1
|
Gallwitz L, Bleibaum F, Voss M, Schweizer M, Spengler K, Winter D, Zöphel F, Müller S, Lichtenthaler S, Damme M, Saftig P. Cellular depletion of major cathepsin proteases reveals their concerted activities for lysosomal proteolysis. Cell Mol Life Sci 2024; 81:227. [PMID: 38775843 PMCID: PMC11111660 DOI: 10.1007/s00018-024-05274-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024]
Abstract
Proteins delivered by endocytosis or autophagy to lysosomes are degraded by exo- and endoproteases. In humans 15 lysosomal cathepsins (CTS) act as important physiological regulators. The cysteine proteases CTSB and CTSL and the aspartic protease CTSD are the most abundant and functional important lysosomal proteinases. Whereas their general functions in proteolysis in the lysosome, their individual substrate, cleavage specificity, and their possible sequential action on substrate proteins have been previously studied, their functional redundancy is still poorly understood. To address a possible common role of highly expressed and functional important CTS proteases, we generated CTSB-, CTSD-, CTSL-, and CTSBDL-triple deficient (KO) human neuroblastoma-derived SH-SY5Y cells and CTSB-, CTSD-, CTSL-, CTSZ and CTSBDLZ-quadruple deficient (KO) HeLa cells. These cells with a combined cathepsin deficiency exhibited enlarged lysosomes and accumulated lipofuscin-like storage material. The lack of the three (SH-SY5Y) or four (HeLa) major CTSs caused an impaired autophagic flux and reduced degradation of endocytosed albumin. Proteome analyses of parental and CTS-depleted cells revealed an enrichment of cleaved peptides, lysosome/autophagy-associated proteins, and potentially endocytosed membrane proteins like the amyloid precursor protein (APP), which can be subject to endocytic degradation. Amino- and carboxyterminal APP fragments accumulated in the multiple CTS-deficient cells, suggesting that multiple CTS-mediated cleavage events regularly process APP. In summary, our analyses support the idea that different lysosomal cathepsins act in concert, have at least partially and functionally redundant substrates, regulate protein degradation in autophagy, and control cellular proteostasis, as exemplified by their involvement in the degradation of APP fragments.
Collapse
Affiliation(s)
- Lisa Gallwitz
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Olshausenstr. 40, 24098, Kiel, Germany
| | - Florian Bleibaum
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Olshausenstr. 40, 24098, Kiel, Germany
| | - Matthias Voss
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Olshausenstr. 40, 24098, Kiel, Germany
| | - Michaela Schweizer
- Center for Molecular Neurobiology (ZMNH), UKE, Falkenried 94, 20251, Hamburg, Germany
| | - Katharina Spengler
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Olshausenstr. 40, 24098, Kiel, Germany
| | - Dominic Winter
- Institute for Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Frederic Zöphel
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Olshausenstr. 40, 24098, Kiel, Germany
| | - Stephan Müller
- German Center for Neurodegenerative Diseases (DZNE), München, Feodor-Lynen-Str. 17, 81377, Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675, Munich, Germany
| | - Stefan Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), München, Feodor-Lynen-Str. 17, 81377, Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Markus Damme
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Olshausenstr. 40, 24098, Kiel, Germany.
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Olshausenstr. 40, 24098, Kiel, Germany.
| |
Collapse
|
2
|
Xu T, Heon-Roberts R, Moore T, Dubot P, Pan X, Guo T, Cairo CW, Holley R, Bigger B, Durcan TM, Levade T, Ausseil J, Amilhon B, Gorelik A, Nagar B, Sturiale L, Palmigiano A, Röckle I, Thiesler H, Hildebrandt H, Garozzo D, Pshezhetsky AV. Secondary deficiency of neuraminidase 1 contributes to CNS pathology in neurological mucopolysaccharidoses via hypersialylation of brain glycoproteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.587986. [PMID: 38712143 PMCID: PMC11071461 DOI: 10.1101/2024.04.26.587986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Mucopolysaccharidoses (MPS) are lysosomal storage diseases caused by defects in catabolism of glycosaminoglycans. MPS I, II, III and VII are associated with lysosomal accumulation of heparan sulphate and manifest with neurological deterioration. Most of these neurological MPS currently lack effective treatments. Here, we report that, compared to controls, neuraminidase 1 (NEU1) activity is drastically reduced in brain tissues of neurological MPS patients and in mouse models of MPS I, II, IIIA, IIIB and IIIC, but not of other neurological lysosomal disorders not presenting with heparan sulphate storage. We further show that accumulated heparan sulphate disrupts the lysosomal multienzyme complex of NEU1 with cathepsin A (CTSA), β-galactosidase (GLB1) and glucosamine-6-sulfate sulfatase (GALNS) necessary to maintain enzyme activity, and that NEU1 deficiency is linked to partial deficiencies of GLB1 and GALNS in cortical tissues and iPSC-derived cortical neurons of neurological MPS patients. Increased sialylation of N-linked glycans in brain samples of human MPS III patients and MPS IIIC mice implicated insufficient processing of brain N-linked sialylated glycans, except for polysialic acid, which was reduced in the brains of MPS IIIC mice. Correction of NEU1 activity in MPS IIIC mice by lentiviral gene transfer ameliorated previously identified hallmarks of the disease, including memory impairment, behavioural traits, and reduced levels of the excitatory synapse markers VGLUT1 and PSD95. Overexpression of NEU1 also restored levels of VGLUT1-/PSD95-positive puncta in cortical neurons derived from iPSC of an MPS IIIA patient. Together, our data demonstrate that heparan sulphate-induced secondary NEU1 deficiency and aberrant sialylation of glycoproteins implicated in synaptogenesis, memory, and behaviour constitute a novel pathological pathway in neurological MPS spectrum crucially contributing to CNS pathology. Graphical abstract
