1
|
Brenna S, Glatzel M, Magnus T, Puig B, Galliciotti G. Neuroserpin and Extracellular Vesicles in Ischemic Stroke: Partners in Neuroprotection? Aging Dis 2024; 15:2191-2204. [PMID: 39191396 PMCID: PMC11346402 DOI: 10.14336/ad.2024.0518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/05/2024] [Indexed: 08/29/2024] Open
Abstract
Ischemic stroke represents a significant global health challenge, often resulting in death or long-term disability, particularly among the elderly, where advancing age stands as the most unmodifiable risk factor. Arising from the blockage of a brain-feeding artery, the only therapies available to date aim at removing the blood clot to restore cerebral blood flow and rescue neuronal cells from death. The prevailing treatment approach involves thrombolysis by administration of recombinant tissue plasminogen activator (tPA), albeit with a critical time constraint. Timely intervention is imperative, given that delayed thrombolysis increases tPA leakage into the brain parenchyma, causing harmful effects. Strategies to preserve tPA's vascular benefits while shielding brain cells from its toxicity have been explored. Notably, administering neuroserpin (Ns), a brain-specific tPA inhibitor, represents one such approach. Following ischemic stroke, Ns levels rise and correlate with favorable post-stroke outcomes. Studies in rodent models of focal cerebral ischemia have demonstrated the beneficial effects of Ns administration. Ns treatment maintains blood-brain barrier (BBB) integrity, reducing stroke volume. Conversely, Ns-deficient animals exhibit larger stroke injury, increased BBB permeability and enhanced microglia activation. Furthermore, Ns administration extends the therapeutic window for tPA intervention, underscoring its potential in stroke management. Remarkably, our investigation reveals the presence of Ns within extracellular vesicles (EVs), small membrane-surrounded particles released by all cells and critical for intercellular communication. EVs influence disease outcome following stroke through cargo transfer between cells. Clarifying the role of EVs containing NS could open up urgently needed novel therapeutic approaches to improve post-ischemic stroke outcome.
Collapse
Affiliation(s)
- Santra Brenna
- Experimental Research in Stroke and Inflammation (ERSI) Group, Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tim Magnus
- Experimental Research in Stroke and Inflammation (ERSI) Group, Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Berta Puig
- Experimental Research in Stroke and Inflammation (ERSI) Group, Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Giovanna Galliciotti
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
2
|
Strand 1A variant in neuroserpin shows increased aggregation and no loss of inhibition: implication in ameliorating polymerization to retain activity. Biosci Rep 2022; 42:232125. [PMID: 36408789 PMCID: PMC9760604 DOI: 10.1042/bsr20221825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/31/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
Neuroserpin (NS) is predominantly expressed in the brain and is the primary inhibitor of tissue plasminogen activator (tPA). NS variants are associated with the neurogenerative disease termed familial encephalopathy with neuroserpin inclusion bodies (FENIB). The disease is characterized by variable age of onset and severity. The reactive center loop (RCL) insertion-based inhibitory mechanism of NS requires a coordinated conformational change leading to a shift in the strands of the β-sheet A and movement of helix F. Strand 1A is connected to the helix F at its C terminal end and with the strand 2A at its N terminal, both these domain move for accommodating the inserting loop; therefore, a variant that influences their movement may alter the inhibition rates. A molecular dynamic simulation analysis of a H138C NS variant from strand 1A showed a large decrease in conformational fluctuations as compared with wild-type NS. H138 was mutated, expressed, purified and a native-PAGE and transmission electron microscopy (TEM) analysis showed that this variant forms large molecular weight aggregates on a slight increase in temperature. However, a circular dichroism analysis showed its secondary structure to be largely conserved. Surprisingly, its tPA inhibition activity and complex formation remain unhindered even after the site-specific labeling of H138C with Alexa fluor C5 maleimide. Further, a helix F-strand 1A (W154C-H138C) double variant still shows appreciable inhibitory activity. Increasingly, it appears that aggregation and not loss of inhibition is the more likely cause of shutter region-based variants phenotypes, indicating that hindering polymer formation using small molecules may retain inhibitory activity in pathological variants of NS.
Collapse
|
3
|
Torrente D, Su EJ, Fredriksson L, Warnock M, Bushart D, Mann KM, Emal CD, Lawrence DA. Compartmentalized Actions of the Plasminogen Activator Inhibitors, PAI-1 and Nsp, in Ischemic Stroke. Transl Stroke Res 2022; 13:801-815. [PMID: 35122213 PMCID: PMC9349468 DOI: 10.1007/s12975-022-00992-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/22/2021] [Accepted: 01/26/2022] [Indexed: 02/08/2023]
Abstract
Tissue plasminogen activator (tPA) is a multifunctional protease. In blood tPA is best understood for its role in fibrinolysis, whereas in the brain tPA is reported to regulate blood-brain barrier (BBB) function and to promote neurodegeneration. Thrombolytic tPA is used for the treatment of ischemic stroke. However, its use is associated with an increased risk of hemorrhagic transformation. In blood the primary regulator of tPA activity is plasminogen activator inhibitor 1 (PAI-1), whereas in the brain, its primary inhibitor is thought to be neuroserpin (Nsp). In this study, we compare the effects of PAI-1 and Nsp deficiency in a mouse model of ischemic stroke and show that tPA has both beneficial and harmful effects that are differentially regulated by PAI-1 and Nsp. Following ischemic stroke Nsp deficiency in mice leads to larger strokes, increased BBB permeability, and increased spontaneous intracerebral hemorrhage. In contrast, PAI-1 deficiency results in smaller infarcts and increased cerebral blood flow recovery. Mechanistically, our data suggests that these differences are largely due to the compartmentalized action of PAI-1 and Nsp, with Nsp deficiency enhancing tPA activity in the CNS which increases BBB permeability and worsens stroke outcomes, while PAI-1 deficiency enhances fibrinolysis and improves recovery. Finally, we show that treatment with a combination therapy that enhances endogenous fibrinolysis by inhibiting PAI-1 with MDI-2268 and reduces BBB permeability by inhibiting tPA-mediated PDGFRα signaling with imatinib significantly reduces infarct size compared to vehicle-treated mice and to mice with either treatment alone.
Collapse
Affiliation(s)
- Daniel Torrente
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Enming Joseph Su
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-0644, USA
| | - Linda Fredriksson
- Biomedicum, Karolinska Institute, Solnavägen 9, Quarter 6D, 17165, Solna, Sweden
| | - Mark Warnock
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-0644, USA
| | - David Bushart
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-0644, USA
- Current affiliation: Ohio State University College of Medicine, Columbus, OH, USA
| | - Kris M Mann
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-0644, USA
| | - Cory D Emal
- Department of Chemistry, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Daniel A Lawrence
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-0644, USA.
| |
Collapse
|
4
|
West J, Satapathy S, Whiten DR, Kelly M, Geraghty NJ, Proctor EJ, Sormanni P, Vendruscolo M, Buxbaum JN, Ranson M, Wilson MR. Neuroserpin and transthyretin are extracellular chaperones that preferentially inhibit amyloid formation. SCIENCE ADVANCES 2021; 7:eabf7606. [PMID: 34890220 PMCID: PMC8664251 DOI: 10.1126/sciadv.abf7606] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 10/21/2021] [Indexed: 06/13/2023]
Abstract
Neuroserpin is a secreted protease inhibitor known to inhibit amyloid formation by the Alzheimer’s beta peptide (Aβ). To test whether this effect was constrained to Aβ, we used a range of in vitro assays to demonstrate that neuroserpin inhibits amyloid formation by several different proteins and protects against the associated cytotoxicity but, unlike other known chaperones, has a poor ability to inhibit amorphous protein aggregation. Collectively, these results suggest that neuroserpin has an unusual chaperone selectivity for intermediates on the amyloid-forming pathway. Bioinformatics analyses identified a highly conserved 14-residue region containing an α helix shared between neuroserpin and the thyroxine-transport protein transthyretin, and we subsequently demonstrated that transthyretin also preferentially inhibits amyloid formation. Last, we used rationally designed neuroserpin mutants to demonstrate a direct involvement of the conserved 14-mer region in its chaperone activity. Identification of this conserved region may prove useful in the future design of anti-amyloid reagents.
Collapse
Affiliation(s)
- Jennifer West
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia
| | - Sandeep Satapathy
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia
| | - Daniel R. Whiten
- Kolling Institute of Medical Research, University of Sydney, NSW 2065, Australia
| | - Megan Kelly
- School of Medicine, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia
| | - Nicholas J. Geraghty
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia
| | - Emma-Jayne Proctor
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia
| | - Pietro Sormanni
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Joel N. Buxbaum
- The Scripps Research Institute, La Jolla, CA, USA
- Protego Biopharma, La Jolla, CA, USA
| | - Marie Ranson
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia
| | - Mark R. Wilson
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW 2522, Australia
| |
Collapse
|
5
|
D'Acunto E, Fra A, Visentin C, Manno M, Ricagno S, Galliciotti G, Miranda E. Neuroserpin: structure, function, physiology and pathology. Cell Mol Life Sci 2021; 78:6409-6430. [PMID: 34405255 PMCID: PMC8558161 DOI: 10.1007/s00018-021-03907-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/24/2022]
Abstract
Neuroserpin is a serine protease inhibitor identified in a search for proteins implicated in neuronal axon growth and synapse formation. Since its discovery over 30 years ago, it has been the focus of active research. Many efforts have concentrated in elucidating its neuroprotective role in brain ischemic lesions, the structural bases of neuroserpin conformational change and the effects of neuroserpin polymers that underlie the neurodegenerative disease FENIB (familial encephalopathy with neuroserpin inclusion bodies), but the investigation of the physiological roles of neuroserpin has increased over the last years. In this review, we present an updated and critical revision of the current literature dealing with neuroserpin, covering all aspects of research including the expression and physiological roles of neuroserpin, both inside and outside the nervous system; its inhibitory and non-inhibitory mechanisms of action; the molecular structure of the monomeric and polymeric conformations of neuroserpin, including a detailed description of the polymerisation mechanism; and the involvement of neuroserpin in human disease, with particular emphasis on FENIB. Finally, we briefly discuss the identification by genome-wide screening of novel neuroserpin variants and their possible pathogenicity.
Collapse
Affiliation(s)
- Emanuela D'Acunto
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Rome, Italy
| | - Annamaria Fra
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Cristina Visentin
- Department of Biosciences, University of Milan, Milan, Italy
- Institute of Molecular and Translational Cardiology, I.R.C.C.S. Policlinico San Donato, Milan, Italy
| | - Mauro Manno
- Institute of Biophysics, National Research Council of Italy, Palermo, Italy
| | - Stefano Ricagno
- Department of Biosciences, University of Milan, Milan, Italy
| | - Giovanna Galliciotti
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elena Miranda
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Rome, Italy.
- Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
6
|
Anfray A, Brodin C, Drieu A, Potzeha F, Dalarun B, Agin V, Vivien D, Orset C. Single- and two- chain tissue type plasminogen activator treatments differentially influence cerebral recovery after stroke. Exp Neurol 2021; 338:113606. [PMID: 33453214 DOI: 10.1016/j.expneurol.2021.113606] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 10/22/2022]
Abstract
Tissue type Plasminogen Activator (tPA), named alteplase (Actilyse®) under its commercial form, is currently the only pharmacological treatment approved during the acute phase of ischemic stroke, used either alone or combined with thrombectomy. Interestingly, the commercial recombinant tPA (rtPA) contains two physiological forms of rtPA: the single chain rtPA (sc-rtPA) and the two-chains rtPA (tc-rtPA), with differential properties demonstrated in vitro. Using a relevant mouse model of thromboembolic stroke, we have investigated the overall effects of these two forms of rtPA when infused early after stroke onset (i.e. 20 min) on recanalization, lesion volumes, alterations of the integrity of the blood brain barrier and functional recovery. Our data reveal that there is no difference in the capacity of sc-rtPA and tc-rtPA to promote fibrinolysis and reperfusion of the tissue. However, compared to sc-rtPA, tc-rtPA is less efficient to reduce lesion volumes and to improve functional recovery, and is associated with an increased opening of the blood brain barrier. These data indicate better understanding of differential effects of these tPA forms might be important to ultimately improve stroke treatment.
Collapse
Affiliation(s)
- Antoine Anfray
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France
| | - Camille Brodin
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France
| | - Antoine Drieu
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France
| | - Fanny Potzeha
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France
| | - Basile Dalarun
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France
| | - Véronique Agin
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France; CHU Caen, Department of Clinical Research, Caen University Hospital, Avenue de la Côte de Nacre, Caen, France.
| | - Cyrille Orset
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France
| |
Collapse
|
7
|
Visentin C, Musso L, Broggini L, Bonato F, Russo R, Moriconi C, Bolognesi M, Miranda E, Dallavalle S, Passarella D, Ricagno S. Embelin as Lead Compound for New Neuroserpin Polymerization Inhibitors. Life (Basel) 2020; 10:life10070111. [PMID: 32664592 PMCID: PMC7400170 DOI: 10.3390/life10070111] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/25/2020] [Accepted: 07/09/2020] [Indexed: 12/13/2022] Open
Abstract
Familial encephalopathy with neuroserpin inclusion bodies (FENIB) is a severe and lethal neurodegenerative disease. Upon specific point mutations in the SERPINI1gene-coding for the human protein neuroserpin (NS) the resulting pathologic NS variants polymerize and accumulate within the endoplasmic reticulum of neurons in the central nervous system. To date, embelin (EMB) is the only known inhibitor of NS polymerization in vitro. This molecule is capable of preventing NS polymerization and dissolving preformed polymers. Here, we show that lowering EMB concentration results in increasing size of NS oligomers in vitro. Moreover, we observe that in cells expressing NS, the polymerization of G392E NS is reduced, but this effect is mediated by an increased proteasomal degradation rather than polymerization impairment. For these reasons we designed a systematic chemical evolution of the EMB scaffold aimed to improve its anti-polymerization properties. The effect of EMB analogs against NS polymerization was assessed in vitro. None of the EMB analogs displayed an anti-polymerization activity better than the one reported for EMB, indicating that the EMB–NS interaction surface is very specific and highly optimized. Thus, our results indicate that EMB is, to date, still the best candidate for developing a treatment against NS polymerization.
Collapse
Affiliation(s)
- Cristina Visentin
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria, 26, 20133 Milan, Italy; (C.V.); (L.B.); (M.B.)
| | - Loana Musso
- Dipartimento di Scienze per gli Alimenti, la Nutrizione e l’Ambiente, Università degli Studi di Milano, Via Celoria, 2, 20133 Milan, Italy; (L.M.); (S.D.)
| | - Luca Broggini
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria, 26, 20133 Milan, Italy; (C.V.); (L.B.); (M.B.)
| | - Francesca Bonato
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi, 19, 20133 Milan, Italy; (F.B.); (D.P.)
| | - Rosaria Russo
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, Via Fratelli Cervi, 93, 20090 Segrate, Italy;
| | - Claudia Moriconi
- Dipartimento di Biologia e Biotecnologie ‘Charles Darwin’, Sapienza Università di Roma, Piazzale Aldo Moro, 5, 00185 Rome, Italy; (C.M.); (E.M.)
| | - Martino Bolognesi
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria, 26, 20133 Milan, Italy; (C.V.); (L.B.); (M.B.)
| | - Elena Miranda
- Dipartimento di Biologia e Biotecnologie ‘Charles Darwin’, Sapienza Università di Roma, Piazzale Aldo Moro, 5, 00185 Rome, Italy; (C.M.); (E.M.)
- Istituto Pasteur—Cenci Bolognetti Foundation, Sapienza Università di Roma, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Sabrina Dallavalle
- Dipartimento di Scienze per gli Alimenti, la Nutrizione e l’Ambiente, Università degli Studi di Milano, Via Celoria, 2, 20133 Milan, Italy; (L.M.); (S.D.)
| | - Daniele Passarella
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi, 19, 20133 Milan, Italy; (F.B.); (D.P.)
| | - Stefano Ricagno
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria, 26, 20133 Milan, Italy; (C.V.); (L.B.); (M.B.)
- Correspondence: ; Tel.: +39-02-5031-4914
| |
Collapse
|
8
|
Visentin C, Broggini L, Sala BM, Russo R, Barbiroli A, Santambrogio C, Nonnis S, Dubnovitsky A, Bolognesi M, Miranda E, Achour A, Ricagno S. Glycosylation Tunes Neuroserpin Physiological and Pathological Properties. Int J Mol Sci 2020; 21:E3235. [PMID: 32375228 PMCID: PMC7247563 DOI: 10.3390/ijms21093235] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 02/03/2023] Open
Abstract
Neuroserpin (NS) is a member of the serine protease inhibitors superfamily. Specific point mutations are responsible for its accumulation in the endoplasmic reticulum of neurons that leads to a pathological condition named familial encephalopathy with neuroserpin inclusion bodies (FENIB). Wild-type NS presents two N-glycosylation chains and does not form polymers in vivo, while non-glycosylated NS causes aberrant polymer accumulation in cell models. To date, all in vitro studies have been conducted on bacterially expressed NS, de facto neglecting the role of glycosylation in the biochemical properties of NS. Here, we report the expression and purification of human glycosylated NS (gNS) using a novel eukaryotic expression system, LEXSY. Our results confirm the correct N-glycosylation of wild-type gNS. The fold and stability of gNS are not altered compared to bacterially expressed NS, as demonstrated by the circular dichroism and intrinsic tryptophan fluorescence assays. Intriguingly, gNS displays a remarkably reduced polymerisation propensity compared to non-glycosylated NS, in keeping with what was previously observed for wild-type NS in vivo and in cell models. Thus, our results support the relevance of gNS as a new in vitro tool to study the molecular bases of FENIB.
Collapse
Affiliation(s)
- Cristina Visentin
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria, 26, 20133 Milan, Italy; (C.V.); (L.B.); (B.M.S.); (M.B.)
| | - Luca Broggini
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria, 26, 20133 Milan, Italy; (C.V.); (L.B.); (B.M.S.); (M.B.)
| | - Benedetta Maria Sala
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria, 26, 20133 Milan, Italy; (C.V.); (L.B.); (B.M.S.); (M.B.)
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, and Division of Infectious Diseases, Karolinska University Hospital, Solna, SE-17176 Stockholm, Sweden;
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, AlbaNova University Center, Royal Institute of Technology, SE-10691 Stockholm, Sweden
| | - Rosaria Russo
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, Via Fratelli Cervi, 93, 20090 Segrate, Italy;
| | - Alberto Barbiroli
- Dipartimento di Scienze per gli Alimenti, la Nutrizione e l′Ambiente, Università degli Studi di Milano, Via Celoria, 2, 20133 Milan, Italy;
| | - Carlo Santambrogio
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi di Milano-Bicocca, Piazza dell’Ateneo Nuovo, 1, 20126 Milan, Italy;
| | - Simona Nonnis
- Departimento di Medicina Veterinaria, Università degli Studi di Milano, Via dell’Università, 6, 26900 Lodi, Italy;
| | - Anatoly Dubnovitsky
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, and Division of Rheumatology, Karolinska University Hospital, Solna, SE-17176 Stockholm, Sweden;
| | - Martino Bolognesi
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria, 26, 20133 Milan, Italy; (C.V.); (L.B.); (B.M.S.); (M.B.)
| | - Elena Miranda
- Dipartimento di Biologia e Biotecnologie ‘Charles Darwin’, and Istituto Pasteur - Fondazione Cenci-Bolognetti, Sapienza Università di Roma, Piazzale Aldo Moro, 5, 00185 Rome, Italy;
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, and Division of Infectious Diseases, Karolinska University Hospital, Solna, SE-17176 Stockholm, Sweden;
| | - Stefano Ricagno
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria, 26, 20133 Milan, Italy; (C.V.); (L.B.); (B.M.S.); (M.B.)
| |
Collapse
|
9
|
Ali MF, Kaushik A, Gupta D, Ansari S, Jairajpuri MA. Changes in strand 6B and helix B during neuroserpin inhibition: Implication in severity of clinical phenotype. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2020; 1868:140363. [PMID: 31954927 DOI: 10.1016/j.bbapap.2020.140363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 12/19/2019] [Accepted: 01/15/2020] [Indexed: 12/13/2022]
Abstract
Neuroserpin (NS) is predominantly expressed in brain and inhibits tissue-type plasminogen activator (tPA) with implications in brain development and memory. Nature of conformational change in pathological variants in strand 6B and helix B of NS that cause a relatively mild to severe epilepsy (and/or dementia) remains largely elusive. MD simulation with wild type (WT) NS, strand 6B and helix B variants indicated that substitution in this region affects the conformation of the strands 5B, 5A and reactive centre loop. Therefore, we designed variants of NS in strand 6B (I46D and F48S) and helix B (A54F, L55A and L55P) to investigate their role in tPA inhibition mechanism and propensity to aggregate. An interaction analysis showed disturbance of a hydrophobic patch centered at strands 5B, 6B and helix B in I46D and F48S but not in A54F, L55A, L55P and WT NS. Purified I46D, F48S and L55P variants showed decrease in fluorescence emission intensity but have similar α-helical content, however results of A54F and L55A were comparable to WT NS. Analysis of tPA inhibition showed marginal effect on A54F and L55A variant with tPA-NS complex formation. In contrast, I46D, F48S and L55P variants showed massive decrease in tPA inhibition, with no tPA-NS complex formation. Analysis of native PAGE under under polymerization condition showed prompt conversion of I46D, F48S and L55P to latent conformation but not A54F and L55A variants. Identification of these novel conformational changes will aid in the understanding of variable clinical phenotype of shutter region NS variants and other serpins.
