MMP-2, and MMP-9 activation, as well as upregulation of IL-1β, TNF-α, MHC-II, and iNOS gene expression, suggestive of induced M1 polarization. Marked elevation of IL-10, Arginase 1, TREM2, RAGE, and TLR4 mRNA levels, together with increased miR-124, miR-146a, and miR-155, at 24 h incubation, suggest the switch to mixed M1 and M2 subpopulations in the exosome-treated N9 microglial cells. Exosomes from mSOD1 NSC-34 MNs also enhanced the number of senescent-like positive N9 cells. Data suggest that miR-124 is translocated from the mSOD1 MNs to exosomes, which determine early and late phenotypic alterations in the recipient N9-microglial cells. In conclusion, modulation of the inflammatory-associated miR-124, in mSOD1 NSC-34 MNs, with potential benefits in the cargo of their exosomes may reveal a promising therapeutic strategy in halting microglia activation and associated effects in MN degeneration.
Collapse
Affiliation(s)
- Sara Pinto
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Carolina Cunha
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Marta Barbosa
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Ana R Vaz
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Dora Brites
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| |
Collapse
|
24
|
Sun J, Wang D, Guo L, Fang S, Wang Y, Xing R. Androgen Receptor Regulates the Growth of Neuroblastoma Cells in vitro and in vivo. Front Neurosci 2017; 11:116. [PMID: 28326012 PMCID: PMC5339338 DOI: 10.3389/fnins.2017.00116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 02/23/2017] [Indexed: 12/18/2022] Open
Abstract
Background: Neuroblastoma is the most common extracranial tumors in children. At present about the true etiology of neuroblastoma is unclear and many studies have tried to find effective treatments for these primary malignant tumors. Although it has been illustrated that androgen receptor (AR) was expressed in neuroblastoma cells in some former reports, the biological role of androgen receptor in the development of neuroblastoma is not fully understood. Methods: Androgen (R1881) and the antagonists of androgen receptor (MDV3100 and ARN509) were used to study the role of the androgen receptor signaling pathway in vitro and in vivo on SH-SY5Y and Neuro-2a (N2a) cell lines. Results: We found that AR expression showed an R1881 dose-dependent manner in neuroblastoma cells in vitro and R1881was able to increase, while both antagonists of androgen receptor (MDV3100 and ARN509) significantly decrease, the proliferation, migration, invasion and sphere formation of SH-SY5Y and N2a cells. Moreover, androgen promoted the growth of N2a tumor in vivo. However, when androgen receptor (AR) was effectively knocked down in the two cell lines by siRNA, either promoting or inhibiting effect of the androgen or androgen receptor antagonists, respectively, was attenuated. Conclusion: Our results suggested that androgen receptor may involve in the progression of neuroblastoma as well as provided insight into a new target for the diagnosis and treatment of neuroblastoma patients.
Collapse
Affiliation(s)
- Junyan Sun
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical UniversityDalian, China; Department of Experimental Functionality, College of Basic Medical SciencesDalian, China
| | - Dongmei Wang
- College of Integrative Medicine, Dalian Medical University Dalian, China
| | - Lianying Guo
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical University Dalian, China
| | - Shengyun Fang
- Center for Biomedical Engineering and Technology, Department of Physiology, Department of Biochemistry and Molecular Biology, University of Maryland, School of Medicine Baltimore, MD, USA
| | - Yang Wang
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical University Dalian, China
| | - Rong Xing
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical University Dalian, China
| |
Collapse
|
25
|
Baranger K, Bonnet AE, Girard SD, Paumier JM, García-González L, Elmanaa W, Bernard A, Charrat E, Stephan D, Bauer C, Moschke K, Lichtenthaler SF, Roman FS, Checler F, Khrestchatisky M, Rivera S. MT5-MMP Promotes Alzheimer's Pathogenesis in the Frontal Cortex of 5xFAD Mice and APP Trafficking in vitro. Front Mol Neurosci 2017; 9:163. [PMID: 28119565 PMCID: PMC5223243 DOI: 10.3389/fnmol.2016.00163] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 12/16/2016] [Indexed: 12/18/2022] Open
Abstract
We previously reported that deficiency of membrane-type five matrix metalloproteinase (MT5-MMP) prevents amyloid pathology in the cortex and hippocampus of 5xFAD mice, and ameliorates the functional outcome. We have now investigated whether the integrity of another important area affected in Alzheimer's disease (AD), the frontal cortex, was also preserved upon MT5-MMP deficiency in 4-month old mice at prodromal stages of the pathology. We used the olfactory H-maze (OHM) to show that learning impairment associated with dysfunctions of the frontal cortex in 5xFAD was prevented in bigenic 5xFAD/MT5-MMP-/- mice. The latter exhibited concomitant drastic reductions of amyloid beta peptide (Aβ) assemblies (soluble, oligomeric and fibrillary) and its immediate precursor, C99. Simultaneously, astrocyte reactivity and tumor necrosis factor alpha (TNF-α) levels were also lowered. Moreover, MT5-MMP deficiency induced a decrease in N-terminal soluble fragments of amyloid precursor protein (APP), including soluble APPα (sAPPα), sAPPβ and the MT5-MMP-linked fragment of 95 kDa, sAPP95. However, the lack of MT5-MMP did not affect the activity of β- and γ-secretases. In cultured HEKswe cells, transiently expressed MT5-MMP localized to early endosomes and increased the content of APP and Aβ40 in these organelles, as well as Aβ levels in cell supernatants. This is the first evidence that the pro-amyloidogenic features of MT5-MMP lie, at least in part, on the ability of the proteinase to promote trafficking into one of the amyloidogenic subcellular loci. Together, our data further support the pathogenic role of MT5-MMP in AD and that its inhibition improves the functional and pathological outcomes, in this case in the frontal cortex. These data also support the idea that MT5-MMP could become a novel therapeutic target in AD.
Collapse
Affiliation(s)
- Kévin Baranger
- Aix Marseille Université, CNRS, NICN UMR 7259 Marseille, France
| | | | | | | | | | - Wejdane Elmanaa
- Université Côte d'Azur, INSERM, CNRS, IPMC, Laboratory of excellence DistALZ, Sophia-Antipolis Valbonne, France
| | - Anne Bernard
- Aix Marseille Université, CNRS, NICN UMR 7259 Marseille, France
| | - Eliane Charrat
- Aix Marseille Université, CNRS, NICN UMR 7259 Marseille, France
| | | | - Charlotte Bauer
- Université Côte d'Azur, INSERM, CNRS, IPMC, Laboratory of excellence DistALZ, Sophia-Antipolis Valbonne, France
| | - Katrin Moschke
- German Center for Neurodegenerative Diseases (DZNE) Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE)Munich, Germany; Neuroproteomics, Klinikum rechts der Isar, and Institute for Advanced Study, Technische Universität München (TUM)Munich, Germany; Munich Cluster for Systems Neurology (SyNergy)Munich, Germany
| | | | - Frédéric Checler
- Université Côte d'Azur, INSERM, CNRS, IPMC, Laboratory of excellence DistALZ, Sophia-Antipolis Valbonne, France
| | | | - Santiago Rivera
- Aix Marseille Université, CNRS, NICN UMR 7259 Marseille, France
| |
Collapse
|
26
|
Padamsey Z, McGuinness L, Bardo SJ, Reinhart M, Tong R, Hedegaard A, Hart ML, Emptage NJ. Activity-Dependent Exocytosis of Lysosomes Regulates the Structural Plasticity of Dendritic Spines. Neuron 2016; 93:132-146. [PMID: 27989455 PMCID: PMC5222721 DOI: 10.1016/j.neuron.2016.11.013] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 09/02/2016] [Accepted: 10/31/2016] [Indexed: 11/28/2022]
Abstract
Lysosomes have traditionally been viewed as degradative organelles, although a growing body of evidence suggests that they can function as Ca2+ stores. Here we examined the function of these stores in hippocampal pyramidal neurons. We found that back-propagating action potentials (bpAPs) could elicit Ca2+ release from lysosomes in the dendrites. This Ca2+ release triggered the fusion of lysosomes with the plasma membrane, resulting in the release of Cathepsin B. Cathepsin B increased the activity of matrix metalloproteinase 9 (MMP-9), an enzyme involved in extracellular matrix (ECM) remodelling and synaptic plasticity. Inhibition of either lysosomal Ca2+ signaling or Cathepsin B release prevented the maintenance of dendritic spine growth induced by Hebbian activity. This impairment could be rescued by exogenous application of active MMP-9. Our findings suggest that activity-dependent exocytosis of Cathepsin B from lysosomes regulates the long-term structural plasticity of dendritic spines by triggering MMP-9 activation and ECM remodelling. Back-propagating action potentials induce Ca2+ release from lysosomes in neurons Lysosomal Ca2+ release triggers exocytosis of the lysosomal protease Cathepsin B Cathepsin B maintains activity-dependent dendritic spine growth by activating MMP-9
Collapse
Affiliation(s)
- Zahid Padamsey
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Lindsay McGuinness
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Scott J Bardo
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Marcia Reinhart
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Rudi Tong
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Anne Hedegaard
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Michael L Hart
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Nigel J Emptage
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK.