Collapse
|
3
|
Itoh K, Tsukimoto J. Lysosomal sialidase NEU1, its intracellular properties, deficiency, and use as a therapeutic agent. Glycoconj J 2023; 40:611-619. [PMID: 38147151 DOI: 10.1007/s10719-023-10135-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 10/14/2023] [Accepted: 10/18/2023] [Indexed: 12/27/2023]
Abstract
Neuraminidase 1 (NEU1) is a lysosomal sialidase that cleaves terminal α-linked sialic acid residues from sialylglycans. NEU1 is biosynthesized in the rough endoplasmic reticulum (RER) lumen as an N-glycosylated protein to associate with its protective protein/cathepsin A (CTSA) and then form a lysosomal multienzyme complex (LMC) also containing β-galactosidase 1 (GLB1). Unlike other mammalian sialidases, including NEU2 to NEU4, NEU1 transport to lysosomes requires association of NEU1 with CTSA, binding of the CTSA carrying terminal mannose 6-phosphate (M6P)-type N-glycan with M6P receptor (M6PR), and intralysosomal NEU1 activation at acidic pH. In contrast, overexpression of the single NEU1 gene in mammalian cells causes intracellular NEU1 protein crystallization in the RER due to self-aggregation when intracellular CTSA is reduced to a relatively low level. Sialidosis (SiD) and galactosialidosis (GS) are autosomal recessive lysosomal storage diseases caused by the gene mutations of NEU1 and CTSA, respectively. These incurable diseases associate with the NEU1 deficiency, excessive accumulation of sialylglycans in neurovisceral organs, and systemic manifestations. We established a novel GS model mouse carrying homozygotic Ctsa IVS6 + 1 g/a mutation causing partial exon 6 skipping with simultaneous deficiency of Ctsa and Neu1. Symptoms developed in the GS mice like those in juvenile/adult GS patients, such as myoclonic seizures, suppressed behavior, gargoyle-like face, edema, proctoptosis due to Neu1 deficiency, and sialylglycan accumulation associated with neurovisceral inflammation. We developed a modified NEU1 (modNEU1), which does not form protein crystals but is transported to lysosomes by co-expressed CTSA. In vivo gene therapy for GS and SiD utilizing a single adeno-associated virus (AAV) carrying modNEU1 and CTSA genes under dual promoter control will be created.
Collapse
Affiliation(s)
- Kohji Itoh
- Department of Medicinal Biotechnology, Institute for Medicinal Biotechnology, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan.
| | - Jun Tsukimoto
- Department of Medicinal Biotechnology, Institute for Medicinal Biotechnology, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
4
|
Kho I, Demina EP, Pan X, Londono I, Cairo CW, Sturiale L, Palmigiano A, Messina A, Garozzo D, Ung RV, Mac-Way F, Bonneil É, Thibault P, Lemaire M, Morales CR, Pshezhetsky AV. Severe kidney dysfunction in sialidosis mice reveals an essential role for neuraminidase 1 in reabsorption. JCI Insight 2023; 8:e166470. [PMID: 37698928 PMCID: PMC10619504 DOI: 10.1172/jci.insight.166470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 09/06/2023] [Indexed: 09/14/2023] Open
Abstract
Sialidosis is an ultra-rare multisystemic lysosomal disease caused by mutations in the neuraminidase 1 (NEU1) gene. The severe type II form of the disease manifests with a prenatal/infantile or juvenile onset, bone abnormalities, severe neuropathology, and visceromegaly. A subset of these patients present with nephrosialidosis, characterized by abrupt onset of fulminant glomerular nephropathy. We studied the pathophysiological mechanism of the disease in 2 NEU1-deficient mouse models, a constitutive Neu1-knockout, Neu1ΔEx3, and a conditional phagocyte-specific knockout, Neu1Cx3cr1ΔEx3. Mice of both strains exhibited terminal urinary retention and severe kidney damage with elevated urinary albumin levels, loss of nephrons, renal fibrosis, presence of storage vacuoles, and dysmorphic mitochondria in the intraglomerular and tubular cells. Glycoprotein sialylation in glomeruli, proximal distal tubules, and distal tubules was drastically increased, including that of an endocytic reabsorption receptor megalin. The pool of megalin bearing O-linked glycans with terminal galactose residues, essential for protein targeting and activity, was reduced to below detection levels. Megalin levels were severely reduced, and the protein was directed to lysosomes instead of the apical membrane. Together, our results demonstrated that desialylation by NEU1 plays a crucial role in processing and cellular trafficking of megalin and that NEU1 deficiency in sialidosis impairs megalin-mediated protein reabsorption.