Collapse
Affiliation(s)
- Mohammad Farhan Ali
- Protein Conformation and Enzymology Lab, Department of Biosciences, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| | - Abhinav Kaushik
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Dinesh Gupta
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Shoyab Ansari
- Protein Conformation and Enzymology Lab, Department of Biosciences, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| | - Mohamad Aman Jairajpuri
- Protein Conformation and Enzymology Lab, Department of Biosciences, Jamia Millia Islamia (A Central University), New Delhi 110025, India.
| |
Collapse
|
10
|
Zhu J, Wan Y, Xu H, Wu Y, Hu B, Jin H. The role of endogenous tissue-type plasminogen activator in neuronal survival after ischemic stroke: friend or foe? Cell Mol Life Sci 2019; 76:1489-1506. [PMID: 30656378 PMCID: PMC11105644 DOI: 10.1007/s00018-019-03005-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/19/2018] [Accepted: 01/03/2019] [Indexed: 12/29/2022]
Abstract
Endogenous protease tissue-type plasminogen activator (tPA) has highly efficient fibrinolytic activity and its recombinant variants alteplase and tenecteplase are established as highly effective thrombolytic drugs for ischemic stroke. Endogenous tPA is constituted of five functional domains through which it interacts with a variety of substrates, binding proteins and receptors, thus having enzymatic and cytokine-like effects to act on all cell types of the brain. In the past 2 decades, numerous studies have explored the clinical relevance of endogenous tPA in neurological diseases, especially in ischemic stroke. tPA is released from many cells within the brain parenchyma exposed to ischemia conditions in vitro and in vivo, which is believed to control neuronal fate. Some studies proved that tPA could induce blood-brain barrier disruption, neural excitotoxicity and inflammation, while others indicated that tPA also has anti-excitotoxic, neurotrophic and anti-apoptotic effects on neurons. Therefore, more work is needed to elucidate how tPA mediates such opposing functions that may amplify tPA from a therapeutic means into a key therapeutic target in endogenous neuroprotection after stroke. In this review, we summarize the biological characteristics and pleiotropic functions of tPA in the brain. Then we focus on possible hypotheses about why and how endogenous tPA mediates ischemic neuronal death and survival. Finally, we analyze how endogenous tPA affects neuron fate in ischemic stroke in a comprehensive view.
Collapse
Affiliation(s)
- Jiayi Zhu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yan Wan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Hexiang Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yulang Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| | - Huijuan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| |
Collapse
|
11
|
Millar LJ, Shi L, Hoerder-Suabedissen A, Molnár Z. Neonatal Hypoxia Ischaemia: Mechanisms, Models, and Therapeutic Challenges. Front Cell Neurosci 2017; 11:78. [PMID: 28533743 PMCID: PMC5420571 DOI: 10.3389/fncel.2017.00078] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/07/2017] [Indexed: 12/11/2022] Open
Abstract
Neonatal hypoxia-ischaemia (HI) is the most common cause of death and disability in human neonates, and is often associated with persistent motor, sensory, and cognitive impairment. Improved intensive care technology has increased survival without preventing neurological disorder, increasing morbidity throughout the adult population. Early preventative or neuroprotective interventions have the potential to rescue brain development in neonates, yet only one therapeutic intervention is currently licensed for use in developed countries. Recent investigations of the transient cortical layer known as subplate, especially regarding subplate's secretory role, opens up a novel set of potential molecular modulators of neonatal HI injury. This review examines the biological mechanisms of human neonatal HI, discusses evidence for the relevance of subplate-secreted molecules to this condition, and evaluates available animal models. Neuroserpin, a neuronally released neuroprotective factor, is discussed as a case study for developing new potential pharmacological interventions for use post-ischaemic injury.
Collapse
Affiliation(s)
- Lancelot J. Millar
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| | - Lei Shi
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan UniversityGuangzhou, China
| | | | - Zoltán Molnár
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| |
Collapse
|
12
|
Ali MF, Kaushik A, Kapil C, Gupta D, Jairajpuri MA. A hydrophobic patch surrounding Trp154 in human neuroserpin controls the helix F dynamics with implications in inhibition and aggregation. Sci Rep 2017; 7:42987. [PMID: 28230174 PMCID: PMC5322333 DOI: 10.1038/srep42987] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 01/17/2017] [Indexed: 01/23/2023] Open
Abstract
Neuroserpin (NS) mediated inhibition of tissue-type plasminogen activator (tPA) is important for brain development, synapse formation and memory. Aberrations in helix F and β-sheet A movement during inhibition can directly lead to epilepsy or dementia. Conserved W154 residue in a hydrophobic patch between helix F and β-sheet A is ideally placed to control their movement during inhibition. Molecular Dynamics (MD) simulation on wild type (WT) NS and its two variants (W154A and W154P) demonstrated partial deformation in helix F and conformational differences in strands 1A and 2A only in W154P. A fluorescence and Circular Dichroism (CD) analysis with purified W154 variants revealed a significant red-shift and an increase in α-helical content in W154P as compared to W154A and WT NS. Kinetics of tPA inhibition showed a decline in association rates (ka) for W154A as compared to WT NS with indication of complex formation. Appearance of cleaved without complex formation in W154P indicates that the variant acts as substrate due to conformational misfolding around helix F. Both the variants however showed increased rate of aggregation as compared to WT NS. The hydrophobic patch identified in this study may have importance in helix F dynamics of NS.
Collapse
Affiliation(s)
- Mohammad Farhan Ali
- Protein Conformation and Enzymology Lab, Department of Biosciences, Jamia Millia Islamia (A Central University), New Delhi-110025, India
| | - Abhinav Kaushik
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi-110067, India
| | - Charu Kapil
- Protein Conformation and Enzymology Lab, Department of Biosciences, Jamia Millia Islamia (A Central University), New Delhi-110025, India
| | - Dinesh Gupta
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi-110067, India
| | - Mohamad Aman Jairajpuri
- Protein Conformation and Enzymology Lab, Department of Biosciences, Jamia Millia Islamia (A Central University), New Delhi-110025, India
| |
Collapse
|
13
|
Lee TW, Tsang VWK, Loef EJ, Birch NP. Physiological and pathological functions of neuroserpin: Regulation of cellular responses through multiple mechanisms. Semin Cell Dev Biol 2017; 62:152-159. [PMID: 27639894 DOI: 10.1016/j.semcdb.2016.09.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 09/09/2016] [Accepted: 09/12/2016] [Indexed: 11/17/2022]
Abstract
It is 27 years since neuroserpin was first discovered in the nervous system and identified as a member of the serpin superfamily. Since that time potential roles for this serine protease inhibitor have been identified in neuronal and non-neuronal systems. Many are linked to inhibition of neuroserpin's principal enzyme target, tissue plasminogen activator (tPA), although some have been suggested to involve alternate non-inhibitory mechanisms. This review focuses mainly on the inhibitory roles of neuroserpin and discusses the evidence supporting tPA as the physiological target. While the major sites of neuroserpin expression are neural, endocrine and immune tissues, most progress on characterizing functional roles for neuroserpin have been in the brain. Roles in emotional behaviour, synaptic plasticity and neuroprotection in stroke and excitotoxicity models are discussed. Current knowledge on three neurological diseases associated with neuroserpin mutation or activity, Familial Encephalopathy with Neuroserpin Inclusion Bodies (FENIB), Alzheimer's disease and brain metastasis is presented. Finally, we consider mechanistic studies that have revealed a distinct inhibitory mechanism for neuroserpin and its possible implications for neuroserpin function.
Collapse
Affiliation(s)
- Tet Woo Lee
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand.
| | - Vicky W K Tsang
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Evert Jan Loef
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Nigel P Birch
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand; Brain Research New Zealand, Rangahau Roro Aotearoa, Auckland, New Zealand.
| |
Collapse
|
14
|
Carlson KSB, Nguyen L, Schwartz K, Lawrence DA, Schwartz BS. Neuroserpin Differentiates Between Forms of Tissue Type Plasminogen Activator via pH Dependent Deacylation. Front Cell Neurosci 2016; 10:154. [PMID: 27378851 PMCID: PMC4908126 DOI: 10.3389/fncel.2016.00154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 05/27/2016] [Indexed: 11/17/2022] Open
Abstract
Tissue-type plasminogen activator (t-PA), initially characterized for its critical role in fibrinolysis, also has key functions in both physiologic and pathologic processes in the CNS. Neuroserpin (NSP) is a t-PA specific serine protease inhibitor (serpin) found almost exclusively in the CNS that regulates t-PA's proteolytic activity and protects against t-PA mediated seizure propagation and blood-brain barrier disruption. This report demonstrates that NSP inhibition of t-PA varies profoundly as a function of pH within the biologically relevant pH range for the CNS, and reflects the stability, rather than the formation of NSP: t-PA acyl-enzyme complexes. Moreover, NSP differentiates between the zymogen-like single chain form (single chain t-PA, sct-PA) and the mature protease form (two chain t-PA, tct-PA) of t-PA, demonstrating different pH profiles for protease inhibition, different pH ranges over which catalytic deacylation occurs, and different pH dependent profiles of deacylation rates for each form of t-PA. NSP's pH dependent inhibition of t-PA is not accounted for by differential acylation, and is specific for the NSP-t-PA serpin-protease pair. These results demonstrate a novel mechanism for the differential regulation of the two forms of t-PA in the CNS, and suggest a potential specific regulatory role for CNS pH in controlling t-PA proteolytic activity.
Collapse
Affiliation(s)
- Karen-Sue B. Carlson
- Department of Biomolecular Chemistry, University of Wisconsin, MadisonWI, USA
- Medical Scientist Training Program, University of Wisconsin, MadisonWI, USA
| | - Lan Nguyen
- Departments of Biochemistry and Medicine, University of Illinois, UrbanaIL, USA
| | - Kat Schwartz
- Departments of Biochemistry and Medicine, University of Illinois, UrbanaIL, USA
| | - Daniel A. Lawrence
- Departments of Medicine and Molecular and Integrative Physiology, University of Michigan, Ann ArborMI, USA
| | - Bradford S. Schwartz
- Department of Biomolecular Chemistry, University of Wisconsin, MadisonWI, USA
- Departments of Biochemistry and Medicine, University of Illinois, UrbanaIL, USA
| |
Collapse
|
15
|
Hébert M, Lesept F, Vivien D, Macrez R. The story of an exceptional serine protease, tissue-type plasminogen activator (tPA). Rev Neurol (Paris) 2015; 172:186-97. [PMID: 26626577 DOI: 10.1016/j.neurol.2015.10.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/08/2015] [Accepted: 10/04/2015] [Indexed: 12/17/2022]
Abstract
The only acute treatment of ischemic stroke approved by the health authorities is tissue recombinant plasminogen activator (tPA)-induced thrombolysis. Under physiological conditions, tPA, belonging to the serine protease family, is secreted by endothelial and brain cells (neurons, astrocytes, microglia, oligodendrocytes). Although revascularisation induced by tPA is beneficial during a stroke, research over the past 20 years shows that tPA can also be deleterious for the brain parenchyma. Thus, in this review of the literature, after a brief history on the discovery of tPA, we reviewed current knowledge of mechanisms by which tPA can influence brain function in physiological and pathological conditions.