| |
Collapse
|
27
|
Lepeta K, Lourenco MV, Schweitzer BC, Martino Adami PV, Banerjee P, Catuara-Solarz S, de La Fuente Revenga M, Guillem AM, Haidar M, Ijomone OM, Nadorp B, Qi L, Perera ND, Refsgaard LK, Reid KM, Sabbar M, Sahoo A, Schaefer N, Sheean RK, Suska A, Verma R, Vicidomini C, Wright D, Zhang XD, Seidenbecher C. Synaptopathies: synaptic dysfunction in neurological disorders - A review from students to students. J Neurochem 2016; 138:785-805. [PMID: 27333343 PMCID: PMC5095804 DOI: 10.1111/jnc.13713] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/03/2016] [Accepted: 06/06/2016] [Indexed: 12/12/2022]
Abstract
Synapses are essential components of neurons and allow information to travel coordinately throughout the nervous system to adjust behavior to environmental stimuli and to control body functions, memories, and emotions. Thus, optimal synaptic communication is required for proper brain physiology, and slight perturbations of synapse function can lead to brain disorders. In fact, increasing evidence has demonstrated the relevance of synapse dysfunction as a major determinant of many neurological diseases. This notion has led to the concept of synaptopathies as brain diseases with synapse defects as shared pathogenic features. In this review, which was initiated at the 13th International Society for Neurochemistry Advanced School, we discuss basic concepts of synapse structure and function, and provide a critical view of how aberrant synapse physiology may contribute to neurodevelopmental disorders (autism, Down syndrome, startle disease, and epilepsy) as well as neurodegenerative disorders (Alzheimer and Parkinson disease). We finally discuss the appropriateness and potential implications of gathering synapse diseases under a single term. Understanding common causes and intrinsic differences in disease-associated synaptic dysfunction could offer novel clues toward synapse-based therapeutic intervention for neurological and neuropsychiatric disorders. In this Review, which was initiated at the 13th International Society for Neurochemistry (ISN) Advanced School, we discuss basic concepts of synapse structure and function, and provide a critical view of how aberrant synapse physiology may contribute to neurodevelopmental (autism, Down syndrome, startle disease, and epilepsy) as well as neurodegenerative disorders (Alzheimer's and Parkinson's diseases), gathered together under the term of synaptopathies. Read the Editorial Highlight for this article on page 783.
Collapse
Affiliation(s)
- Katarzyna Lepeta
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Barbara C Schweitzer
- Department for Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany
| | - Pamela V Martino Adami
- Laboratory of Amyloidosis and Neurodegeneration, Fundación Instituto Leloir-IIBBA-CONICET, Buenos Aires, Argentina
| | - Priyanjalee Banerjee
- Department of Biochemistry, Institute of Post Graduate Medical Education & Research, Kolkata, West Bengal, India
| | - Silvina Catuara-Solarz
- Systems Biology Program, Cellular and Systems Neurobiology, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Mario de La Fuente Revenga
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Alain Marc Guillem
- Laboratorio de Neurotoxicología, Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México D.F. 07000, Mexico
| | - Mouna Haidar
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Omamuyovwi M Ijomone
- Department of Human Anatomy, Cross River University of Technology, Okuku Campus, Cross River, Nigeria
| | - Bettina Nadorp
- The Department of Biological Chemistry, The Edmond and Lily Safra Center for Brain Sciences, The Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Israel
| | - Lin Qi
- Laboratory of Molecular Neuro-Oncology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, United States of America
| | - Nirma D Perera
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Louise K Refsgaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Kimberley M Reid
- Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Mariam Sabbar
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Arghyadip Sahoo
- Department of Biochemistry, Midnapore Medical College, West Bengal University of Health Sciences, West Bengal, India
| | - Natascha Schaefer
- Institute for Clinical Neurobiology, Julius-Maximilians-University of Wuerzburg, Wuerzburg, Germany
| | - Rebecca K Sheean
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Anna Suska
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Rajkumar Verma
- Department of Neurosciences Uconn Health Center, Farmington, CT, United States of America
| | | | - Dean Wright
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Xing-Ding Zhang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Constanze Seidenbecher
- Department for Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany. .,Center for Behavioral Brain Sciences (CBBS) Magdeburg, Magdeburg, Germany.
| |
Collapse
|
28
|
The multifaceted role of metalloproteinases in physiological and pathological conditions in embryonic and adult brains. Prog Neurobiol 2016; 155:36-56. [PMID: 27530222 DOI: 10.1016/j.pneurobio.2016.08.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 07/10/2016] [Accepted: 08/08/2016] [Indexed: 02/07/2023]
Abstract
Matrix metalloproteinases (MMPs) are a large family of ubiquitous extracellular endopeptidases, which play important roles in a variety of physiological and pathological conditions, from the embryonic stages throughout adult life. Their extraordinary physiological "success" is due to concomitant broad substrate specificities and strict regulation of their expression, activation and inhibition levels. In recent years, MMPs have gained increasing attention as significant effectors in various aspects of central nervous system (CNS) physiology. Most importantly, they have been recognized as main players in a variety of brain disorders having different etiologies and evolution. A common aspect of these pathologies is the development of acute or chronic neuroinflammation. MMPs play an integral part in determining the result of neuroinflammation, in some cases turning its beneficial outcome into a harmful one. This review summarizes the most relevant studies concerning the physiology of MMPs, highlighting their involvement in both the developing and mature CNS, in long-lasting and acute brain diseases and, finally, in nervous system repair. Recently, a concerted effort has been made in identifying therapeutic strategies for major brain diseases by targeting MMP activities. However, from this revision of the literature appears clear that MMPs have multifaceted functional characteristics, which modulate physiological processes in multiple ways and with multiple consequences. Therefore, when choosing MMPs as possible targets, great care must be taken to evaluate the delicate balance between their activation and inhibition and to determine at which stage of the disease and at what level they become active in order maximize chances of success.
Collapse
|
29
|
Azevedo A, Prado A, Issa J, Gerlach R. Matrix metalloproteinase 2 fused to GFP, expressed in E. coli, successfully tracked MMP-2 distribution in vivo. Int J Biol Macromol 2016; 89:737-45. [DOI: 10.1016/j.ijbiomac.2016.05.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 01/22/2023]
|
30
|
Magnowska M, Gorkiewicz T, Suska A, Wawrzyniak M, Rutkowska-Wlodarczyk I, Kaczmarek L, Wlodarczyk J. Transient ECM protease activity promotes synaptic plasticity. Sci Rep 2016; 6:27757. [PMID: 27282248 PMCID: PMC4901294 DOI: 10.1038/srep27757] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 05/25/2016] [Indexed: 12/20/2022] Open
Abstract
Activity-dependent proteolysis at a synapse has been recognized as a pivotal factor in controlling dynamic changes in dendritic spine shape and function; however, excessive proteolytic activity is detrimental to the cells. The exact mechanism of control of these seemingly contradictory outcomes of protease activity remains unknown. Here, we reveal that dendritic spine maturation is strictly controlled by the proteolytic activity, and its inhibition by the endogenous inhibitor (Tissue inhibitor of matrix metalloproteinases-1 – TIMP-1). Excessive proteolytic activity impairs long-term potentiation of the synaptic efficacy (LTP), and this impairment could be rescued by inhibition of protease activity. Moreover LTP is altered persistently when the ability of TIMP-1 to inhibit protease activity is abrogated, further demonstrating the role of such inhibition in the promotion of synaptic plasticity under well-defined conditions. We also show that dendritic spine maturation involves an intermediate formation of elongated spines, followed by their conversion into mushroom shape. The formation of mushroom-shaped spines is accompanied by increase in AMPA/NMDA ratio of glutamate receptors. Altogether, our results identify inhibition of protease activity as a critical regulatory mechanism for dendritic spines maturation.