Collapse
Affiliation(s)
- Ikhui Kho
- CHU Sainte-Justine Research Center, University of Montreal, Montreal, Québec, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, Québec, Canada
| | - Ekaterina P. Demina
- CHU Sainte-Justine Research Center, University of Montreal, Montreal, Québec, Canada
| | - Xuefang Pan
- CHU Sainte-Justine Research Center, University of Montreal, Montreal, Québec, Canada
| | - Irene Londono
- CHU Sainte-Justine Research Center, University of Montreal, Montreal, Québec, Canada
| | | | - Luisa Sturiale
- CNR, Institute for Polymers, Composites and Biomaterials, Catania, Italy
| | - Angelo Palmigiano
- CNR, Institute for Polymers, Composites and Biomaterials, Catania, Italy
| | - Angela Messina
- CNR, Institute for Polymers, Composites and Biomaterials, Catania, Italy
| | - Domenico Garozzo
- CNR, Institute for Polymers, Composites and Biomaterials, Catania, Italy
| | - Roth-Visal Ung
- CHU de Québec Research Center, L’Hôtel-Dieu de Québec Hospital, Faculty and Department of Medicine, University Laval, Québec City, Québec, Canada
| | - Fabrice Mac-Way
- CHU de Québec Research Center, L’Hôtel-Dieu de Québec Hospital, Faculty and Department of Medicine, University Laval, Québec City, Québec, Canada
| | - Éric Bonneil
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Québec, Canada
| | - Pierre Thibault
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Québec, Canada
| | - Mathieu Lemaire
- Division of Nephrology, The Hospital for Sick Kids, Faculty of Medicine, University of Toronto, Ontario, Canada
- Cell Biology Program, SickKids Research Institute, Toronto, Ontario, Canada
| | - Carlos R. Morales
- Department of Anatomy and Cell Biology, McGill University, Montreal, Québec, Canada
| | - Alexey V. Pshezhetsky
- CHU Sainte-Justine Research Center, University of Montreal, Montreal, Québec, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, Québec, Canada
| |
Collapse
|
5
|
Huertas J, Lee HT. Multi‑faceted roles of cathepsins in ischemia reperfusion injury (Review). Mol Med Rep 2022; 26:368. [PMID: 36300202 PMCID: PMC9644425 DOI: 10.3892/mmr.2022.12885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022] Open
Abstract
Cathepsins are one of the most abundant proteases within the lysosomes with diverse physiological effects ranging from immune responses, cell death and intracellular protein degradation. Cathepsins are involved in extracellular and systemic functions such as systemic inflammation and extracellular matrix degradation. Ischemia reperfusion (IR) injury is responsible for numerous diseases including myocardial infarction, acute kidney injury, stroke and acute graft failure after transplant surgery. Inflammation plays a major role in the reperfusion phase of IR injury and previous research has shown that cathepsins are key mediators of the inflammation cascade as well as apoptosis. Taken together, cathepsins modulation could provide potential therapeutic approaches to attenuate IR injury. The present review summarized the current understanding of various cathepsin subtypes, their major physiologic functions, their roles in multi‑organ IR injury and detailed selective cathepsin inhibitors with therapeutic potential.
Collapse
Affiliation(s)
- Jaime Huertas
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, NY 10032-3784, USA
| | - H. Thomas Lee
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, NY 10032-3784, USA
| |
Collapse
|
6
|
Del Grosso A, Parlanti G, Mezzena R, Cecchini M. Current treatment options and novel nanotechnology-driven enzyme replacement strategies for lysosomal storage disorders. Adv Drug Deliv Rev 2022; 188:114464. [PMID: 35878795 DOI: 10.1016/j.addr.2022.114464] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/26/2022] [Accepted: 07/19/2022] [Indexed: 11/01/2022]
Abstract
Lysosomal storage disorders (LSDs) are a vast group of more than 50 clinically identified metabolic diseases. They are singly rare, but they affect collectively 1 on 5,000 live births. They result in most of the cases from an enzymatic defect within lysosomes, which causes the subsequent augmentation of unwanted substrates. This accumulation process leads to plenty of clinical signs, determined by the specific substrate and accumulation area. The majority of LSDs present a broad organ and tissue engagement. Brain, connective tissues, viscera and bones are usually afflicted. Among them, brain disease is markedly frequent (two-thirds of LSDs). The most clinically employed approach to treat LSDs is enzyme replacement therapy (ERT), which is practiced by administering systemically the missed or defective enzyme. It represents a healthful strategy for 11 LSDs at the moment, but it solves the pathology only in the case of Gaucher disease. This approach, in fact, is not efficacious in the case of LSDs that have an effect on the central nervous system (CNS) due to the existence of the blood-brain barrier (BBB). Additionally, ERT suffers from several other weak points, such as low penetration of the exogenously administered enzyme to poorly vascularized areas, the development of immunogenicity and infusion-associated reactions (IARs), and, last but not least, the very high cost and lifelong needed. To ameliorate these weaknesses lot of efforts have been recently spent around the development of innovative nanotechnology-driven ERT strategies. They may boost the power of ERT and minimize adverse reactions by loading enzymes into biodegradable nanomaterials. Enzyme encapsulation into biocompatible liposomes, micelles, and polymeric nanoparticles, for example, can protect enzymatic activity, eliminating immunologic reactions and premature enzyme degradation. It can also permit a controlled release of the payload, ameliorating pharmacokinetics and pharmacodynamics of the drug. Additionally, the potential to functionalize the surface of the nanocarrier with targeting agents (antibodies or peptides), could promote the passage through biological barriers. In this review we examined the clinically applied ERTs, highlighting limitations that do not allow to completely cure the specific LSD. Later, we critically consider the nanotechnology-based ERT strategies that have beenin-vitroand/orin-vivotested to improve ERT efficacy.