Collapse
Affiliation(s)
- M Hébert
- Inserm, UMR-S U919 serine proteases and pathophysiology of the neurovascular unit, 14000 Caen, France
| | - F Lesept
- Inserm, UMR-S U919 serine proteases and pathophysiology of the neurovascular unit, 14000 Caen, France
| | - D Vivien
- Inserm, UMR-S U919 serine proteases and pathophysiology of the neurovascular unit, 14000 Caen, France
| | - R Macrez
- Inserm, UMR-S U919 serine proteases and pathophysiology of the neurovascular unit, 14000 Caen, France.
| |
Collapse
|
16
|
The plasminogen activation system in neuroinflammation. Biochim Biophys Acta Mol Basis Dis 2015; 1862:395-402. [PMID: 26493446 DOI: 10.1016/j.bbadis.2015.10.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 10/08/2015] [Accepted: 10/15/2015] [Indexed: 01/30/2023]
Abstract
The plasminogen activation (PA) system consists in a group of proteases and protease inhibitors regulating the activation of the zymogen plasminogen into its proteolytically active form, plasmin. Here, we give an update of the current knowledge about the role of the PA system on different aspects of neuroinflammation. These include modification in blood-brain barrier integrity, leukocyte diapedesis, removal of fibrin deposits in nervous tissues, microglial activation and neutrophil functions. Furthermore, we focus on the molecular mechanisms (some of them independent of plasmin generation and even of proteolysis) and target receptors responsible for these effects. The description of these mechanisms of action may help designing new therapeutic strategies targeting the expression, activity and molecular mediators of the PA system in neurological disorders involving neuroinflammatory processes. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
|
17
|
Chevilley A, Lesept F, Lenoir S, Ali C, Parcq J, Vivien D. Impacts of tissue-type plasminogen activator (tPA) on neuronal survival. Front Cell Neurosci 2015; 9:415. [PMID: 26528141 PMCID: PMC4607783 DOI: 10.3389/fncel.2015.00415] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 10/01/2015] [Indexed: 11/18/2022] Open
Abstract
Tissue-type plasminogen activator (tPA) a serine protease is constituted of five functional domains through which it interacts with different substrates, binding proteins, and receptors. In the last years, great interest has been given to the clinical relevance of targeting tPA in different diseases of the central nervous system, in particular stroke. Among its reported functions in the central nervous system, tPA displays both neurotrophic and neurotoxic effects. How can the protease mediate such opposite functions remain unclear but several hypotheses have been proposed. These include an influence of the degree of maturity and/or the type of neurons, of the level of tPA, of its origin (endogenous or exogenous) or of its form (single chain tPA versus two chain tPA). In this review, we will provide a synthetic snapshot of our current knowledge regarding the natural history of tPA and discuss how it sustains its pleiotropic functions with focus on excitotoxic/ischemic neuronal death and neuronal survival.
Collapse
Affiliation(s)
- Arnaud Chevilley
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| | - Flavie Lesept
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| | - Sophie Lenoir
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| | - Carine Ali
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| | - Jérôme Parcq
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| | - Denis Vivien
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| |
Collapse
|
18
|
Lee TW, Tsang VWK, Birch NP. Physiological and pathological roles of tissue plasminogen activator and its inhibitor neuroserpin in the nervous system. Front Cell Neurosci 2015; 9:396. [PMID: 26528129 PMCID: PMC4602146 DOI: 10.3389/fncel.2015.00396] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 09/22/2015] [Indexed: 12/03/2022] Open
Abstract
Although its roles in the vascular space are most well-known, tissue plasminogen activator (tPA) is widely expressed in the developing and adult nervous system, where its activity is believed to be regulated by neuroserpin, a predominantly brain-specific member of the serpin family of protease inhibitors. In the normal physiological state, tPA has been shown to play roles in the development and plasticity of the nervous system. Ischemic damage, however, may lead to excess tPA activity in the brain and this is believed to contribute to neurodegeneration. In this article, we briefly review the physiological and pathological roles of tPA in the nervous system, which includes neuronal migration, axonal growth, synaptic plasticity, neuroprotection and neurodegeneration, as well as a contribution to neurological disease. We summarize tPA's multiple mechanisms of action and also highlight the contributions of the inhibitor neuroserpin to these processes.
Collapse
Affiliation(s)
- Tet Woo Lee
- School of Biological Sciences and Centre for Brain Research, University of Auckland Auckland, New Zealand
| | - Vicky W K Tsang
- School of Biological Sciences and Centre for Brain Research, University of Auckland Auckland, New Zealand
| | - Nigel P Birch
- School of Biological Sciences and Centre for Brain Research, University of Auckland Auckland, New Zealand ; Brain Research New Zealand, Rangahau Roro Aotearoa Auckland, New Zealand
| |
Collapse
|
19
|
Caccia S, Ricagno S, Bolognesi M. Molecular bases of neuroserpin function and pathology. Biomol Concepts 2015; 1:117-30. [PMID: 25961991 DOI: 10.1515/bmc.2010.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Serpins build a large and evolutionary widespread protein superfamily, hosting members that are mainly Ser-protease inhibitors. Typically, serpins display a conserved core domain composed of three main β-sheets and 9-10 α-helices, for a total of approximately 350 amino acids. Neuroserpin (NS) is mostly expressed in neurons and in the central and peripheral nervous systems, where it targets tissue-type plasminogen activator. NS activity is relevant for axogenesis, synaptogenesis and synaptic plasticity. Five (single amino acid) NS mutations are associated with severe neurodegenerative disease in man, leading to early onset dementia, epilepsy and neuronal death. The functional aspects of NS protease inhibition are linked to the presence of a long exposed loop (reactive center loop, RCL) that acts as bait for the incoming partner protease. Large NS conformational changes, associated with the cleavage of the RCL, trap the protease in an acyl-enzyme complex. Contrary to other serpins, this complex has a half-life of approximately 10 min. Conformational flexibility is held to be at the bases of NS polymerization leading to Collins bodies intracellular deposition and neuronal damage in the pathological NS variants. Two main general mechanisms of serpin polymerization are currently discussed. Both models require the swapping of the RCL among neighboring serpin molecules. Specific differences in the size of swapped regions, as well as differences in the folding stage at which polymerization can occur, distinguish the two models. The results provided by recent crystallographic and biophysical studies allow rationalization of the functional and pathological roles played by NS based on the analysis of four three-dimensional structures.
Collapse
|
20
|
Lee TW, Yang ASP, Brittain T, Birch NP. An analysis approach to identify specific functional sites in orthologous proteins using sequence and structural information: application to neuroserpin reveals regions that differentially regulate inhibitory activity. Proteins 2015; 83:135-52. [PMID: 25363759 DOI: 10.1002/prot.24711] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 10/22/2014] [Accepted: 10/27/2014] [Indexed: 01/12/2023]
Abstract
The analysis of sequence conservation is commonly used to predict functionally important sites in proteins. We have developed an approach that first identifies highly conserved sites in a set of orthologous sequences using a weighted substitution-matrix-based conservation score and then filters these conserved sites based on the pattern of conservation present in a wider alignment of sequences from the same family and structural information to identify surface-exposed sites. This allows us to detect specific functional sites in the target protein and exclude regions that are likely to be generally important for the structure or function of the wider protein family. We applied our method to two members of the serpin family of serine protease inhibitors. We first confirmed that our method successfully detected the known heparin binding site in antithrombin while excluding residues known to be generally important in the serpin family. We next applied our sequence analysis approach to neuroserpin and used our results to guide site-directed polyalanine mutagenesis experiments. The majority of the mutant neuroserpin proteins were found to fold correctly and could still form inhibitory complexes with tissue plasminogen activator (tPA). Kinetic analysis of tPA inhibition, however, revealed altered inhibitory kinetics in several of the mutant proteins, with some mutants showing decreased association with tPA and others showing more rapid dissociation of the covalent complex. Altogether, these results confirm that our sequence analysis approach is a useful tool that can be used to guide mutagenesis experiments for the detection of specific functional sites in proteins.
Collapse
Affiliation(s)
- Tet Woo Lee
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | | | | | | |
Collapse
|
21
|
Tsang VWK, Young D, During MJ, Birch NP. AAV-mediated overexpression of neuroserpin in the hippocampus decreases PSD-95 expression but does not affect hippocampal-dependent learning and memory. PLoS One 2014; 9:e91050. [PMID: 24608243 PMCID: PMC3946662 DOI: 10.1371/journal.pone.0091050] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 02/07/2014] [Indexed: 01/06/2023] Open
Abstract
Neuroserpin is a serine protease inhibitor, or serpin, that is expressed in the nervous system and inhibits the protease tissue plasminogen activator (tPA). Neuroserpin has been suggested to play a role in learning and memory but direct evidence for such a role is lacking. Here we have used an adeno-associated virus (AAV) vector expression system to investigate the effect of neuroserpin on hippocampal-dependent learning and memory in the young adult rat. A FLAG-tagged neuroserpin construct was initially characterized by in vitro transcription/translation and transfection into HEK293 cells and shown to interact with tPA and be targeted to the secretory pathway. Targeted injection of a chimeric AAV1/2 vector expressing FLAG-neuroserpin resulted in localized overexpression in the dorsal hippocampus. Neuroserpin overexpression led to the appearance of an unstable neuroserpin:tPA complex in zymographic assays consistent with interaction with endogenous tPA in vivo. Rats overexpressing neuroserpin also showed a significant decrease in the levels of postsynaptic density protein 95, a major postsynaptic scaffolding protein. Three weeks after injection, a range of behavioural tests was performed to measure spatial and associative learning and memory, as well as innate and acquired fear. These tests provided no evidence of a role for neuroserpin in hippocampal-dependent learning and memory. In summary this study does not support a role for neuroserpin in hippocampal-dependent learning and memory in young adult rats but does suggest an involvement of neuroserpin in hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- Vicky W. K. Tsang
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Deborah Young
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Matthew J. During
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University, Columbus, Ohio, United States of America
| | - Nigel P. Birch
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
22
|
Lasierra-Cirujeda J, Coronel P, Aza M, Gimeno M. Beta-amyloidolysis and glutathione in Alzheimer's disease. J Blood Med 2013; 4:31-8. [PMID: 23650462 PMCID: PMC3640603 DOI: 10.2147/jbm.s35496] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In this review, we hypothesized the importance of the interaction between the brain glutathione (GSH) system, the proteolytic tissue plasminogen activator (t-PA)/plasminogen/ plasmin system, regulated by plasminogen activator inhibitor (PAI-1), and neuroserpin in the pathogenesis of Alzheimer’s disease. The histopathological characteristic hallmark that gives personality to the diagnosis of Alzheimer’s disease is the accumulation of neurofibroid tangles located intracellularly in the brain, such as the protein tau and extracellular senile plaques made primarily of amyloidal substance. These formations of complex etiology are intimately related to GSH, brain protective antioxidants, and the proteolytic system, in which t-PA plays a key role. There is scientific evidence that suggests a relationship between aging, a number of neurodegenerative disorders, and the excessive production of reactive oxygen species and accompanying decreased brain proteolysis. The plasminogen system in the brain is an essential proteolytic mechanism that effectively degrades amyloid peptides (“beta-amyloidolysis”) through action of the plasmin, and this physiologic process may be considered to be a means of prevention of neurodegenerative disorders. In parallel to the decrease in GSH levels seen in aging, there is also a decrease in plasmin brain activity and a progressive decrease of t-PA activity, caused by a decrease in the expression of the t-PA together with an increase of the PAI-1 levels, which rise to an increment in the production of amyloid peptides and a lesser clearance of them. Better knowledge of the GSH mechanism and cerebral proteolysis will allow us to hypothesize about therapeutic practices.
Collapse
|
23
|
Parcq J, Bertrand T, Montagne A, Baron AF, Macrez R, Billard JM, Briens A, Hommet Y, Wu J, Yepes M, Lijnen HR, Dutar P, Anglés-Cano E, Vivien D. Unveiling an exceptional zymogen: the single-chain form of tPA is a selective activator of NMDA receptor-dependent signaling and neurotoxicity. Cell Death Differ 2012; 19:1983-91. [PMID: 22743997 DOI: 10.1038/cdd.2012.86] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Unlike other serine proteases that are zymogens, the single-chain form of tissue plasminogen activator (sc-tPA) exhibits an intrinsic activity similar to that of its cleaved two-chain form (tc-tPA), especially in the presence of fibrin. In the central nervous system tPA controls brain functions and dysfunctions through its proteolytic activity. We demonstrated here, both in vitro and in vivo, that the intrinsic activity of sc-tPA selectively modulates N-methyl-D-aspartate receptor (NMDAR) signaling as compared with tc-tPA. Thus, sc-tPA enhances NMDAR-mediated calcium influx, Erk(½) activation and neurotoxicity in cultured cortical neurons, excitotoxicity in the striatum and NMDAR-dependent long-term potentiation in the hippocampal CA-1 network. As the first demonstration of a differential function for sc-tPA and tc-tPA, this finding opens a new area of investigations on tPA functions in the absence of its allosteric regulator, fibrin.