Collapse
Affiliation(s)
- Marta Magnowska
- Department of Molecular and Cellular Neurobiology, Nencki Institute, Pasteura 3, Warsaw, 02-093, Poland
| | - Tomasz Gorkiewicz
- Department of Neurophysiology, Nencki Institute, Pasteura 3, Warsaw, 02-093, Poland.,Department of Physics, Warsaw University of Life Sciences, Nowoursynowska 159, Warsaw, 02-776, Poland
| | - Anna Suska
- Department of Molecular and Cellular Neurobiology, Nencki Institute, Pasteura 3, Warsaw, 02-093, Poland
| | - Marcin Wawrzyniak
- Department of Molecular and Cellular Neurobiology, Nencki Institute, Pasteura 3, Warsaw, 02-093, Poland
| | | | - Leszek Kaczmarek
- Department of Molecular and Cellular Neurobiology, Nencki Institute, Pasteura 3, Warsaw, 02-093, Poland
| | - Jakub Wlodarczyk
- Department of Molecular and Cellular Neurobiology, Nencki Institute, Pasteura 3, Warsaw, 02-093, Poland
| |
Collapse
|
31
|
Baranger K, Marchalant Y, Bonnet AE, Crouzin N, Carrete A, Paumier JM, Py NA, Bernard A, Bauer C, Charrat E, Moschke K, Seiki M, Vignes M, Lichtenthaler SF, Checler F, Khrestchatisky M, Rivera S. MT5-MMP is a new pro-amyloidogenic proteinase that promotes amyloid pathology and cognitive decline in a transgenic mouse model of Alzheimer's disease. Cell Mol Life Sci 2016; 73:217-36. [PMID: 26202697 PMCID: PMC4700096 DOI: 10.1007/s00018-015-1992-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/08/2015] [Accepted: 07/10/2015] [Indexed: 01/22/2023]
Abstract
Membrane-type 5-matrix metalloproteinase (MT5-MMP) is a proteinase mainly expressed in the nervous system with emerging roles in brain pathophysiology. The implication of MT5-MMP in Alzheimer's disease (AD), notably its interplay with the amyloidogenic process, remains elusive. Accordingly, we crossed the genetically engineered 5xFAD mouse model of AD with MT5-MMP-deficient mice and examined the impact of MT5-MMP deficiency in bigenic 5xFAD/MT5-MMP(-/-) mice. At early stages (4 months) of the pathology, the levels of amyloid beta peptide (Aβ) and its amyloid precursor protein (APP) C-terminal fragment C99 were largely reduced in the cortex and hippocampus of 5xFAD/MT5-MMP(-/-), compared to 5xFAD mice. Reduced amyloidosis in bigenic mice was concomitant with decreased glial reactivity and interleukin-1β (IL-1β) levels, and the preservation of long-term potentiation (LTP) and spatial learning, without changes in the activity of α-, β- and γ-secretases. The positive impact of MT5-MMP deficiency was still noticeable at 16 months of age, as illustrated by reduced amyloid burden and gliosis, and a better preservation of the cortical neuronal network and synaptophysin levels in bigenic mice. MT5-MMP expressed in HEKswe cells colocalized and co-immunoprecipitated with APP and significantly increased the levels of Aβ and C99. MT5-MMP also promoted the release of a soluble APP fragment of 95 kDa (sAPP95) in HEKswe cells. sAPP95 levels were significantly reduced in brain homogenates of 5xFAD/MT5-MMP(-/-) mice, supporting altogether the idea that MT5-MMP influences APP processing. MT5-MMP emerges as a new pro-amyloidogenic regulator of APP metabolism, whose deficiency alleviates amyloid pathology, neuroinflammation and cognitive decline.
Collapse
Affiliation(s)
- Kévin Baranger
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Yannick Marchalant
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
- Psychology Department, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Amandine E Bonnet
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Nadine Crouzin
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Alex Carrete
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | | | - Nathalie A Py
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Anne Bernard
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Charlotte Bauer
- Labex DistAlz, IPMC UMR 7275 CNRS-UNS, 06560, Valbonne, France
| | - Eliane Charrat
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Katrin Moschke
- German Center for Neurodegenerative Diseases (DZNE) and Neuroproteomics, Munich, Germany
- Klinikum rechts der Isar, and Institute for Advanced Study, Technische Universität München (TUM), 81675, Munich, Germany
| | - Mothoharu Seiki
- Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Michel Vignes
- UMR5247 IBMM CNRS University of Montpellier 1 and University of Montpellier 2, 34095, Montepellier, France
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE) and Neuroproteomics, Munich, Germany
- Klinikum rechts der Isar, and Institute for Advanced Study, Technische Universität München (TUM), 81675, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 80336, Munich, Germany
| | | | | | - Santiago Rivera
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France.
| |
Collapse
|
32
|
Wiera G, Mozrzymas JW. Extracellular proteolysis in structural and functional plasticity of mossy fiber synapses in hippocampus. Front Cell Neurosci 2015; 9:427. [PMID: 26582976 PMCID: PMC4631828 DOI: 10.3389/fncel.2015.00427] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/09/2015] [Indexed: 02/04/2023] Open
Abstract
Brain is continuously altered in response to experience and environmental changes. One of the underlying mechanisms is synaptic plasticity, which is manifested by modification of synapse structure and function. It is becoming clear that regulated extracellular proteolysis plays a pivotal role in the structural and functional remodeling of synapses during brain development, learning and memory formation. Clearly, plasticity mechanisms may substantially differ between projections. Mossy fiber synapses onto CA3 pyramidal cells display several unique functional features, including pronounced short-term facilitation, a presynaptically expressed long-term potentiation (LTP) that is independent of NMDAR activation, and NMDA-dependent metaplasticity. Moreover, structural plasticity at mossy fiber synapses ranges from the reorganization of projection topology after hippocampus-dependent learning, through intrinsically different dynamic properties of synaptic boutons to pre- and postsynaptic structural changes accompanying LTP induction. Although concomitant functional and structural plasticity in this pathway strongly suggests a role of extracellular proteolysis, its impact only starts to be investigated in this projection. In the present report, we review the role of extracellular proteolysis in various aspects of synaptic plasticity in hippocampal mossy fiber synapses. A growing body of evidence demonstrates that among perisynaptic proteases, tissue plasminogen activator (tPA)/plasmin system, β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) and metalloproteinases play a crucial role in shaping plastic changes in this projection. We discuss recent advances and emerging hypotheses on the roles of proteases in mechanisms underlying mossy fiber target specific synaptic plasticity and memory formation.