Collapse
Affiliation(s)
- Ambra Del Grosso
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Gabriele Parlanti
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Roberta Mezzena
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Marco Cecchini
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| |
Collapse
|
7
|
Yu L, Peng J, Mineo C. Lipoprotein sialylation in atherosclerosis: Lessons from mice. Front Endocrinol (Lausanne) 2022; 13:953165. [PMID: 36157440 PMCID: PMC9498574 DOI: 10.3389/fendo.2022.953165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/15/2022] [Indexed: 11/22/2022] Open
Abstract
Sialylation is a dynamically regulated modification, which commonly occurs at the terminal of glycan chains in glycoproteins and glycolipids in eukaryotic cells. Sialylation plays a key role in a wide array of biological processes through the regulation of protein-protein interactions, intracellular localization, vesicular trafficking, and signal transduction. A majority of the proteins involved in lipoprotein metabolism and atherogenesis, such as apolipoproteins and lipoprotein receptors, are sialylated in their glycan structures. Earlier studies in humans and in preclinical models found a positive correlation between low sialylation of lipoproteins and atherosclerosis. More recent works using loss- and gain-of-function approaches in mice have revealed molecular and cellular mechanisms by which protein sialylation modulates causally the process of atherosclerosis. The purpose of this concise review is to summarize these findings in mouse models and to provide mechanistic insights into lipoprotein sialylation and atherosclerosis.
Collapse
Affiliation(s)
- Liming Yu
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jun Peng
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- *Correspondence: Chieko Mineo,
| |
Collapse
|
8
|
Abstract
Significance: The vascular extracellular matrix (ECM) not only provides mechanical stability but also manipulates vascular cell behaviors, which are crucial for vascular function and homeostasis. ECM remodeling, which alters vascular wall mechanical properties and exposes vascular cells to bioactive molecules, is involved in the development and progression of hypertension. Recent Advances: This brief review summarized the dynamic changes in ECM components and their modification and degradation during hypertension and after antihypertensive treatment. We also discussed how alterations in the ECM amount, assembly, mechanical properties, and degradation fragment generation provide input into the pathological process of hypertension. Critical Issues: Although the relevance between ECM remodeling and hypertension has been recognized, the underlying mechanism by which ECM remodeling initiates the development of hypertension remains unclear. Therefore, the modulation of ECM remodeling on arterial stiffness and hypertension in genetically modified rodent models is summarized in this review. The circulating biomarkers based on ECM metabolism and therapeutic strategies targeting ECM disorders in hypertension are also introduced. Future Directions: Further research will provide more comprehensive understanding of ECM remodeling in hypertension by the application of matridomic and degradomic approaches. The better understanding of mechanisms underlying vascular ECM remodeling may provide novel potential therapeutic strategies for preventing and treating hypertension. Antioxid. Redox Signal. 34, 765-783.
Collapse
Affiliation(s)
- Zeyu Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Li Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| |
Collapse
|
9
|
Cadaoas J, Hu H, Boyle G, Gomero E, Mosca R, Jayashankar K, Machado M, Cullen S, Guzman B, van de Vlekkert D, Annunziata I, Vellard M, Kakkis E, Koppaka V, d’Azzo A. Galactosialidosis: preclinical enzyme replacement therapy in a mouse model of the disease, a proof of concept. Mol Ther Methods Clin Dev 2021; 20:191-203. [PMID: 33426146 PMCID: PMC7782203 DOI: 10.1016/j.omtm.2020.11.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022]
Abstract
Galactosialidosis is a rare lysosomal storage disease caused by a congenital defect of protective protein/cathepsin A (PPCA) and secondary deficiency of neuraminidase-1 and β-galactosidase. PPCA is a lysosomal serine carboxypeptidase that functions as a chaperone for neuraminidase-1 and β-galactosidase within a lysosomal multi-protein complex. Combined deficiency of the three enzymes leads to accumulation of sialylated glycoproteins and oligosaccharides in tissues and body fluids and manifests in a systemic disease pathology with severity mostly correlating with the type of mutation(s) and age of onset of the symptoms. Here, we describe a proof-of-concept, preclinical study toward the development of enzyme replacement therapy for galactosialidosis, using a recombinant human PPCA. We show that the recombinant enzyme, taken up by patient-derived fibroblasts, restored cathepsin A, neuraminidase-1, and β-galactosidase activities. Long-term, bi-weekly injection of the recombinant enzyme in a cohort of mice with null mutation at the PPCA (CTSA) locus (PPCA -/- ), a faithful model of the disease, demonstrated a dose-dependent, systemic internalization of the enzyme by cells of various organs, including the brain. This resulted in restoration/normalization of the three enzyme activities, resolution of histopathology, and reduction of sialyloligosacchariduria. These positive results underscore the benefits of a PPCA-mediated enzyme replacement therapy for the treatment of galactosialidosis.