Collapse
Affiliation(s)
- J Parcq
- Inserm U919, Serine Proteases and Pathophysiology of the neurovascular Unit, Université de Caen Basse-Normandie, GIP Cyceron, Caen, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lee TW, Montgomery JM, Birch NP. The serine protease inhibitor neuroserpin regulates the growth and maturation of hippocampal neurons through a non-inhibitory mechanism. J Neurochem 2012; 121:561-74. [PMID: 22191421 DOI: 10.1111/j.1471-4159.2011.07639.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Neuroserpin is a brain-specific serine protease inhibitor that is expressed in the developing and adult nervous system. Its expression profile led to suggestions that it played roles in neuronal growth and connectivity. In this study, we provide direct evidence to support a role for neuroserpin in axon and dendritic growth. We report that axon growth is enhanced while axon and dendrite diameter are reduced following neuroserpin treatment of hippocampal neurons. More complex effects are seen on dendritic growth and branching with neuroserpin-stimulating dendritic growth and branching in young neurons but switching to an inhibitory response in older neurons. The protease inhibitory activity of neuroserpin is not required to activate changes in neuronal morphology and a proportion of responses are modulated by an antagonist to the LRP1 receptor. Collectively, these findings support a key role for neuroserpin as a regulator of neuronal development through a non-inhibitory mechanism and suggest a basis for neuroserpin's effects on complex emotional behaviours and recent link to schizophrenia.
Collapse
Affiliation(s)
- Tet Woo Lee
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | | | | |
Collapse
|
25
|
Noto R, Santangelo MG, Ricagno S, Mangione MR, Levantino M, Pezzullo M, Martorana V, Cupane A, Bolognesi M, Manno M. The tempered polymerization of human neuroserpin. PLoS One 2012; 7:e32444. [PMID: 22412873 PMCID: PMC3295756 DOI: 10.1371/journal.pone.0032444] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 01/31/2012] [Indexed: 11/24/2022] Open
Abstract
Neuroserpin, a member of the serpin protein superfamily, is an inhibitor of proteolytic activity that is involved in pathologies such as ischemia, Alzheimer's disease, and Familial Encephalopathy with Neuroserpin Inclusion Bodies (FENIB). The latter belongs to a class of conformational diseases, known as serpinopathies, which are related to the aberrant polymerization of serpin mutants. Neuroserpin is known to polymerize, even in its wild type form, under thermal stress. Here, we study the mechanism of neuroserpin polymerization over a wide range of temperatures by different techniques. Our experiments show how the onset of polymerization is dependent on the formation of an intermediate monomeric conformer, which then associates with a native monomer to yield a dimeric species. After the formation of small polymers, the aggregation proceeds via monomer addition as well as polymer-polymer association. No further secondary mechanism takes place up to very high temperatures, thus resulting in the formation of neuroserpin linear polymeric chains. Most interesting, the overall aggregation is tuned by the co-occurrence of monomer inactivation (i.e. the formation of latent neuroserpin) and by a mechanism of fragmentation. The polymerization kinetics exhibit a unique modulation of the average mass and size of polymers, which might suggest synchronization among the different processes involved. Thus, fragmentation would control and temper the aggregation process, instead of enhancing it, as typically observed (e.g.) for amyloid fibrillation.
Collapse
Affiliation(s)
- Rosina Noto
- Institute of Biophysics, National Research Council of Italy, Palermo, Italy
| | | | - Stefano Ricagno
- Department of Biomolecular Sciences and Biotechnology, Institute of Biophysics CNR and CIMAINA, University of Milano, Milan, Italy
| | | | | | - Margherita Pezzullo
- Department of Biomolecular Sciences and Biotechnology, Institute of Biophysics CNR and CIMAINA, University of Milano, Milan, Italy
| | - Vincenzo Martorana
- Institute of Biophysics, National Research Council of Italy, Palermo, Italy
| | - Antonio Cupane
- Department of Physics, University of Palermo, Palermo, Italy
| | - Martino Bolognesi
- Department of Biomolecular Sciences and Biotechnology, Institute of Biophysics CNR and CIMAINA, University of Milano, Milan, Italy
| | - Mauro Manno
- Institute of Biophysics, National Research Council of Italy, Palermo, Italy
| |
Collapse
|
26
|
Sarkar A, Zhou C, Meklemburg R, Wintrode P. Local conformational flexibility provides a basis for facile polymer formation in human neuroserpin. Biophys J 2011; 101:1758-65. [PMID: 21961602 PMCID: PMC3183757 DOI: 10.1016/j.bpj.2011.08.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 08/23/2011] [Accepted: 08/25/2011] [Indexed: 10/17/2022] Open
Abstract
Neuroserpin is a regulator of neuronal growth and plasticity. Like other members of the serpin family, neuroserpin undergoes a large conformational change as part of its function. Unlike other serpins such as α(1)-antitrypsin, wild-type neuroserpin will polymerize under near-physiological conditions, and will spontaneously transition to the latent state. To probe the origins of this conformational lability, we have performed hydrogen exchange measurements and molecular-dynamics simulations on human neuroserpin. Hydrogen exchange indicates that neuroserpin has greater flexibility in the breach region and in β-strand 1C compared with α(1)-antitrypsin. Molecular-dynamics simulations show that the distance between the top of β-strands 3 and 5A averages 4.6 Å but becomes as large as 7.5 Å in neuroserpin while it remains stable at ∼3.5 Å in α(1)-antitrypsin. Further simulations show that the stabilizing S340A mutation suppresses these fluctuations in neuroserpin. The first principal component calculated from the simulations shows a movement of helix F away from the face of β-sheet A in neuroserpin while no such movement is evident in α(1)-antitrypsin. The increased mobility of these regions in neuroserpin relative to α(1)-antitrypsin provides a basis for neuroserpin's increased tendency toward the formation of polymers and/or the latent state.
Collapse
Affiliation(s)
| | | | | | - Patrick L. Wintrode
- Department of Physiology & Biophysics, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
27
|
Almonte AG, Sweatt JD. Serine proteases, serine protease inhibitors, and protease-activated receptors: roles in synaptic function and behavior. Brain Res 2011; 1407:107-22. [PMID: 21782155 DOI: 10.1016/j.brainres.2011.06.042] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 06/03/2011] [Accepted: 06/16/2011] [Indexed: 12/11/2022]
Abstract
Serine proteases, serine protease inhibitors, and protease-activated receptors have been intensively investigated in the periphery and their roles in a wide range of processes-coagulation, inflammation, and digestion, for example-have been well characterized (see Coughlin, 2000; Macfarlane et al., 2001; Molinari et al., 2003; Wang et al., 2008; Di Cera, 2009 for reviews). A growing number of studies demonstrate that these protein systems are widely expressed in many cell types and regions in mammalian brains. Accumulating lines of evidence suggest that the brain has co-opted the activities of these interesting proteins to regulate various processes underlying synaptic activity and behavior. In this review, we discuss emerging roles for serine proteases in the regulation of mechanisms underlying synaptic plasticity and memory formation.
Collapse
Affiliation(s)
- Antoine G Almonte
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
28
|
Olson ST, Gettins PGW. Regulation of proteases by protein inhibitors of the serpin superfamily. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 99:185-240. [PMID: 21238937 DOI: 10.1016/b978-0-12-385504-6.00005-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The serpins comprise an ancient superfamily of proteins, found abundantly in eukaryotes and even in some bacteria and archea, that have evolved to regulate proteases of both serine and cysteine mechanistic classes. Unlike the thermodynamically determined lock-and-key type inhibitors, such as those of the Kunitz and Kazal families, serpins use conformational change and consequent kinetic trapping of an enzyme intermediate to effect inhibition. By combining interactions of both an exposed reactive center loop and exosites outside this loop with the active site and complementary exosites on the target protease, serpins can achieve remarkable specificity. Together with the frequent use of regulatory cofactors, this permits a sophisticated time- and location-dependent mode of protease regulation. An understanding of the structure and function of serpins has suggested that they may provide novel scaffolds for engineering protease inhibitors of desired specificity for therapeutic use.
Collapse
Affiliation(s)
- Steven T Olson
- Center for Molecular Biology of Oral Diseases, University of Illinois at Chicago, Chicago, Illinois, USA
| | | |
Collapse
|
29
|
Belorgey D, Irving JA, Ekeowa UI, Freeke J, Roussel BD, Miranda E, Pérez J, Robinson CV, Marciniak SJ, Crowther DC, Michel CH, Lomas DA. Characterisation of serpin polymers in vitro and in vivo. Methods 2010; 53:255-66. [PMID: 21115126 DOI: 10.1016/j.ymeth.2010.11.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 11/21/2010] [Indexed: 10/18/2022] Open
Abstract
Neuroserpin is a member of the serine protease inhibitor or serpin superfamily of proteins. It is secreted by neurones and plays an important role in the regulation of tissue plasminogen activator at the synapse. Point mutations in the neuroserpin gene cause the autosomal dominant dementia familial encephalopathy with neuroserpin inclusion bodies or FENIB. This is one of a group of disorders caused by mutations in the serpins that are collectively known as the serpinopathies. Others include α(1)-antitrypsin deficiency and deficiency of C1 inhibitor, antithrombin and α(1)-antichymotrypsin. The serpinopathies are characterised by delays in protein folding and the retention of ordered polymers of the mutant serpin within the cell of synthesis. The clinical phenotype results from either a toxic gain of function from the inclusions or a loss of function, as there is insufficient protease inhibitor to regulate important proteolytic cascades. We describe here the methods required to characterise the polymerisation of neuroserpin and draw parallels with the polymerisation of α(1)-antitrypsin. It is important to recognise that the conditions in which experiments are performed will have a major effect on the findings. For example, incubation of monomeric serpins with guanidine or urea will produce polymers that are not found in vivo. The characterisation of the pathological polymers requires heating of the folded protein or alternatively the assessment of ordered polymers from cell and animal models of disease or from the tissues of humans who carry the mutation.
Collapse
Affiliation(s)
- Didier Belorgey
- Dept. of Medicine, University of Cambridge, Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Ricagno S, Pezzullo M, Barbiroli A, Manno M, Levantino M, Santangelo MG, Bonomi F, Bolognesi M. Two latent and two hyperstable polymeric forms of human neuroserpin. Biophys J 2010; 99:3402-11. [PMID: 21081089 PMCID: PMC2980742 DOI: 10.1016/j.bpj.2010.09.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 09/08/2010] [Accepted: 09/09/2010] [Indexed: 02/02/2023] Open
Abstract
Human neuroserpin (hNS) is a serine protease inhibitor that belongs to the serpin superfamily and is expressed in nervous tissues. The serpin fold is generally characterized by a long exposed loop, termed the reactive center loop, that acts as bait for the target protease. Intramolecular insertion of the reactive center loop into the main serpin β-sheet leads to the serpin latent form. As with other known serpins, hNS pathological mutants have been shown to accumulate as polymers composed of quasi-native protein molecules. Although hNS polymerization has been intensely studied, a general agreement about serpin polymer organization is still lacking. Here we report a biophysical characterization of native hNS that is shown to undergo two distinct conformational transitions, at 55°C and 85°C, both leading to distinct latent and polymeric species. The latent and polymer hNS forms obtained at 45°C and 85°C differ in their chemical and thermal stabilities; furthermore, the hNS polymers also differ in size and morphology. Finally, the 85°C polymer shows a higher content of intermolecular β-sheet interactions than the 45°C polymer. Together, these results suggest a more complex conformational scenario than was previously envisioned, and, in a general context, may help reconcile the current contrasting views on serpin polymerization.