Collapse
Affiliation(s)
- Grzegorz Wiera
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University Wroclaw, Poland ; Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University Wroclaw, Poland
| | - Jerzy W Mozrzymas
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University Wroclaw, Poland ; Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University Wroclaw, Poland
| |
Collapse
|
33
|
Duellman T, Burnett J, Yang J. Functional Roles of N-Linked Glycosylation of Human Matrix Metalloproteinase 9. Traffic 2015. [PMID: 26207422 DOI: 10.1111/tra.12312] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Matrix metalloproteinase-9 (MMP-9) is a secreted endoproteinase with a critical role in the regulation of the extracellular matrix and proteolytic activation of signaling molecules. Human (h)MMP-9 has two well-defined N-glycosylation sites at residues N38 and N120; however, their role has remained mostly unexplored partly because expression of the N-glycosylation-deficient N38S has been difficult due to a recently discovered single nucleotide polymorphism-dependent miRNA-mediated inhibitory mechanism. hMMP-9 cDNA encoding amino acid substitutions at residues 38 (modified-S38, mS38) or 120 (N120S) were created in the background of a miRNA-binding site disrupted template and expressed by transient transfection. hMMP-9 harboring a single mS38 replacement secreted well, whereas N120S, or a double mS38/N120S hMMP-9 demonstrated much reduced secretion. Imaging indicated endoplasmic reticulum (ER) retention of the non-secreted variants and co-immunoprecipitation confirmed an enhanced strong interaction between the non-secreted hMMP-9 and the ER-resident protein calreticulin (CALR). Removal of N-glycosylation at residue 38 revealed an amino acid-dependent strong interaction with CALR likely preventing unloading of the misfolded protein from the ER chaperone down the normal secretory pathway. As with other glycoproteins, N-glycosylation strongly regulates hMMP-9 secretion. This is mediated, however, through a novel mechanism of cloaking an N-glycosylation-independent strong interaction with the ER-resident CALR.
Collapse
Affiliation(s)
- Tyler Duellman
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53705, USA.,Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - John Burnett
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jay Yang
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53705, USA.,Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| |
Collapse
|
34
|
Conant K, Allen M, Lim ST. Activity dependent CAM cleavage and neurotransmission. Front Cell Neurosci 2015; 9:305. [PMID: 26321910 PMCID: PMC4531370 DOI: 10.3389/fncel.2015.00305] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/27/2015] [Indexed: 12/13/2022] Open
Abstract
Spatially localized proteolysis represents an elegant means by which neuronal activity dependent changes in synaptic structure, and thus experience dependent learning and memory, can be achieved. In vitro and in vivo studies suggest that matrix metalloproteinase and adamalysin activity is concentrated at the cell surface, and emerging evidence suggests that increased peri-synaptic expression, release and/or activation of these proteinases occurs with enhanced excitatory neurotransmission. Synaptically expressed cell adhesion molecules (CAMs) could therefore represent important targets for neuronal activity-dependent proteolysis. Several CAM subtypes are expressed at the synapse, and their cleavage can influence the efficacy of synaptic transmission through a variety of non-mutually exclusive mechanisms. In the following review, we discuss mechanisms that regulate neuronal activity-dependent synaptic CAM shedding, including those that may be calcium dependent. We also highlight CAM targets of activity-dependent proteolysis including neuroligin and intercellular adhesion molecule-5 (ICAM-5). We include discussion focused on potential consequences of synaptic CAM shedding, with an emphasis on interactions between soluble CAM cleavage products and specific pre- and post-synaptic receptors.
Collapse
Affiliation(s)
- Katherine Conant
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center Washington, DC, USA
| | - Megan Allen
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center Washington, DC, USA
| | - Seung T Lim
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center Washington, DC, USA
| |
Collapse
|
35
|
Wójtowicz T, Brzdąk P, Mozrzymas JW. Diverse impact of acute and long-term extracellular proteolytic activity on plasticity of neuronal excitability. Front Cell Neurosci 2015; 9:313. [PMID: 26321914 PMCID: PMC4530619 DOI: 10.3389/fncel.2015.00313] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 07/28/2015] [Indexed: 12/13/2022] Open
Abstract
Learning and memory require alteration in number and strength of existing synaptic connections. Extracellular proteolysis within the synapses has been shown to play a pivotal role in synaptic plasticity by determining synapse structure, function, and number. Although synaptic plasticity of excitatory synapses is generally acknowledged to play a crucial role in formation of memory traces, some components of neural plasticity are reflected by nonsynaptic changes. Since information in neural networks is ultimately conveyed with action potentials, scaling of neuronal excitability could significantly enhance or dampen the outcome of dendritic integration, boost neuronal information storage capacity and ultimately learning. However, the underlying mechanism is poorly understood. With this regard, several lines of evidence and our most recent study support a view that activity of extracellular proteases might affect information processing in neuronal networks by affecting targets beyond synapses. Here, we review the most recent studies addressing the impact of extracellular proteolysis on plasticity of neuronal excitability and discuss how enzymatic activity may alter input-output/transfer function of neurons, supporting cognitive processes. Interestingly, extracellular proteolysis may alter intrinsic neuronal excitability and excitation/inhibition balance both rapidly (time of minutes to hours) and in long-term window. Moreover, it appears that by cleavage of extracellular matrix (ECM) constituents, proteases may modulate function of ion channels or alter inhibitory drive and hence facilitate active participation of dendrites and axon initial segments (AISs) in adjusting neuronal input/output function. Altogether, a picture emerges whereby both rapid and long-term extracellular proteolysis may influence some aspects of information processing in neurons, such as initiation of action potential, spike frequency adaptation, properties of action potential and dendritic backpropagation.
Collapse
Affiliation(s)
- Tomasz Wójtowicz
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University Wroclaw, Poland
| | - Patrycja Brzdąk
- Department of Animal Physiology, Institute of Experimental Biology, Wroclaw University Wroclaw, Poland
| | - Jerzy W Mozrzymas
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University Wroclaw, Poland ; Department of Animal Physiology, Institute of Experimental Biology, Wroclaw University Wroclaw, Poland
| |
Collapse
|
36
|
Pei J, Park IH, Ryu HH, Li SY, Li CH, Lim SH, Wen M, Jang WY, Jung S. Sublethal dose of irradiation enhances invasion of malignant glioma cells through p53-MMP 2 pathway in U87MG mouse brain tumor model. Radiat Oncol 2015; 10:164. [PMID: 26245666 PMCID: PMC4554349 DOI: 10.1186/s13014-015-0475-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 07/26/2015] [Indexed: 02/27/2023] Open
Abstract
Background Glioblastoma is a highly lethal neoplasm that frequently recurs locally after radiotherapy, and most of these recurrences originate from near the irradiated target field. In the present study, we identified the effects of radiation on glioma invasion and p53, TIMP-2, and MMP-2 expression through in vitro and in vivo experiments. Methods The U87MG (wt p53) and U251 (mt p53) human malignant glioma cell lines were prepared, and the U2OS (wt 53) and Saos2 (del p53) osteosarcoma cell lines were used as p53 positive and negative controls. The four cell lines and p53 knock-downed U87MG cells received radiation (2–6 Gy) and were analyzed for expression of p53 and TIMP-2 by Western blot, and MMP-2 activity was detected by zymography. In addition, the effects of irradiation on directional invasion of malignant glioma were evaluated by implanting nude mice with bioluminescent u87-Fluc in vivo followed by MMP-2, p53, and TIMP-2 immunohisto-chemistry and in situ zymography. Results MMP-2 activity and p53 expression increased in proportional to the radiation dose in cell lines with wt p53, but not in the cell lines with del or mt p53. TIMP-2 expression did not increase in U87MG cells. MMP-2 activity decreased in p53 knock-downed U87MG cells but increased in the control group. Furthermore, radiation enhanced MMP-2 activity and increased tumor margin invasiveness in vivo. Tumor cells invaded by radiation overexpressed MMP-2 and p53 and revealed high gelatinolytic activity compared with those of non-radiated tumor cells. Conclusion Radiation-induced upregulation of p53 modulated MMP-2 activity, and the imbalance between MMP-2 and TIMP-2 may have an important role in glioblastoma invasion by degrading the extracellular matrix. Bioluminescent “U87-Fluc”was useful for observing tumor formation without sacrifice after implanting tumor cells in the mouse brain. These findings suggest that the radiotherapy involved field for malignant glioma needs to be reconsidered, and that future trials should investigate concurrent pharmacologic therapies that inhibit invasion associated with radiotherapy. Electronic supplementary material The online version of this article (doi:10.1186/s13014-015-0475-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jian Pei
- Department of Neurosurgery, Chonnam National University Hwasun Hospital and Mediacal School, 322 Seoyang-ro, Hwasun-eup, Hwasun-gun, Jeonnam, 519-763, Republic of Korea
| | - In-Ho Park
- Department of Neurosurgery, Chonnam National University Hwasun Hospital and Mediacal School, 322 Seoyang-ro, Hwasun-eup, Hwasun-gun, Jeonnam, 519-763, Republic of Korea
| | - Hyang-Hwa Ryu
- Brain Tumor Research Laboratory, and Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, 322 Seoyang-ro, Hwasun-eup, Hwasun-gun, Jeonnam, 519-763, Republic of Korea
| | - Song-Yuan Li
- Brain Tumor Research Laboratory, and Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, 322 Seoyang-ro, Hwasun-eup, Hwasun-gun, Jeonnam, 519-763, Republic of Korea
| | - Chun-Hao Li
- Brain Tumor Research Laboratory, and Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, 322 Seoyang-ro, Hwasun-eup, Hwasun-gun, Jeonnam, 519-763, Republic of Korea
| | - Sa-Hoe Lim
- Department of Neurosurgery, Chonnam National University Hwasun Hospital and Mediacal School, 322 Seoyang-ro, Hwasun-eup, Hwasun-gun, Jeonnam, 519-763, Republic of Korea.,Brain Tumor Research Laboratory, and Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, 322 Seoyang-ro, Hwasun-eup, Hwasun-gun, Jeonnam, 519-763, Republic of Korea
| | - Min Wen
- Brain Tumor Research Laboratory, and Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, 322 Seoyang-ro, Hwasun-eup, Hwasun-gun, Jeonnam, 519-763, Republic of Korea
| | - Woo-Youl Jang
- Department of Neurosurgery, Chonnam National University Hwasun Hospital and Mediacal School, 322 Seoyang-ro, Hwasun-eup, Hwasun-gun, Jeonnam, 519-763, Republic of Korea
| | - Shin Jung
- Department of Neurosurgery, Chonnam National University Hwasun Hospital and Mediacal School, 322 Seoyang-ro, Hwasun-eup, Hwasun-gun, Jeonnam, 519-763, Republic of Korea. .,Brain Tumor Research Laboratory, and Chonnam National University Research Institute of Medical Sciences, Chonnam National University Hwasun Hospital and Medical School, 322 Seoyang-ro, Hwasun-eup, Hwasun-gun, Jeonnam, 519-763, Republic of Korea.