Collapse
Affiliation(s)
| | - Huimin Hu
- Department of Genetics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | | | - Elida Gomero
- Department of Genetics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Rosario Mosca
- Department of Genetics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | | | - Mike Machado
- Ultragenyx Pharmaceutical, Novato, CA 94949, USA
| | - Sean Cullen
- Ultragenyx Pharmaceutical, Novato, CA 94949, USA
| | - Belle Guzman
- Ultragenyx Pharmaceutical, Novato, CA 94949, USA
| | - Diantha van de Vlekkert
- Department of Genetics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Ida Annunziata
- Department of Genetics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | | | - Emil Kakkis
- Ultragenyx Pharmaceutical, Novato, CA 94949, USA
| | - Vish Koppaka
- Ultragenyx Pharmaceutical, Novato, CA 94949, USA
| | - Alessandra d’Azzo
- Department of Genetics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| |
Collapse
|
10
|
Demina EP, Smutova V, Pan X, Fougerat A, Guo T, Zou C, Chakraberty R, Snarr BD, Shiao TC, Roy R, Orekhov AN, Miyagi T, Laffargue M, Sheppard DC, Cairo CW, Pshezhetsky AV. Neuraminidases 1 and 3 Trigger Atherosclerosis by Desialylating Low-Density Lipoproteins and Increasing Their Uptake by Macrophages. J Am Heart Assoc 2021; 10:e018756. [PMID: 33554615 PMCID: PMC7955353 DOI: 10.1161/jaha.120.018756] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Chronic vascular disease atherosclerosis starts with an uptake of atherogenic modified low-density lipoproteins (LDLs) by resident macrophages, resulting in formation of arterial fatty streaks and eventually atheromatous plaques. Increased plasma sialic acid levels, increased neuraminidase activity, and reduced sialic acid LDL content have been previously associated with atherosclerosis and coronary artery disease in human patients, but the mechanism underlying this association has not been explored. Methods and Results We tested the hypothesis that neuraminidases contribute to development of atherosclerosis by removing sialic acid residues from glycan chains of the LDL glycoprotein and glycolipids. Atherosclerosis progression was investigated in apolipoprotein E and LDL receptor knockout mice with genetic deficiency of neuraminidases 1, 3, and 4 or those treated with specific neuraminidase inhibitors. We show that desialylation of the LDL glycoprotein, apolipoprotein B 100, by human neuraminidases 1 and 3 increases the uptake of human LDL by human cultured macrophages and by macrophages in aortic root lesions in Apoe-/- mice via asialoglycoprotein receptor 1. Genetic inactivation or pharmacological inhibition of neuraminidases 1 and 3 significantly delays formation of fatty streaks in the aortic root without affecting the plasma cholesterol and LDL levels in Apoe-/- and Ldlr-/- mouse models of atherosclerosis. Conclusions Together, our results suggest that neuraminidases 1 and 3 trigger the initial phase of atherosclerosis and formation of aortic fatty streaks by desialylating LDL and increasing their uptake by resident macrophages.
Collapse
Affiliation(s)
- Ekaterina P Demina
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Victoria Smutova
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Xuefang Pan
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Anne Fougerat
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| | - Tianlin Guo
- Department of Chemistry University of Alberta Edmonton Alberta Canada
| | - Chunxia Zou
- Department of Chemistry University of Alberta Edmonton Alberta Canada
| | | | - Brendan D Snarr
- Departments of Medicine, Microbiology and Immunology McGill University Montreal Quebec Canada
| | - Tze C Shiao
- Department of Chemistry Université du Québec à Montréal Montreal Quebec Canada
| | - Rene Roy
- Department of Chemistry Université du Québec à Montréal Montreal Quebec Canada
| | | | - Taeko Miyagi
- Miyagi Cancer Center Research Institute Natori Miyagi Japan
| | - Muriel Laffargue
- Institut National de la Santé et de la Recherche MédicaleUMR 1048Institute of Metabolic and Cardiovascular Diseases Toulouse France
| | - Donald C Sheppard
- Departments of Medicine, Microbiology and Immunology McGill University Montreal Quebec Canada
| | | | - Alexey V Pshezhetsky
- Departments of Pediatrics and Biochemistry Sainte-Justine University Hospital Research CenterUniversity of Montreal Quebec Canada
| |
Collapse
|
11
|
Hohl M, Mayr M, Lang L, Nickel AG, Barallobre-Barreiro J, Yin X, Speer T, Selejan SR, Goettsch C, Erb K, Fecher-Trost C, Reil JC, Linz B, Ruf S, Hübschle T, Maack C, Böhm M, Sadowski T, Linz D. Cathepsin A contributes to left ventricular remodeling by degrading extracellular superoxide dismutase in mice. J Biol Chem 2020; 295:12605-12617. [PMID: 32647007 DOI: 10.1074/jbc.ra120.013488] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/29/2020] [Indexed: 11/06/2022] Open
Abstract
In the heart, the serine carboxypeptidase cathepsin A (CatA) is distributed between lysosomes and the extracellular matrix (ECM). CatA-mediated degradation of extracellular peptides may contribute to ECM remodeling and left ventricular (LV) dysfunction. Here, we aimed to evaluate the effects of CatA overexpression on LV remodeling. A proteomic analysis of the secretome of adult mouse cardiac fibroblasts upon digestion by CatA identified the extracellular antioxidant enzyme superoxide dismutase (EC-SOD) as a novel substrate of CatA, which decreased EC-SOD abundance 5-fold. In vitro, both cardiomyocytes and cardiac fibroblasts expressed and secreted CatA protein, and only cardiac fibroblasts expressed and secreted EC-SOD protein. Cardiomyocyte-specific CatA overexpression and increased CatA activity in the LV of transgenic mice (CatA-TG) reduced EC-SOD protein levels by 43%. Loss of EC-SOD-mediated antioxidative activity resulted in significant accumulation of superoxide radicals (WT, 4.54 μmol/mg tissue/min; CatA-TG, 8.62 μmol/mg tissue/min), increased inflammation, myocyte hypertrophy (WT, 19.8 μm; CatA-TG, 21.9 μm), cellular apoptosis, and elevated mRNA expression of hypertrophy-related and profibrotic marker genes, without affecting intracellular detoxifying proteins. In CatA-TG mice, LV interstitial fibrosis formation was enhanced by 19%, and the type I/type III collagen ratio was shifted toward higher abundance of collagen I fibers. Cardiac remodeling in CatA-TG was accompanied by an increased LV weight/body weight ratio and LV end diastolic volume (WT, 50.8 μl; CatA-TG, 61.9 μl). In conclusion, CatA-mediated EC-SOD reduction in the heart contributes to increased oxidative stress, myocyte hypertrophy, ECM remodeling, and inflammation, implicating CatA as a potential therapeutic target to prevent ventricular remodeling.