Collapse
Affiliation(s)
- Stefano Ricagno
- Dipartimento di Scienze Biomolecolari e Biotecnologie, Centro Interdisciplinare Materiali e Interfacce Nanostrutturati, Università di Milano, Milan, Italy
- Dipartimento di Biochimica, Università di Pavia, Pavia, Italy
- Laboratori di Biotecnologie, Istituto Di Ricovero e Cura a Carattere Scientifico Fondazione Policlinico San Matteo, Pavia, Italy
| | - Margherita Pezzullo
- Dipartimento di Scienze Biomolecolari e Biotecnologie, Centro Interdisciplinare Materiali e Interfacce Nanostrutturati, Università di Milano, Milan, Italy
| | - Alberto Barbiroli
- Sezione di Biochimica, Dipartimento di Scienze Molecolari Agroalimentari, Università di Milano, Milan, Italy
| | - Mauro Manno
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, CNR, Palermo, Italy
| | - Matteo Levantino
- Dipartimento di Scienze Fisiche ed Astronomiche, Università of Palermo, Palermo, Italy
| | | | - Francesco Bonomi
- Sezione di Biochimica, Dipartimento di Scienze Molecolari Agroalimentari, Università di Milano, Milan, Italy
| | - Martino Bolognesi
- Dipartimento di Scienze Biomolecolari e Biotecnologie, Centro Interdisciplinare Materiali e Interfacce Nanostrutturati, Università di Milano, Milan, Italy
| |
Collapse
|
31
|
Belorgey D, Hägglöf P, Onda M, Lomas DA. pH-dependent stability of neuroserpin is mediated by histidines 119 and 138; implications for the control of beta-sheet A and polymerization. Protein Sci 2010; 19:220-8. [PMID: 19953505 PMCID: PMC2865726 DOI: 10.1002/pro.299] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Revised: 09/24/2009] [Accepted: 11/16/2009] [Indexed: 11/09/2022]
Abstract
Neuroserpin is a member of the serpin superfamily. Point mutations in the neuroserpin gene underlie the autosomal dominant dementia, familial encephalopathy with neuroserpin inclusion bodies. This is characterized by the retention of ordered polymers of neuroserpin within the endoplasmic reticulum of neurons. pH has been shown to affect the propensity of several serpins to form polymers. In particular, low pH favors the formation of polymers of both alpha(1)-antitrypsin and antithrombin. We report here opposite effects in neuroserpin, with a striking resistance to polymer formation at acidic pH. Mutation of specific histidine residues showed that this effect is not attributable to the shutter domain histidine as would be predicted by analogy with other serpins. Indeed, mutation of the shutter domain His338 decreased neuroserpin stability but had no effect on the pH dependence of polymerization when compared with the wild-type protein. In contrast, mutation of His119 or His138 reduced the polymerization of neuroserpin at both acidic and neutral pH. These residues are at the lower pole of neuroserpin and provide a novel mechanism to control the opening of beta-sheet A and hence polymerization. This mechanism is likely to have evolved to protect neuroserpin from the acidic environment of the secretory granules.
Collapse
Affiliation(s)
- Didier Belorgey
- Department of Medicine, University of Cambridge, Cambridge Institute for Medical Research, Cambridge CB2 0XY, United Kingdom.
| | | | | | | |
Collapse
|
32
|
Synaptic plasticity-associated proteases and protease inhibitors in the brain linked to the processing of extracellular matrix and cell adhesion molecules. ACTA ACUST UNITED AC 2009; 4:223-34. [DOI: 10.1017/s1740925x09990172] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Research on the molecular and cellular basis of learning and memory has focused on the mechanisms that underlie the induction and expression of synaptic plasticity. There is increasing evidence that structural changes at the synapse are associated with synaptic plasticity and that extracellular matrix (ECM) components and cell adhesion molecules are associated with these changes. The functions of both groups of molecules can be regulated by proteolysis. In this article we review the roles of selected proteases and protease inhibitors in perisynaptic proteolysis of the ECM and synaptic adhesion proteins and the impact of proteolysis on synaptic modification and cognitive function.
Collapse
|
33
|
Takehara S, Onda M, Zhang J, Nishiyama M, Yang X, Mikami B, Lomas DA. The 2.1-A crystal structure of native neuroserpin reveals unique structural elements that contribute to conformational instability. J Mol Biol 2009; 388:11-20. [PMID: 19285087 DOI: 10.1016/j.jmb.2009.03.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Revised: 03/03/2009] [Accepted: 03/04/2009] [Indexed: 11/23/2022]
Abstract
Neuroserpin is a selective inhibitor of tissue-type plasminogen activator (tPA) that plays an important role in neuronal plasticity, memory, and learning. We report here the crystal structure of native human neuroserpin at 2.1 A resolution. The structure has a helical reactive center loop and an omega loop between strands 1B and 2B. The omega loop contributes to the inhibition of tPA, as deletion of this motif reduced the association rate constant with tPA by threefold but had no effect on the kinetics of interaction with urokinase. Point mutations in neuroserpin cause the formation of ordered intracellular polymers that underlie dementia familial encephalopathy with neuroserpin inclusion bodies (FENIB). Wild-type neuroserpin is also unstable and readily forms polymers under near-physiological conditions in vitro. This is, in part, due to the substitution of a conserved alanine for serine at position 340. The replacement of Ser340 by Ala increased the melting temperature by 3 degrees C and reduced polymerization as compared to wild-type neuroserpin. Similarly, neuroserpin has Asn-Leu-Val at the end of helix F and thus differs markedly from the Gly-X-Ile consensus sequence of the serpins. Restoration of these amino acids to the consensus sequence increased thermal stability and reduced the polymerization of neuroserpin and its transition to the latent conformer. Moreover, introduction of the consensus sequence into S49P neuroserpin that causes FENIB increased the stability and inhibitory activity of the mutant, as well as blocked polymerization and increased the yield of protein during refolding. These data provide a molecular explanation for the inherent instability of neuroserpin and the effect of point mutations that underlie the dementia FENIB.
Collapse
Affiliation(s)
- Sayaka Takehara
- Division of Applied Life Sciences, The Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Ricagno S, Caccia S, Sorrentino G, Antonini G, Bolognesi M. Human neuroserpin: structure and time-dependent inhibition. J Mol Biol 2009; 388:109-21. [PMID: 19265707 DOI: 10.1016/j.jmb.2009.02.056] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 02/19/2009] [Accepted: 02/24/2009] [Indexed: 11/23/2022]
Abstract
Human neuroserpin (hNS) is a protein serine protease inhibitor expressed mainly in the nervous system, where it plays key roles in neural development and plasticity by primarily targeting tissue plasminogen activator (tPA). Four hNS mutations are associated to a form of autosomal dominant dementia, known as familial encephalopathy with neuroserpin inclusion bodies. The medical interest in and the lack of structural information on hNS prompted us to study the crystal structure of native and cleaved hNS, reported here at 3.15 and 1.85 A resolution, respectively. In the light of the three-dimensional structures, we focus on the hNS reactive centre loop in its intact and cleaved conformations relative to the current serpin polymerization models and discuss the protein sites hosting neurodegenerative mutations. On the basis of homologous serpin structures, we suggest the location of a protein surface site that may stabilize the hNS native (metastable) form. In parallel, we present the results of kinetic studies on hNS inhibition of tPA. Our data analysis stresses the instability of the hNS-tPA complex with a dissociation half-life of minutes compared to a half-life of weeks observed for other serpin-cognate protease complexes.
Collapse
Affiliation(s)
- Stefano Ricagno
- Department of Biomolecular Sciences and Biotechnology, CNR-INFM and CIMAINA, University of Milano, Via Celoria 26, 20133 Milan, Italy
| | | | | | | | | |
Collapse
|
35
|
Fabbro S, Seeds NW. Plasminogen activator activity is inhibited while neuroserpin is up-regulated in the Alzheimer disease brain. J Neurochem 2009; 109:303-15. [PMID: 19222708 DOI: 10.1111/j.1471-4159.2009.05894.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Amyloid-beta plaques are a pathological hallmark of Alzheimer's disease. Several proteases are known to cleave/remove amyloid-beta, including plasmin, the product of tissue plasminogen activator cleavage of the pro-enzyme plasminogen. Although plasmin levels are lower in Alzheimer brain, there has been little analysis of the plasminogen activator/plasmin system in the brains of Alzheimer patients. In this study, zymography, immunocapture, and ELISAs were utilized to show that tissue plasminogen activator activity in frontal cortex tissue of Alzheimer patients is dramatically reduced compared with age-matched controls, while tissue plasminogen activator and plasminogen protein levels are unchanged; suggesting that plasminogen activator activity is inhibited in the Alzheimer brain. Analysis of endogenous plasminogen activator inhibitors shows that while plasminogen activator inhibitor-1 and protease nexin-1 levels are unchanged, the neuroserpin levels are significantly elevated in brains of Alzheimer patients. Furthermore, elevated amounts of tissue plasminogen activator-neuroserpin complexes are seen in the Alzheimer brain, and immunohistochemical studies demonstrate that both tissue plasminogen activator and neuroserpin are associated with amyloid-beta plaques in Alzheimer brain tissue. Thus, neuroserpin inhibition of tissue plasminogen activator activity leads to reduced plasmin and may be responsible for reduced clearance of amyloid-beta in the Alzheimer disease brain. Furthermore, decreased tissue plasminogen activator activity in the Alzheimer brain may directly influence synaptic activity and impair cognitive function.
Collapse
Affiliation(s)
- Shay Fabbro
- Neuroscience Program and Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, USA
| | | |
Collapse
|
36
|
Huang X, Swanson R, Broze GJ, Olson ST. Kinetic characterization of the protein Z-dependent protease inhibitor reaction with blood coagulation factor Xa. J Biol Chem 2008; 283:29770-83. [PMID: 18768472 PMCID: PMC2573075 DOI: 10.1074/jbc.m805214200] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Revised: 09/03/2008] [Indexed: 11/06/2022] Open
Abstract
Protein Z-dependent protease inhibitor (ZPI) is a recently identified member of the serpin superfamily that functions as a cofactor-dependent regulator of blood coagulation factors Xa (FXa) and XIa. Here we show that ZPI and its cofactor, protein Z (PZ), inhibit procoagulant membrane-bound factor Xa by the branched pathway acyl-intermediate trapping mechanism used by other serpins, but with significant variations of this mechanism that are unique to ZPI. Rapid kinetic analyses showed that the reaction proceeded by the initial assembly of a membrane-associated PZ-ZPI-FXa Michaelis complex (K(M) 53+/-5 nM) followed by conversion to a stable ZPI-FXa complex (k(lim) 1.2+/-0.1 s(-1)). Cofactor premixing experiments together with independent kinetic analyses of ZPI-PZ and factor Xa-PZ-membrane complex formation suggested that assembly of the Michaelis complex through either ZPI-PZ-lipid or factor Xa-PZ-lipid intermediates was rate-limiting. Reaction stoichiometry analyses and native PAGE showed that for every factor Xa molecule inhibited by ZPI, two serpin molecules were cleaved. Native PAGE and immunoblotting showed that PZ dissociated from ZPI once ZPI forms a stable complex with FXa, and kinetic analyses confirmed that PZ acted catalytically to accelerate the membrane-dependent ZPI-factor Xa reaction. The ZPI-FXa complex was only transiently stable and dissociated with a rate constant that showed a bell-shaped pH dependence indicative of participation of factor Xa active-site residues. The complex was detectable by SDS-PAGE when denatured at low pH, consistent with it being a kinetically trapped covalent acyl-intermediate. Together our findings show that ZPI functions like other serpins to regulate the activity of FXa but in a manner uniquely dependent on protein Z, procoagulant membranes, and pH.