| |
Collapse
|
37
|
Ruiz-Perera L, Muniz M, Vierci G, Bornia N, Baroncelli L, Sale A, Rossi FM. Fluoxetine increases plasticity and modulates the proteomic profile in the adult mouse visual cortex. Sci Rep 2015. [PMID: 26205348 PMCID: PMC4513348 DOI: 10.1038/srep12517] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The scarce functional recovery of the adult CNS following injuries or diseases is largely due to its reduced potential for plasticity, the ability to reorganize neural connections as a function of experience. Recently, some new strategies restoring high levels of plasticity in the adult brain have been identified, especially in the paradigmatic model of the visual system. A chronic treatment with the anti-depressant fluoxetine reinstates plasticity in the adult rat primary visual cortex, inducing recovery of vision in amblyopic animals. The molecular mechanisms underlying this effect remain largely unknown. Here, we explored fluoxetine effects on mouse visual cortical plasticity, and exploited a proteomic approach to identify possible candidates mediating the outcome of the antidepressant treatment on adult cortical plasticity. We showed that fluoxetine restores ocular dominance plasticity in the adult mouse visual cortex, and identified 31 differentially expressed protein spots in fluoxetine-treated animals vs. controls. MALDITOF/TOF mass spectrometry identification followed by bioinformatics analysis revealed that these proteins are involved in the control of cytoskeleton organization, endocytosis, molecular transport, intracellular signaling, redox cellular state, metabolism and protein degradation. Altogether, these results indicate a complex effect of fluoxetine on neuronal signaling mechanisms potentially involved in restoring plasticity in the adult brain.
Collapse
Affiliation(s)
- L Ruiz-Perera
- Laboratorio de Neurociencias "Neuroplasticity Unit", Facultad de Ciencias, UdelaR, Montevideo, Uruguay
| | - M Muniz
- Laboratorio de Neurociencias "Neuroplasticity Unit", Facultad de Ciencias, UdelaR, Montevideo, Uruguay
| | - G Vierci
- Laboratorio de Neurociencias "Neuroplasticity Unit", Facultad de Ciencias, UdelaR, Montevideo, Uruguay
| | - N Bornia
- Laboratorio de Neurociencias "Neuroplasticity Unit", Facultad de Ciencias, UdelaR, Montevideo, Uruguay
| | - L Baroncelli
- Institute of Neuroscience, Consiglio Nazionale delle Ricerche, Pisa, Italy
| | - A Sale
- Institute of Neuroscience, Consiglio Nazionale delle Ricerche, Pisa, Italy
| | - F M Rossi
- Laboratorio de Neurociencias "Neuroplasticity Unit", Facultad de Ciencias, UdelaR, Montevideo, Uruguay
| |
Collapse
|
38
|
Tojima T, Kamiguchi H. Exocytic and endocytic membrane trafficking in axon development. Dev Growth Differ 2015; 57:291-304. [DOI: 10.1111/dgd.12218] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 04/09/2015] [Accepted: 04/09/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Takuro Tojima
- Laboratory for Neuronal Growth Mechanisms; RIKEN Brain Science Institute; 2-1 Hirosawa Wako Saitama 351-0198 Japan
| | - Hiroyuki Kamiguchi
- Laboratory for Neuronal Growth Mechanisms; RIKEN Brain Science Institute; 2-1 Hirosawa Wako Saitama 351-0198 Japan
| |
Collapse
|
39
|
Liao G, Wang Z, Lee E, Moreno S, Abuelnasr O, Baudry M, Bi X. Enhanced expression of matrix metalloproteinase-12 contributes to Npc1 deficiency-induced axonal degeneration. Exp Neurol 2015; 269:67-74. [PMID: 25864931 DOI: 10.1016/j.expneurol.2015.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 01/08/2015] [Accepted: 04/02/2015] [Indexed: 11/28/2022]
Abstract
Niemann-Pick type C (NPC) disease is a genetic disorder associated with intracellular cholesterol accumulation in the brain and other organs, and neurodegeneration is generally believed to be the fatal cause of the disease. In view of the emerging role of matrix metalloproteinase-12 (MMP-12) in neuronal injury, we investigated its expression and potential roles in axonal degeneration in Npc1-/- mouse brain. Microarray and quantitative real-time reversed transcription PCR analysis indicated a marked increase in MMP-12 mRNA levels in cerebellum of 3 week-old Npc1-/- mice, as compared to wild-type littermates. Western blots showed that the ratio of mature MMP-12 over pro-MMP-12 was significantly increased in cerebellum of Npc1-/-, as compared to wild-type mice. Immunohistochemical studies confirmed that MMP-12 expression was increased, especially in the cell bodies of Purkinje neurons in Npc1-/- mice. Neuritic growth was significantly reduced by Npc1 siRNA knockdown in nerve growth factor-differentiated PC-12 cells, and this effect was completely reversed by treatment with an MMP-12 specific inhibitor. Furthermore, in vivo experiments showed that chronic treatment with the MMP-12 inhibitor ameliorated Npc1 deficiency-induced axonal pathology in the striatum. Our results indicate that abnormal neuronal expression of MMP-12 may contribute to axonal degeneration in NPC disease, thus providing a potential novel target for treatment.