Collapse
Affiliation(s)
- Mathias Hohl
- Klinik für Innere Medizin III, Universität des Saarlandes, Homburg/Saar, Germany
| | - Manuel Mayr
- King's BHF Centre of Research Excellence, The James Black Centre, London, United Kingdom
| | - Lisa Lang
- Klinik für Innere Medizin III, Universität des Saarlandes, Homburg/Saar, Germany
| | - Alexander G Nickel
- Klinik für Innere Medizin III, Universität des Saarlandes, Homburg/Saar, Germany.,Universitätsklinikum Würzburg, Deutsches Zentrum für Herzinsuffizienz (DZHI), Comprehensive Heart Failure Center (CHFC), Würzburg, Germany
| | | | - Xiaoke Yin
- King's BHF Centre of Research Excellence, The James Black Centre, London, United Kingdom
| | - Thimoteus Speer
- Klinik für Innere Medizin IV, Universität des Saarlandes, Homburg/Saar, Germany
| | | | - Claudia Goettsch
- Medizinische Fakultät, Medizinische Klinik 1, Kardiologie, Universitätsklinikum, Aachen, Germany
| | - Katharina Erb
- Klinik für Innere Medizin III, Universität des Saarlandes, Homburg/Saar, Germany
| | - Claudia Fecher-Trost
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie Universität des Saarlandes, Homburg/Saar, Germany
| | - Jan-Christian Reil
- Klinik für Innere Medizin III, Universität des Saarlandes, Homburg/Saar, Germany.,Klinik für Innere Medizin II, Universitäres Herzzentrum, Lübeck, Germany
| | - Benedikt Linz
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sven Ruf
- Sanofi-Aventis Deutschland GmbH, Frankfurt, Germany
| | | | - Christoph Maack
- Klinik für Innere Medizin III, Universität des Saarlandes, Homburg/Saar, Germany.,Universitätsklinikum Würzburg, Deutsches Zentrum für Herzinsuffizienz (DZHI), Comprehensive Heart Failure Center (CHFC), Würzburg, Germany
| | - Michael Böhm
- Klinik für Innere Medizin III, Universität des Saarlandes, Homburg/Saar, Germany
| | | | - Dominik Linz
- Klinik für Innere Medizin III, Universität des Saarlandes, Homburg/Saar, Germany .,University Maastricht, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands.,Department of Cardiology, Maastricht University Medical Centre, Maastricht, the Netherlands.,Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
| |
Collapse
|
12
|
Bouknight KD, Jurkouich KM, Compton JR, Khavrutskii IV, Guelta MA, Harvey SP, Legler PM. Structural and kinetic evidence of aging after organophosphate inhibition of human Cathepsin A. Biochem Pharmacol 2020; 177:113980. [PMID: 32305437 DOI: 10.1016/j.bcp.2020.113980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/14/2020] [Indexed: 11/30/2022]
Abstract
Human Cathepsin A (CatA) is a lysosomal serine carboxypeptidase of the renin-angiotensin system (RAS) and is structurally similar to acetylcholinesterase (AChE). CatA can remove the C-terminal amino acids of endothelin I, angiotensin I, Substance P, oxytocin, and bradykinin, and can deamidate neurokinin A. Proteomic studies identified CatA and its homologue, SCPEP1, as potential targets of organophosphates (OP). CatA could be stably inhibited by low µM to high nM concentrations of racemic sarin (GB), soman (GD), cyclosarin (GF), VX, and VR within minutes to hours at pH 7. Cyclosarin was the most potent with a kinetically measured dissociation constant (KI) of 2 µM followed by VR (KI = 2.8 µM). Bimolecular rate constants for inhibition by cyclosarin and VR were 1.3 × 103 M-1sec-1 and 1.2 × 103 M-1sec-1, respectively, and were approximately 3-orders of magnitude lower than those of human AChE indicating slower reactivity. Notably, both AChE and CatA bound diisopropylfluorophosphate (DFP) comparably and had KIDFP = 13 µM and 11 µM, respectively. At low pH, greater than 85% of the enzyme spontaneously reactivated after OP inhibition, conditions under which OP-adducts of cholinesterases irreversibly age. At pH 6.5 CatA remained stably inhibited by GB and GF and <10% of the enzyme spontaneously reactivated after 200 h. A crystal structure of DFP-inhibited CatA was determined and contained an aged adduct. Similar to AChE, CatA appears to have a "backdoor" for product release. CatA has not been shown previously to age. These results may have implications for: OP-associated inflammation; cardiovascular effects; and the dysregulation of RAS enzymes by OP.