Collapse
Affiliation(s)
- Xin Huang
- Center for Molecular Biology of Oral Diseases, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | | | |
Collapse
|
37
|
Miranda E, MacLeod I, Davies MJ, Pérez J, Römisch K, Crowther DC, Lomas DA. The intracellular accumulation of polymeric neuroserpin explains the severity of the dementia FENIB. Hum Mol Genet 2008; 17:1527-39. [PMID: 18267959 PMCID: PMC2387220 DOI: 10.1093/hmg/ddn041] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2007] [Accepted: 02/07/2008] [Indexed: 11/13/2022] Open
Abstract
Familial encephalopathy with neuroserpin inclusion bodies (FENIB) is an autosomal dominant dementia that is characterized by the retention of polymers of neuroserpin as inclusions within the endoplasmic reticulum (ER) of neurons. We have developed monoclonal antibodies that detect polymerized neuroserpin and have used COS-7 cells, stably transfected PC12 cell lines and transgenic Drosophila melanogaster to characterize the cellular handling of all four mutant forms of neuroserpin that cause FENIB. We show a direct correlation between the severity of the disease-causing mutation and the accumulation of neuroserpin polymers in cell and fly models of the disease. Moreover, mutant neuroserpin causes locomotor deficits in the fly allowing us to demonstrate a direct link between polymer accumulation and neuronal toxicity.
Collapse
Affiliation(s)
- Elena Miranda
- Department of Medicine, University of Cambridge, Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, UK.
| | | | | | | | | | | | | |
Collapse
|
38
|
Li SH, Gorlatova NV, Lawrence DA, Schwartz BS. Structural differences between active forms of plasminogen activator inhibitor type 1 revealed by conformationally sensitive ligands. J Biol Chem 2008; 283:18147-57. [PMID: 18436534 DOI: 10.1074/jbc.m709455200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Plasminogen activator inhibitor type 1 (PAI-1) is a serine protease inhibitor (serpin) in which the reactive center loop (RCL) spontaneously inserts into a central beta-sheet, beta-sheet A, resulting in inactive inhibitor. Available x-ray crystallographic studies of PAI-1 in an active conformation relied on the use of stabilizing mutations. Recently it has become evident that these structural models do not adequately explain the behavior of wild-type PAI-1 (wtPAI-1) in solution. To probe the structure of native wtPAI-1, we used three conformationally sensitive ligands: the physiologic cofactor, vitronectin; a monoclonal antibody, 33B8, that binds preferentially to RCL-inserted forms of PAI-1; and RCL-mimicking peptides that insert into beta-sheet A. From patterns of interaction with wtPAI-1 and the stable mutant, 14-1B, we propose a model of the native conformation of wtPAI-1 in which the bottom of the central sheet is closed, whereas the top of the beta-sheet A is open to allow partial insertion of the RCL. Because the incorporation of RCL-mimicking peptides into wtPAI-1 is accelerated by vitronectin, we further propose that vitronectin alters the conformation of the RCL to allow increased accessibility to beta-sheet A, yielding a structural hypothesis that is contradictory to the current structural model of PAI-1 in solution and its interaction with vitronectin.
Collapse
Affiliation(s)
- Shih-Hon Li
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA
| | | | | | | |
Collapse
|
39
|
Paterson M, Horvath A, Pike R, Coughlin P. Molecular characterization of centerin, a germinal centre cell serpin. Biochem J 2007; 405:489-94. [PMID: 17447896 PMCID: PMC2267310 DOI: 10.1042/bj20070174] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Centerin [SERPINA9/GCET1 (germinal centre B-cell-expressed transcript 1)] is a serpin (serine protease inhibitor) whose expression is restricted to germinal centre B-cells and lymphoid malignancies with germinal centre B-cell maturation. Expression of centerin, together with bcl-6 and GCET2, constitutes a germinal centre B-cell signature, which is associated with a good prognosis in diffuse large B-cell lymphomas, but the molecular basis for this remains to be elucidated. We report here the cloning, expression and molecular characterization of bacterial recombinant centerin. Biophysical studies demonstrated that centerin was able to undergo the 'stressed to relaxed' conformational change which is an absolute requirement for protease inhibitory activity. Kinetic analysis showed that centerin rapidly inhibited the serine protease trypsin (k(a)=1.9x10(5) M(-1) x s(-1)) and also demonstrated measurable inhibition of thrombin (k(a)=1.17x10(3) M(-1) x s(-1)) and plasmin (k(a)=1.92x10(3) M(-1) x s(-1)). Centerin also bound DNA and unfractionated heparin, although there was no functionally significant impact on the rate of inhibition. These results suggest that centerin is likely to function in vivo in the germinal centre as an efficient inhibitor of a trypsin-like protease.
Collapse
Affiliation(s)
- Melinda A. Paterson
- *Australian Centre for Blood Diseases, Monash University, Level 6 Burnet Tower, 89 Commercial Road, Melbourne 3004, VIC, Australia
| | - Anita J. Horvath
- *Australian Centre for Blood Diseases, Monash University, Level 6 Burnet Tower, 89 Commercial Road, Melbourne 3004, VIC, Australia
| | - Robert N. Pike
- †Department of Biochemistry and Molecular Biology, Monash University, Wellington Road, Clayton 3800, VIC, Australia
| | - Paul B. Coughlin
- *Australian Centre for Blood Diseases, Monash University, Level 6 Burnet Tower, 89 Commercial Road, Melbourne 3004, VIC, Australia
- To whom correspondence should be addressed (email )
| |
Collapse
|
40
|
Sharp LK, Mallya M, Kinghorn KJ, Wang Z, Crowther DC, Huntington JA, Belorgey D, Lomas DA. Sugar and alcohol molecules provide a therapeutic strategy for the serpinopathies that cause dementia and cirrhosis. FEBS J 2006; 273:2540-52. [PMID: 16704419 DOI: 10.1111/j.1742-4658.2006.05262.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Mutations in neuroserpin and alpha1-antitrypsin cause these proteins to form ordered polymers that are retained within the endoplasmic reticulum of neurones and hepatocytes, respectively. The resulting inclusions underlie the dementia familial encephalopathy with neuroserpin inclusion bodies (FENIB) and Z alpha1-antitrypsin-associated cirrhosis. Polymers form by a sequential linkage between the reactive centre loop of one molecule and beta-sheet A of another, and strategies that block polymer formation are likely to be successful in treating the associated disease. We show here that glycerol, the sugar alcohol erythritol, the disaccharide trehalose and its breakdown product glucose reduce the rate of polymerization of wild-type neuroserpin and the Ser49Pro mutant that causes dementia. They also attenuate the polymerization of the Z variant of alpha1-antitrypsin. The effect on polymerization was apparent even when these agents had been removed from the buffer. None of these agents had any detectable effect on the structure or inhibitory activity of neuroserpin or alpha1-antitrypsin. These data demonstrate that sugar and alcohol molecules can reduce the polymerization of serpin mutants that cause disease, possibly by binding to and stabilizing beta-sheet A.
Collapse
|
41
|
Bruno MA, Cuello AC. Activity-dependent release of precursor nerve growth factor, conversion to mature nerve growth factor, and its degradation by a protease cascade. Proc Natl Acad Sci U S A 2006; 103:6735-40. [PMID: 16618925 PMCID: PMC1458950 DOI: 10.1073/pnas.0510645103] [Citation(s) in RCA: 268] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this report, we provide direct demonstration that the neurotrophin nerve growth factor (NGF) is released in the extracellular space in an activity-dependent manner in its precursor form (proNGF) and that it is in this compartment that its maturation and degradation takes place because of the coordinated release and the action of proenzymes and enzyme regulators. This converting protease cascade and its endogenous regulators (including tissue plasminogen activator, plasminogen, neuroserpin, precursor matrix metalloproteinase 9, and tissue inhibitor metalloproteinase 1) are colocalized in neurons of the cerebral cortex and released upon neuronal stimulation. We also provide evidence that this mechanism operates in in vivo conditions, as the CNS application of inhibitors of converting and degrading enzymes lead to dramatic alterations in the tissue levels of either precursor NGF or mature NGF. Pathological alterations of this cascade in the CNS might cause or contribute to a lack of proper neuronal trophic support in conditions such as cerebral ischemia, seizure and Alzheimer's disease or, conversely, to excessive local production of neurotrophins as reported in inflammatory arthritis pain.
Collapse
Affiliation(s)
| | - A. Claudio Cuello
- Departments of *Pharmacology and Therapeutics
- Anatomy and Cell Biology, and
- Neurology and Neurosurgery, McGill University, Montreal, QC, Canada H3G 1Y6
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
42
|
Lebeurrier N, Vivien D, Ali C. The complexity of tissue-type plasminogen activator: can serine protease inhibitors help in stroke management? Expert Opin Ther Targets 2006; 8:309-20. [PMID: 15268626 DOI: 10.1517/14728222.8.4.309] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Stroke, the third leading cause of death in industrialised countries, represents a major burden on healthcare authorities. The elucidation of molecular events sustaining infarct evolution in experimental models has allowed the development of putative therapeutic agents. However, despite marked benefits in animals, most of them have failed in clinical trials. At present, the only approved therapy for stroke is early reperfusion by intravenous injection of the thrombolytic agent, tissue-type plasminogen activator (tPA). tPA-dependent thrombolysis sometimes promotes haemorrhage, but improves neurological outcome in a great proportion of patients, provided it is performed within the recommended therapeutic window. In addition to the benefit of tPA injection in the vascular compartment, this endogenously produced serine protease could also promote excitotoxic processes within the cerebral parenchyma. This article reviews the various aspects of tPA during stroke, and discusses potential improvements to current clinical management, with a particular emphasis on targeting the deleterious actions of tPA through endogenous serine protease inhibitors (serpins).