Collapse
Affiliation(s)
- Guanghong Liao
- Department of Basic Medical Sciences, COMP, Western University of Health Sciences, 701 E. Second Street, Pomona, CA 91766, USA
| | - Zhuangjun Wang
- Department of Basic Medical Sciences, COMP, Western University of Health Sciences, 701 E. Second Street, Pomona, CA 91766, USA
| | - Erik Lee
- Department of Basic Medical Sciences, COMP, Western University of Health Sciences, 701 E. Second Street, Pomona, CA 91766, USA
| | - Stephanie Moreno
- Department of Basic Medical Sciences, COMP, Western University of Health Sciences, 701 E. Second Street, Pomona, CA 91766, USA
| | - Omar Abuelnasr
- Department of Basic Medical Sciences, COMP, Western University of Health Sciences, 701 E. Second Street, Pomona, CA 91766, USA
| | - Michel Baudry
- Graduate College of Biomedical Sciences, Western University of Health Sciences, 701 E. Second Street, Pomona, CA 91766, USA
| | - Xiaoning Bi
- Department of Basic Medical Sciences, COMP, Western University of Health Sciences, 701 E. Second Street, Pomona, CA 91766, USA.
| |
Collapse
|
40
|
Lee S, Hennigar SR, Alam S, Nishida K, Kelleher SL. Essential Role for Zinc Transporter 2 (ZnT2)-mediated Zinc Transport in Mammary Gland Development and Function during Lactation. J Biol Chem 2015; 290:13064-78. [PMID: 25851903 DOI: 10.1074/jbc.m115.637439] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Indexed: 01/28/2023] Open
Abstract
The zinc transporter ZnT2 (SLC30A2) imports zinc into vesicles in secreting mammary epithelial cells (MECs) and is critical for zinc efflux into milk during lactation. Recent studies show that ZnT2 also imports zinc into mitochondria and is expressed in the non-lactating mammary gland and non-secreting MECs, highlighting the importance of ZnT2 in general mammary gland biology. In this study we used nulliparous and lactating ZnT2-null mice and characterized the consequences on mammary gland development, function during lactation, and milk composition. We found that ZnT2 was primarily expressed in MECs and to a limited extent in macrophages in the nulliparous mammary gland and loss of ZnT2 impaired mammary expansion during development. Secondly, we found that lactating ZnT2-null mice had substantial defects in mammary gland architecture and MEC function during secretion, including fewer, condensed and disorganized alveoli, impaired Stat5 activation, and unpolarized MECs. Loss of ZnT2 led to reduced milk volume and milk containing less protein, fat, and lactose compared with wild-type littermates, implicating ZnT2 in the regulation of mammary differentiation and optimal milk production during lactation. Together, these results demonstrate that ZnT2-mediated zinc transport is critical for mammary gland function, suggesting that defects in ZnT2 not only reduce milk zinc concentration but may compromise breast health and increase the risk for lactation insufficiency in lactating women.
Collapse
Affiliation(s)
- Sooyeon Lee
- From the Interdisciplinary Graduate Physiology Program and Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, Departments of Cellular and Molecular Physiology
| | - Stephen R Hennigar
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Samina Alam
- Departments of Cellular and Molecular Physiology, Surgery, Penn State Hershey College of Medicine, Hershey, Pennsylvania 17033
| | - Keigo Nishida
- Laboratory for Homeostatic Network, RCAI, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Yokohama 230-0045, Japan, and Laboratory of Immune Regulation, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka 513-8670, Japan
| | - Shannon L Kelleher
- From the Interdisciplinary Graduate Physiology Program and Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, Departments of Cellular and Molecular Physiology, Surgery, Penn State Hershey College of Medicine, Hershey, Pennsylvania 17033, Pharmacology, and
| |
Collapse
|
41
|
Yu S, Yehia G, Wang J, Stypulkowski E, Sakamori R, Jiang P, Hernandez-Enriquez B, Tran TS, Bonder EM, Guo W, Gao N. Global ablation of the mouse Rab11a gene impairs early embryogenesis and matrix metalloproteinase secretion. J Biol Chem 2014; 289:32030-32043. [PMID: 25271168 DOI: 10.1074/jbc.m113.538223] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Rab11a has been conceived as a prominent regulatory component of the recycling endosome, which acts as a nexus in the endo- and exocytotic networks. The precise in vivo role of Rab11a in mouse embryonic development is unknown. We globally ablated Rab11a and examined the phenotypic and molecular outcomes in Rab11a(null) blastocysts and mouse embryonic fibroblasts. Using multiple trafficking assays and complementation analyses, we determined, among multiple important membrane-associated and soluble cargos, the critical contribution of Rab11a vesicular traffic to the secretion of multiple soluble MMPs. Rab11a(null) embryos were able to properly form normal blastocysts but died at peri-implantation stages. Our data suggest that Rab11a critically controls mouse blastocyst development and soluble matrix metalloproteinase secretion.
Collapse
Affiliation(s)
- Shiyan Yu
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Ghassan Yehia
- Transgenic Core Facility, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Juanfei Wang
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, and
| | - Ewa Stypulkowski
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Ryotaro Sakamori
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Ping Jiang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724
| | | | - Tracy S Tran
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Wei Guo
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, and
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102,.
| |
Collapse
|
42
|
Py NA, Bonnet AE, Bernard A, Marchalant Y, Charrat E, Checler F, Khrestchatisky M, Baranger K, Rivera S. Differential spatio-temporal regulation of MMPs in the 5xFAD mouse model of Alzheimer's disease: evidence for a pro-amyloidogenic role of MT1-MMP. Front Aging Neurosci 2014; 6:247. [PMID: 25278878 PMCID: PMC4166961 DOI: 10.3389/fnagi.2014.00247] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 09/02/2014] [Indexed: 12/03/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are pleiotropic endopeptidases involved in a variety of neurodegenerative/neuroinflammatory processes through their interactions with a large number of substrates. Among those, the amyloid precursor protein (APP) and the beta amyloid peptide (Aβ) are largely associated with the development of Alzheimer’s disease (AD). However, the regulation and potential contribution of MMPs to AD remains unclear. In this study, we investigated the evolution of the expression of MMP-2, MMP-9, and membrane-type 1-MMP (MT1-MMP) in the hippocampus at different stages of the pathology (asymptomatic, prodromal-like and symptomatic) in the 5xFAD transgenic mouse AD model. In parallel we also followed the expression of functionally associated factors. Overall, the expression of MMP-2, MMP-9, and MT1-MMP was upregulated concomitantly with the tissue inhibitor of MMPs-1 (TIMP-1) and several markers of inflammatory/glial response. The three MMPs exhibited age- and cell-dependent upregulation of their expression, with MMP-2 and MMP-9 being primarily located to astrocytes, and MT1-MMP to neurons. MMP-9 and MT1-MMP were also prominently present in amyloid plaques. The levels of active MT1-MMP were highly upregulated in membrane-enriched fractions of hippocampus at 6 months of age (symptomatic phase), when the levels of APP, its metabolites APP C-terminal fragments (CTFs), and Aβ trimers were the highest. Overexpression of MT1-MMP in HEK cells carrying the human APP Swedish mutation (HEKswe) strongly increased β-secretase derived C-terminal APP fragment (C99) and Aβ levels, whereas MMP-2 overexpression nearly abolished Aβ production without affecting C99. Our data consolidate the emerging idea of a regulatory interplay between MMPs and the APP/Aβ system, and demonstrate for the first time the pro-amyloidogenic features of MT1-MMP. Further investigation will be justified to evaluate this MMP as a novel potential therapeutic target in AD.
Collapse
Affiliation(s)
- Nathalie A Py
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France
| | - Amandine E Bonnet
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France
| | - Anne Bernard
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France
| | - Yannick Marchalant
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France
| | - Eliane Charrat
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France
| | | | - Michel Khrestchatisky
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France
| | - Kévin Baranger
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France ; Department of Neurology and Neuropsychology, APHM, CHU La Timone Marseille, France
| | - Santiago Rivera
- Faculty of Medicine, Aix-Marseille Université, CNRS, NICN, UMR7259 Marseille, France
| |
Collapse
|
43
|
Stawarski M, Stefaniuk M, Wlodarczyk J. Matrix metalloproteinase-9 involvement in the structural plasticity of dendritic spines. Front Neuroanat 2014; 8:68. [PMID: 25071472 PMCID: PMC4091410 DOI: 10.3389/fnana.2014.00068] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 06/25/2014] [Indexed: 01/01/2023] Open
Abstract
Dendritic spines are the locus for excitatory synaptic transmission in the brain and thus play a major role in neuronal plasticity. The ability to alter synaptic connections includes volumetric changes in dendritic spines that are driven by scaffolds created by the extracellular matrix (ECM). Here, we review the effects of the proteolytic activity of ECM proteases in physiological and pathological structural plasticity. We use matrix metalloproteinase-9 (MMP-9) as an example of an ECM modifier that has recently emerged as a key molecule in regulating the morphology and dysmorphology of dendritic spines that underlie synaptic plasticity and neurological disorders, respectively. We summarize the influence of MMP-9 on the dynamic remodeling of the ECM via the cleavage of extracellular substrates. We discuss its role in the formation, modification, and maintenance of dendritic spines in learning and memory. Finally, we review research that implicates MMP-9 in aberrant synaptic plasticity and spine dysmorphology in neurological disorders, with a focus on morphological abnormalities of dendritic protrusions that are associated with epilepsy.