Collapse
Affiliation(s)
- Kayla D Bouknight
- Hampton University, 100 E Queen St, Hampton, VA 23668, United States
| | - Kayla M Jurkouich
- Case Western Reserve University, Dept. of Biomedical Engineering, Cleveland, 10900 Euclid Avenue, OH 44106, United States
| | - Jaimee R Compton
- U.S. Naval Research Laboratory, 4555 Overlook Ave., Washington, DC 20375, United States
| | - Ilja V Khavrutskii
- Uniformed Services University, Armed Forces Radiobiology Research Institute, 4301 Jones Bridge Rd., Bethesda, MD 20889-5648, United States
| | - Mark A Guelta
- U.S. Army Combat Capabilities and Development Command Chemical Biological Center, 5183 Blackhawk Road, Aberdeen Proving Ground, MD 21010, United States
| | - Steven P Harvey
- U.S. Army Combat Capabilities and Development Command Chemical Biological Center, 5183 Blackhawk Road, Aberdeen Proving Ground, MD 21010, United States
| | - Patricia M Legler
- U.S. Naval Research Laboratory, 4555 Overlook Ave., Washington, DC 20375, United States.
| |
Collapse
|
13
|
De Pasquale V, Moles A, Pavone LM. Cathepsins in the Pathophysiology of Mucopolysaccharidoses: New Perspectives for Therapy. Cells 2020; 9:cells9040979. [PMID: 32326609 PMCID: PMC7227001 DOI: 10.3390/cells9040979] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
Cathepsins (CTSs) are ubiquitously expressed proteases normally found in the endolysosomal compartment where they mediate protein degradation and turnover. However, CTSs are also found in the cytoplasm, nucleus, and extracellular matrix where they actively participate in cell signaling, protein processing, and trafficking through the plasma and nuclear membranes and between intracellular organelles. Dysregulation in CTS expression and/or activity disrupts cellular homeostasis, thus contributing to many human diseases, including inflammatory and cardiovascular diseases, neurodegenerative disorders, diabetes, obesity, cancer, kidney dysfunction, and others. This review aimed to highlight the involvement of CTSs in inherited lysosomal storage disorders, with a primary focus to the emerging evidence on the role of CTSs in the pathophysiology of Mucopolysaccharidoses (MPSs). These latter diseases are characterized by severe neurological, skeletal and cardiovascular phenotypes, and no effective cure exists to date. The advance in the knowledge of the molecular mechanisms underlying the activity of CTSs in MPSs may open a new challenge for the development of novel therapeutic approaches for the cure of such intractable diseases.
Collapse
Affiliation(s)
- Valeria De Pasquale
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy;
| | - Anna Moles
- Institute of Biomedical Research of Barcelona, Spanish Research Council, 08036 Barcelona, Spain;
| | - Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy;
- Correspondence: ; Tel.: +39-081-7463043
| |
Collapse
|
14
|
Davison JE. Eye involvement in inherited metabolic disorders. Ther Adv Ophthalmol 2020; 12:2515841420979109. [PMID: 33447730 PMCID: PMC7780305 DOI: 10.1177/2515841420979109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 11/12/2020] [Indexed: 12/22/2022] Open
Abstract
Inherited metabolic disorders are a large group of rare disorders affecting normal biochemical pathways. Many metabolic disorders can present with symptoms affecting the eye, and eye disorders can evolve later in the natural history of an already diagnosed metabolic disorder. The ophthalmic involvement can be very varied affecting any part of the eye, including abnormalities of cornea, lens dislocation and cataracts, retina and the distal optic pathway, and extraocular muscles. Awareness of inherited metabolic disorders is important to facilitate early diagnosis and in some cases instigate early treatment if a patient presents with eye involvement suggestive of a metabolic disorder. Ophthalmological interventions are also an important component of the multisystem holistic approach to treating patients with metabolic disorders.
Collapse
Affiliation(s)
- James E. Davison
- Metabolic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, NIHR GOSH Biomedical Research Centre (BRC), London WC1N 3JH, UK
| |
Collapse
|
15
|
Daamen WF, Quaglino D. Signaling pathways in elastic tissues. Cell Signal 2019; 63:109364. [DOI: 10.1016/j.cellsig.2019.109364] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 07/22/2019] [Indexed: 02/06/2023]
|
16
|
The role of elastin-derived peptides in human physiology and diseases. Matrix Biol 2019; 84:81-96. [PMID: 31295577 DOI: 10.1016/j.matbio.2019.07.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/03/2019] [Accepted: 07/07/2019] [Indexed: 12/12/2022]
Abstract
Once considered as inert, the extracellular matrix recently revealed to be biologically active. Elastin is one of the most important components of the extracellular matrix. Many vital organs including arteries, lungs and skin contain high amounts of elastin to assure their correct function. Physiologically, the organism contains a determined quantity of elastin from the early development which may remain physiologically constant due to its very long half-life and very low turnover. Taking into consideration the continuously ongoing challenges during life, there is a physiological degradation of elastin into elastin-derived peptides which is accentuated in several disease states such as obstructive pulmonary diseases, atherosclerosis and aortic aneurysm. These elastin-derived peptides have been shown to have various biological effects mediated through their interaction with their cognate receptor called elastin receptor complex eliciting several signal transduction pathways. In this review, we will describe the production and the biological effects of elastin-derived peptides in physiology and pathology.