Collapse
|
43
|
de Groot DM, Pol C, Martens GJM. Comparative analysis and expression of neuroserpin in Xenopus laevis. Neuroendocrinology 2005; 82:11-20. [PMID: 16319501 DOI: 10.1159/000090011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2005] [Accepted: 10/06/2005] [Indexed: 01/22/2023]
Abstract
Serine protease inhibitors form a diverse family of proteins of which most members inhibit target serine proteases. Neuroserpin is a member of this family. Here, we have characterized neuroserpin in the nonmammalian species Xenopus laevis and found a high degree of aminoacid sequence conservation, especially of the reactive center loop, of the Xenopus protein compared to mammalian and chicken neuroserpin sequences, suggesting a conserved target specificity. Neuroserpin mRNA and protein were expressed throughout Xenopus development, while in the adult frog high mRNA expression was found in neuronal and neuroendocrine tissues, and the reproductive organs, and the neuroserpin protein was detected mainly in brain and pituitary. More specifically, in Xenopus pituitary neuroserpin mRNA was expressed higher in the neurointermediate lobe than in the pars distalis. At the protein level, we detected a 55-kDa neuroserpin protein in the pars nervosa, two neuroserpin proteins of 44- and 50-kDa in the melanotrope cells of the pars intermedia, and a 46-kDa product in the pars distalis. On the basis of its relatively high degree of sequence conservation and its expression pattern, we conclude that Xenopus neuroserpin may play an important physiological role, e.g. as a serine protease inhibitor during development, and for proper neuronal and neuroendocrine cell functioning.
Collapse
Affiliation(s)
- Dorien M de Groot
- Department of Molecular Animal Physiology, Institute for Neuroscience, Nijmegen Center for Molecular Life Sciences, Radboud University, Nijmegen, The Netherlands
| | | | | |
Collapse
|
44
|
Onda M, Belorgey D, Sharp LK, Lomas DA. Latent S49P neuroserpin forms polymers in the dementia familial encephalopathy with neuroserpin inclusion bodies. J Biol Chem 2005; 280:13735-41. [PMID: 15664988 DOI: 10.1074/jbc.m413282200] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The serpinopathies result from conformational transitions in members of the serine proteinase inhibitor superfamily with aberrant tissue deposition or loss of function. They are typified by mutants of neuroserpin that are retained within the endoplasmic reticulum of neurons as ordered polymers in association with dementia. We show here that the S49P mutant of neuroserpin that causes the dementia familial encephalopathy with neuroserpin inclusion bodies (FENIB) forms a latent species in vitro and in vivo in addition to the formation of polymers. Latent neuroserpin is thermostable and inactive as a proteinase inhibitor, but activity can be restored by refolding. Strikingly, latent S49P neuroserpin is unlike any other latent serine proteinase inhibitor (serpin) in that it spontaneously forms polymers under physiological conditions. These data provide an alternative method for the inactivation of mutant neuroserpin as a proteinase inhibitor in FENIB and demonstrate a second pathway for the formation of intracellular polymers in association with disease.
Collapse
Affiliation(s)
- Maki Onda
- Department of Medicine, University of Cambridge, Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 2XY, United Kingdom.
| | | | | | | |
Collapse
|
45
|
Belorgey D, Sharp LK, Crowther DC, Onda M, Johansson J, Lomas DA. Neuroserpin Portland (Ser52Arg) is trapped as an inactive intermediate that rapidly forms polymers: implications for the epilepsy seen in the dementia FENIB. EUROPEAN JOURNAL OF BIOCHEMISTRY 2004; 271:3360-7. [PMID: 15291813 DOI: 10.1111/j.1432-1033.2004.04270.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The dementia familial encephalopathy with neuroserpin inclusion bodies (FENIB) is caused by point mutations in the neuroserpin gene. We have shown a correlation between the predicted effect of the mutation and the number of intracerebral inclusions, and an inverse relationship with the age of onset of disease. Our previous work has shown that the intraneuronal inclusions in FENIB result from the sequential interaction between the reactive centre loop of one neuroserpin molecule with beta-sheet A of the next. We show here that neuroserpin Portland (Ser52Arg), which causes a severe form of FENIB, also forms loop-sheet polymers but at a faster rate, in keeping with the more severe clinical phenotype. The Portland mutant has a normal unfolding transition in urea and a normal melting temperature but is inactive as a proteinase inhibitor. This results in part from the reactive loop being in a less accessible conformation to bind to the target enzyme, tissue plasminogen activator. These results, with those of the CD analysis, are in keeping with the reactive centre loop of neuroserpin Portland being partially inserted into beta-sheet A to adopt a conformation similar to an intermediate on the polymerization pathway. Our data provide an explanation for the number of inclusions and the severity of dementia in FENIB associated with neuroserpin Portland. Moreover the inactivity of the mutant may result in uncontrolled activity of tissue plasminogen activator, and so explain the epileptic seizures seen in individuals with more severe forms of the disease.
Collapse
Affiliation(s)
- Didier Belorgey
- Cambridge Institute for Medical Research, Department of Medicine, University of Cambridge, UK.
| | | | | | | | | | | |
Collapse
|
46
|
Miranda E, Römisch K, Lomas DA. Mutants of neuroserpin that cause dementia accumulate as polymers within the endoplasmic reticulum. J Biol Chem 2004; 279:28283-91. [PMID: 15090543 DOI: 10.1074/jbc.m313166200] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The dementia familial encephalopathy with neuroserpin inclusion bodies (FENIB) is caused by the accumulation of mutant neuroserpin within neurons (Davis, R. L., Shrimpton, A. E., Holohan, P. D., Bradshaw, C., Feiglin, D., Sonderegger, P., Kinter, J., Becker, L. M., Lacbawan, F., Krasnewich, D., Muenke, M., Lawrence, D. A., Yerby, M. S., Shaw, C.-M., Gooptu, B., Elliott, P. R., Finch, J. T., Carrell, R. W., and Lomas, D. A. (1999) Nature 401, 376-379), but little is known about the trafficking of wild type and mutant neuroserpins. We have established a cell model to study the processing of wild type neuroserpin and the Syracuse (S49P) and Portland (S52R) mutants that cause FENIB. Here we show that Syracuse and Portland neuroserpin are retained soon after their synthesis in the endoplasmic reticulum and that the limiting step in their processing is the transport to the Golgi complex. This is in contrast to the wild type protein, which is secreted into the culture medium. Mutant neuroserpin is retained within the endoplasmic reticulum as polymers, similar to those isolated from the intraneuronal inclusions in the brains of individuals with FENIB. Remarkably, the Portland mutant showed faster accumulation and slower secretion compared with the Syracuse mutant, in keeping with the more severe clinical phenotype found in patients with the Portland variant of neuroserpin. Both mutant and wild type neuroserpin were partially degraded by proteasomes. Taken together, our results provide further understanding of how cells handle defective but ordered mutant proteins and provide strong support for a common mechanism of disease caused by mutants of the serine protease inhibitor superfamily.
Collapse
Affiliation(s)
- Elena Miranda
- Departments of Medicine and Clinical Biochemistry, University of Cambridge, Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council Building, Hills Road, Cambridge CB2 2XY, United Kingdom.
| | | | | |
Collapse
|
47
|
Navarro-Yubero C, Cuadrado A, Sonderegger P, Muñoz A. Neuroserpin is post-transcriptionally regulated by thyroid hormone. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2004; 123:56-65. [PMID: 15046866 DOI: 10.1016/j.molbrainres.2003.12.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/31/2003] [Indexed: 12/17/2022]
Abstract
Neuroserpin is a serine protease inhibitor expressed in the developing and the adult nervous system. Studies with genetically modified mice indicate a role of neuroserpin in the regulation of anxiety. Mutations in the neuroserpin gene cause protein polymerization and formation of inclusion bodies leading to progressive myoclonic epilepsy and neurodegeneration. Here we demonstrate that neuroserpin expression is regulated by thyroid hormone (T3). Neuroserpin RNA levels are down-regulated in cortical layers II/III and VIa, the hippocampus, the retrosplenial cortex and the medial habenular nucleus, but not in cortical layer V or other areas of the hypothyroid rat brain. Concordantly, neuroserpin RNA and protein expression was induced by T3 in rat PC12 cells containing appropriate thyroid hormone receptor levels. In run-on assays T3 did not affect the transcription rate of the neuroserpin gene, indicating that regulation was post-transcriptional. Moreover, T3 increased in vitro binding of cytoplasmic proteins to neuroserpin 3'-UTR RNA and caused biphasic regulation of the stability of this transcript in PC12 cells. Ectopic neuroserpin expression induced neurite extension in PC12 cells and enhanced neuritogenesis triggered by nerve growth factor. In summary, these results indicate that neuroserpin expression is post-transcriptionally regulated by T3 at the level of RNA stability.
Collapse
Affiliation(s)
- Cristina Navarro-Yubero
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, E-28029 Madrid, Spain
| | | | | | | |
Collapse
|
48
|
Makarova A, Mikhailenko I, Bugge TH, List K, Lawrence DA, Strickland DK. The low density lipoprotein receptor-related protein modulates protease activity in the brain by mediating the cellular internalization of both neuroserpin and neuroserpin-tissue-type plasminogen activator complexes. J Biol Chem 2003; 278:50250-8. [PMID: 14522960 DOI: 10.1074/jbc.m309150200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Proteases contribute to a variety of processes in the brain; consequently, their activity is carefully regulated by protease inhibitors, such as neuroserpin. This inhibitor is thought to be secreted by axons at synaptic regions where it controls tissue-type plasminogen activator (tPA) activity. Mechanisms regulating neuroserpin are not known, and the current studies were undertaken to define the cellular pathways involved in neuroserpin catabolism. We found that both active neuroserpin and neuroserpin.tPA complexes were internalized by mouse cortical cultures and embryonic fibroblasts in a process mediated by the low density lipoprotein receptor-related protein (LRP). Surprisingly, despite the fact that active neuroserpin is internalized by LRP, this form of the molecule does not directly bind to LRP on its own, indicating the requirement of a cofactor for neuroserpin internalization. Our studies ruled out the possibility that endogenously produced plasminogen activators (i.e. tPA and urokinase-type plasminogen activator) are responsible for the LRP-mediated internalization of active neuroserpin, but could not rule out the possibility that another cell-associated proteases capable of binding active neuroserpin functions in this capacity. In summary, neuroserpin levels appear to be carefully regulated by LRP and an unidentified cofactor, and this pathway may be critical for maintaining the balance between proteases and inhibitors.
Collapse
Affiliation(s)
- Alexandra Makarova
- Department of Vascular Biology, Holland Laboratory, American Red Cross, Rockville, Maryland 20855, USA
| | | | | | | | | | | |
Collapse
|
49
|
Molinari F, Meskanaite V, Munnich A, Sonderegger P, Colleaux L. Extracellular proteases and their inhibitors in genetic diseases of the central nervous system. Hum Mol Genet 2003; 12 Spec No 2:R195-200. [PMID: 12925575 DOI: 10.1093/hmg/ddg276] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cumulative evidence has shown that a delicate balance between serine proteases and their inhibitors is crucial for normal functioning of several biological pathways. The importance of proteases and their inhibitors is well documented in several human diseases. Among them, the best documented are hemophilia B, a genetic deficiency of the serine protease coagulation factor IX and serpinophathies. Alpha-1-antitrypsin deficiency (MIM 107400), is associated with early-onset emphysema and liver disease, while hereditary angioedema (HANE; MIM 106100) is caused by mutations in the C1 inhibitor, a serpin involved in the regulation of the complement cascade. Recently, two human genetic diseases of the central nervous system have been related to mutations in components of extracellular proteolytic systems. Here, we review the recent advances in this field.
Collapse
Affiliation(s)
- Florence Molinari
- Départment de Génétique et Unité de Recherche sur les Handicaps Génetiques de l'Enfant, INSERM U393, Hôpital Necker-Enfants Malades, Cedex 15, France.
| | | | | | | | | |
Collapse
|