Collapse
Affiliation(s)
- Michal Stawarski
- Laboratory of Cell Biophysics, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology Warsaw, Mazowieckie, Poland
| | - Marzena Stefaniuk
- Laboratory of Neurobiology, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology Warsaw, Mzowieckie, Poland
| | - Jakub Wlodarczyk
- Laboratory of Cell Biophysics, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology Warsaw, Mazowieckie, Poland
| |
Collapse
|
44
|
Matrix metalloproteinases and their multiple roles in Alzheimer's disease. BIOMED RESEARCH INTERNATIONAL 2014; 2014:908636. [PMID: 25050378 PMCID: PMC4094696 DOI: 10.1155/2014/908636] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 06/09/2014] [Accepted: 06/10/2014] [Indexed: 01/09/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent type of dementia. Pathological changes in the AD brain include amyloid-β (Aβ) plaques and neurofibrillary tangles (NFTs), as well as neuronal death and synaptic loss. Matrix metalloproteinases (MMPs) play an important role as inflammatory components in the pathogenesis of AD. MMP-2 might be assumed to have a protective role in AD and is the major MMP which is directly linked to Aβ in the brain. Synthesis of MMP-9 can be induced by Aβ, and the enzymes appear to exert multiple effects in AD in senile plaque homoeostasis. The proaggregatory influence on tau oligomer formation in strategic brain regions may be a potential neurotoxic side effect of MMP-9. MMP-3 levels are correlated to the duration of AD and correlate with the CSF T-tau and P-tau levels in the elderly controls. Elevated brain levels of MMP-3 might result in increased MMP-9 activity and indirectly facilitate tau aggregation. At present, the clinical utility of these proteins, particularly in plasma or serum, as potential early diagnostic biomarkers for AD remains to be established. More research is needed to understand the diverse roles of these proteases to design specific drugs and devise therapeutic strategies for AD.
Collapse
|
45
|
Baranger K, Rivera S, Liechti FD, Grandgirard D, Bigas J, Seco J, Tarrago T, Leib SL, Khrestchatisky M. Endogenous and synthetic MMP inhibitors in CNS physiopathology. PROGRESS IN BRAIN RESEARCH 2014; 214:313-51. [DOI: 10.1016/b978-0-444-63486-3.00014-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
46
|
Tsilibary E, Tzinia A, Radenovic L, Stamenkovic V, Lebitko T, Mucha M, Pawlak R, Frischknecht R, Kaczmarek L. Neural ECM proteases in learning and synaptic plasticity. PROGRESS IN BRAIN RESEARCH 2014; 214:135-57. [PMID: 25410356 DOI: 10.1016/b978-0-444-63486-3.00006-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recent studies implicate extracellular proteases in synaptic plasticity, learning, and memory. The data are especially strong for such serine proteases as thrombin, tissue plasminogen activator, neurotrypsin, and neuropsin as well as matrix metalloproteinases, MMP-9 in particular. The role of those enzymes in the aforementioned phenomena is supported by the experimental results on the expression patterns (at the gene expression and protein and enzymatic activity levels) and functional studies, including knockout mice, specific inhibitors, etc. Counterintuitively, the studies have shown that the extracellular proteolysis is not responsible mainly for an overall degradation of the extracellular matrix (ECM) and loosening perisynaptic structures, but rather allows for releasing signaling molecules from the ECM, transsynaptic proteins, and latent form of growth factors. Notably, there are also indications implying those enzymes in the major neuropsychiatric disorders, probably by contributing to synaptic aberrations underlying such diseases as schizophrenia, bipolar, autism spectrum disorders, and drug addiction.
Collapse
Affiliation(s)
- Effie Tsilibary
- Institute of Biosciences and Applications, NCSR "Demokritos", Athens, Greece
| | - Athina Tzinia
- Institute of Biosciences and Applications, NCSR "Demokritos", Athens, Greece
| | - Lidija Radenovic
- Center for Laser Microscopy, Institute for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Vera Stamenkovic
- Center for Laser Microscopy, Institute for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Tomasz Lebitko
- Department of Molecular and Cellular Neurobiology, Nencki Institute, Warsaw, Poland
| | | | | | - Renato Frischknecht
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Leszek Kaczmarek
- Department of Molecular and Cellular Neurobiology, Nencki Institute, Warsaw, Poland.
| |
Collapse
|
47
|
Ogden A, Rida PCG, Aneja R. Heading off with the herd: how cancer cells might maneuver supernumerary centrosomes for directional migration. Cancer Metastasis Rev 2013; 32:269-87. [PMID: 23114845 DOI: 10.1007/s10555-012-9413-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The complicity of centrosomes in carcinogenesis is unmistakable. Mounting evidence clearly implicates a robust correlation between centrosome amplification (CA) and malignant transformation in diverse tissue types. Furthermore, CA has been suggested as a marker of cancer aggressiveness, in particular the invasive phenotype, in breast and prostate cancers. One means by which CA promotes malignancy is through induction of transient spindle multipolarity during mitosis, which predisposes the cell to karyotypic changes arising from low-grade chromosome mis-segregation. It is well recognized that during cell migration in interphase, centrosome-mediated nucleation of a radial microtubule array is crucial for establishing a polarized Golgi apparatus, without which directionality is precluded. The question of how cancer cells maneuver their supernumerary centrosomes to achieve directionality during cell migration is virtually uncharted territory. Given that CA is a hallmark of cancers and has been correlated with cancer aggressiveness, malignant cells are presumably competent in managing their centrosome surfeit during directional migration, although the cellular logistics of this process remain unexplored. Another key angle worth pondering is whether an overabundance of centrosomes confers some advantage on cancer cells in terms of their migratory and invasive capabilities. Recent studies have uncovered a remarkable strategy that cancer cells employ to deal with the problem of excess centrosomes and ensure bipolar mitoses, viz., centrosome clustering. This review aims to change the narrative by exploring how an increased centrosome complement may, via aneuploidy-independent modulation of the microtubule cytoskeleton, enhance directional migration and invasion of malignant cells. We postulate that CA imbues cancer cells with cytoskeletal advantages that enhance cell polarization, Golgi-dependent vesicular trafficking, stromal invasion, and other aspects of metastatic progression. We also propose that centrosome declustering may represent a novel, cancer cell-specific antimetastatic strategy, as cancer cells may rely on centrosome clustering during migration as they do in mitosis. Elucidation of these details offers an exciting avenue for future research, as does investigating how CA may promote metastasis through enhanced directional migration.
Collapse
Affiliation(s)
- Angela Ogden
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | | | | |
Collapse
|
48
|
Lonskaya I, Partridge J, Lalchandani RR, Chung A, Lee T, Vicini S, Hoe HS, Lim ST, Conant K. Soluble ICAM-5, a product of activity dependent proteolysis, increases mEPSC frequency and dendritic expression of GluA1. PLoS One 2013; 8:e69136. [PMID: 23844251 PMCID: PMC3699500 DOI: 10.1371/journal.pone.0069136] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 06/12/2013] [Indexed: 11/23/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are zinc dependent endopeptidases that can be released from neurons in an activity dependent manner to play a role in varied forms of learning and memory. MMP inhibitors impair hippocampal long term potentiation (LTP), spatial memory, and behavioral correlates of drug addiction. Since MMPs are thought to influence LTP through a β1 integrin dependent mechanism, it has been suggested that these enzymes cleave specific substrates to generate integrin binding ligands. In previously published work, we have shown that neuronal activity stimulates rapid MMP dependent shedding of intercellular adhesion molecule-5 (ICAM-5), a synaptic adhesion molecule expressed on dendrites of the telencephalon. We have also shown that the ICAM-5 ectodomain can interact with β1 integrins to stimulate integrin dependent phosphorylation of cofilin, an event that occurs with dendritic spine maturation and LTP. In the current study, we investigate the potential for the ICAM-5 ectodomain to stimulate changes in α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptor (AMPAR) dependent glutamatergic transmission. Single cell recordings show that the ICAM-5 ectodomain stimulates an increase in the frequency, but not the amplitude, of AMPA mini excitatory post synaptic currents (mEPSCs). With biotinylation and precipitation assays, we also show that the ICAM-5 ectodomain stimulates an increase in membrane levels of GluA1, but not GluA2, AMPAR subunits. In addition, we observe an ICAM-5 associated increase in GluA1 phosphorylation at serine 845. Concomitantly, ICAM-5 affects an increase in GluA1 surface staining along dendrites without affecting an increase in dendritic spine number. Together these data are consistent with the possibility that soluble ICAM-5 increases glutamatergic transmission and that post-synaptic changes, including increased phosphorylation and dendritic insertion of GluA1, could contribute. We suggest that future studies are warranted to determine whether ICAM-5 is one of a select group of synaptic CAMs whose shedding contributes to MMP dependent effects on learning and memory.
Collapse
Affiliation(s)
- Irina Lonskaya
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C., United States of America
| | - John Partridge
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D.C., United States of America
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Rupa R. Lalchandani
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Andrew Chung
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Taehee Lee
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Stefano Vicini
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D.C., United States of America
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Hyang-Sook Hoe
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C., United States of America
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Seung T. Lim
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C., United States of America
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Katherine Conant
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C., United States of America
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C., United States of America
- * E-mail:
| |
Collapse
|
49
|
Ould-Yahoui A, Sbai O, Baranger K, Bernard A, Gueye Y, Charrat E, Clément B, Gigmes D, Dive V, Girard SD, Féron F, Khrestchatisky M, Rivera S. Role of Matrix Metalloproteinases in Migration and Neurotrophic Properties of Nasal Olfactory Stem and Ensheathing Cells. Cell Transplant 2013; 22:993-1010. [DOI: 10.3727/096368912x657468] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Adult olfactory ectomesenchymal stem cells (OE-MSCs) and olfactory ensheathing cells (OECs), both from the nasal olfactory lamina propria, display robust regenerative properties when transplanted into the nervous system, but the mechanisms supporting such therapeutic effects remain unknown. Matrix metalloproteinases (MMPs) are an important family of proteinases contributing to cell motility and axonal outgrowth across the extracellular matrix (ECM) in physiological and pathological conditions. In this study, we have characterized for the first time in nasal human OE-MSCs the expression profile of some MMPs currently associated with cell migration and invasiveness. We demonstrate different patterns of expression for MMP-1, MMP-2, MMP-9, and MT1-MMP upon cell migration when compared with nonmigrating cells. Our results establish a correspondence between the localization of these proteinases in the migration front with the ability of cells to migrate. Using various modulators of MMP activity, we also show that at least MMP-2, MMP-9, and MT1-MMP contribute to OE-MSC migration in an in vitro 3D test. Furthermore, we demonstrate under the same conditions of culture used for in vivo transplantation that OE-MSCs and OECs secrete neurotrophic factors that promote neurite outgrowth of cortical and dorsal root ganglia (DRG) neurons, as well as axo-dendritic differentiation of cortical neurons. These effects were abolished by the depletion of MMP-2 and MMP-9 from the culture conditioned media. Altogether, our results provide the first evidence that MMPs may contribute to the therapeutic features of OE-MSCs and OECs through the control of their motility and/or their neurotrophic properties. Our data provide new insight into the mechanisms of neuroregeneration and will contribute to optimization of cell therapy strategies.
Collapse
Affiliation(s)
- Adlane Ould-Yahoui
- Aix-Marseille Univ, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
- CNRS, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
| | - Oualid Sbai
- Aix-Marseille Univ, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
- CNRS, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
| | - Kévin Baranger
- Aix-Marseille Univ, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
- CNRS, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
| | - Anne Bernard
- Aix-Marseille Univ, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
- CNRS, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
| | - Yatma Gueye
- Aix-Marseille Univ, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
- CNRS, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
| | - Eliane Charrat
- Aix-Marseille Univ, Institut de Chimie Radicalaire, Equipe Chimie Radicalaire, Organique et Polymères de Spécialité, UMR 7273, Marseille, France
- CNRS, Institut de Chimie Radicalaire, Equipe Chimie Radicalaire, Organique et Polymères de Spécialité, UMR 7273, Marseille, France
| | - Benoît Clément
- Aix-Marseille Univ, Institut de Chimie Radicalaire, Equipe Chimie Radicalaire, Organique et Polymères de Spécialité, UMR 7273, Marseille, France
- CNRS, Institut de Chimie Radicalaire, Equipe Chimie Radicalaire, Organique et Polymères de Spécialité, UMR 7273, Marseille, France
| | - Didier Gigmes
- Aix-Marseille Univ, Institut de Chimie Radicalaire, Equipe Chimie Radicalaire, Organique et Polymères de Spécialité, UMR 7273, Marseille, France
- CNRS, Institut de Chimie Radicalaire, Equipe Chimie Radicalaire, Organique et Polymères de Spécialité, UMR 7273, Marseille, France
| | - Vincent Dive
- Département d'Ingénierie et d'Etudes des Protéines (DIEP), CEA/Saclay, Gif-sur-Yvette, France
| | - Stéphane D. Girard
- Aix-Marseille Univ, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
- CNRS, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
| | - François Féron
- Aix-Marseille Univ, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
- CNRS, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
| | - Michel Khrestchatisky
- Aix-Marseille Univ, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
- CNRS, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
| | - Santiago Rivera
- Aix-Marseille Univ, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
- CNRS, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, 13344, Marseille, France
| |
Collapse
|
50
|
Furmanova-Hollenstein P, Broggini-Tenzer A, Eggel M, Millard AL, Pruschy M. The microtubule stabilizer patupilone counteracts ionizing radiation-induced matrix metalloproteinase activity and tumor cell invasion. Radiat Oncol 2013; 8:105. [PMID: 23631818 PMCID: PMC3661365 DOI: 10.1186/1748-717x-8-105] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 04/23/2013] [Indexed: 02/06/2023] Open
Abstract
Background Ionizing radiation (IR) in combination with microtubule stabilizing agents (MSA) is a promising combined treatment modality. Supra-additive treatment responses might result from direct tumor cell killing and cooperative indirect, tumor cell-mediated effects on the tumor microenvironment. Here we investigated deregulation of matrix metalloproteinase (MMP) activity, as an important component of the tumor microenvironment, by the combined treatment modality of IR with the clinically relevant MSA patupilone. Methods Expression, secretion and activity of MMPs and related tissue inhibitors of metalloproteinases (TIMPs) were determined in cell extracts and conditioned media derived from human fibrosarcoma HT1080 and human glioblastoma U251 tumor cells in response to treatment with IR and the MSA patupilone. Treatment-dependent changes of the invasive capacities of these tumor cell lines were analysed using a Transwell invasion assay. Control experiments were performed using TIMP-directed siRNA and TIMP-directed inhibitory antibodies. Results Enzymatic activity of secreted MMPs was determined after treatment with patupilone and irradiation in the human fibrosarcoma HT1080 and the human glioblastoma U251 tumor cell line. IR enhanced the activity of secreted MMPs up to 2-fold and cellular pretreatment with low dose patupilone (0.05-0.2 nM) counteracted specifically the IR-induced MMP activity. The cell invasive capacity of HT1080 and U251 cells was increased after irradiation with 2 Gy by 30% and 50%, respectively, and patupilone treatment completely abrogated IR-induced cell invasion. Patupilone did not alter the level of MMP expression, but interestingly, the protein level of secreted TIMP-1 and TIMP-2 was lower after combined treatment than after irradiation treatment alone. Furthermore, siRNA depletion of TIMP-1 or TIMP-2 prevented IR-mediated induction of MMP activity and cell invasion. Conclusions These results indicate that patupilone counteracts an IR-induced MMP activation process by the reduction of secreted TIMP-1 and TIMP-2 proteins, which are required for activation of MMPs. Since IR-induced MMP activity could contribute to tumor progression, treatment combination of IR with patupilone might be of great clinical benefit for tumor therapy.
Collapse
|