Collapse
|
17
|
Bennasroune A, Romier-Crouzet B, Blaise S, Laffargue M, Efremov RG, Martiny L, Maurice P, Duca L. Elastic fibers and elastin receptor complex: Neuraminidase-1 takes the center stage. Matrix Biol 2019; 84:57-67. [PMID: 31226402 DOI: 10.1016/j.matbio.2019.06.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/11/2019] [Accepted: 06/17/2019] [Indexed: 01/02/2023]
Abstract
Extracellular matrix (ECM) has for a long time being considered as a simple architectural support for cells. It is now clear that ECM presents a fundamental influence on cells driving their phenotype and fate. This complex network is highly specialized and the different classes of macromolecules that comprise the ECM determine its biological functions. For instance, collagens are responsible for the tensile strength of tissues, proteoglycans and glycosaminoglycans are essential for hydration and resistance to compression, and glycoproteins such as laminins facilitate cell attachment. The largest structures of the ECM are the elastic fibers found in abundance in tissues suffering high mechanical constraints such as skin, lungs or arteries. These structures present a very complex composition whose core is composed of elastin surrounded by a microfibrils mantle. Elastogenesis is a tightly regulated process involving the sialidase activity of the Neuraminidase-1 (Neu-1) sub-unit of the Elastin Receptor Complex. Interestingly, Neu-1 subunit also serves as a sensor of elastin degradation via its ability to transmit elastin-derived peptides signaling. Finally, reports showing that neuraminidase activity is able to regulate TGF-β activation raises questions about a possible role for Neu-1 in elastic fibers remodeling. In this mini review, we develop the concept of the regulation of the whole life of elastic fibers through an original scope, the key role of Neu-1 sialidase enzymatic activity.
Collapse
Affiliation(s)
- Amar Bennasroune
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | | | - Sébastien Blaise
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Muriel Laffargue
- UMR INSERM 1048 I2MC, Université Paul Sabatier, Toulouse, France
| | - Roman G Efremov
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Higher School of Economics, Myasnitskaya ul. 20, 101000 Moscow, Russia
| | - Laurent Martiny
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Pascal Maurice
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Laurent Duca
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France.
| |
Collapse
|
18
|
Hohl M, Erb K, Lang L, Ruf S, Hübschle T, Dhein S, Linz W, Elliott AD, Sanders P, Zamyatkin O, Böhm M, Schotten U, Sadowski T, Linz D. Cathepsin A Mediates Ventricular Remote Remodeling and Atrial Cardiomyopathy in Rats With Ventricular Ischemia/Reperfusion. JACC Basic Transl Sci 2019; 4:332-344. [PMID: 31312757 PMCID: PMC6609908 DOI: 10.1016/j.jacbts.2019.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/08/2019] [Accepted: 01/11/2019] [Indexed: 01/14/2023]
Abstract
After myocardial infarction, remote ventricular remodeling and atrial cardiomyopathy progress despite successful revascularization. In a rat model of ventricular ischemia/reperfusion, pharmacological inhibition of the protease activity of cathepsin A initiated at the time point of reperfusion prevented extracellular matrix remodeling in the atrium and the ventricle remote from the infarcted area. This scenario was associated with preservation of more viable ventricular myocardium and the prevention of an arrhythmogenic and functional substrate for atrial fibrillation. Remote ventricular extracellular matrix remodeling and atrial cardiomyopathy may represent a promising target for pharmacological atrial fibrillation upstream therapy following myocardial infarction.
Collapse
Key Words
- AF, atrial fibrillation
- CatA, cathepsin A
- Cx43, connexin 43
- ECM, extracellular matrix
- I/R, ischemia/reperfusion
- ICM, ischemic cardiomyopathy
- LA, left atrial
- LAD, left anterior descending coronary artery
- LV, left ventricular
- MRI, magnetic resonance imaging
- PL, permanent left anterior descending ligation
- SAR, (S)-3-{[1-(2-Fluoro-phenyl)-5-methoxy-1H-pyrazole-3-carbonyl]-amino}-3-o-tolyl-propionic-acid
- atrial cardiomyopathy
- atrial fibrillation
- ischemia/reperfusion
- mRNA, messenger ribonucleic acid
- myocardial infarction
- remote remodeling
Collapse
Affiliation(s)
- Mathias Hohl
- Klinik für Innere Medizin III, Universität des Saarlandes, Homburg/Saar, Germany
| | - Katharina Erb
- Klinik für Innere Medizin III, Universität des Saarlandes, Homburg/Saar, Germany
| | - Lisa Lang
- Klinik für Innere Medizin III, Universität des Saarlandes, Homburg/Saar, Germany
| | - Sven Ruf
- Sanofi-Aventis Deutschland GmbH, Frankfurt, Germany
| | | | - Stefan Dhein
- Herzzentrum Leipzig Abt. Herzchirurgie, Leipzig, Germany
| | | | - Adrian D. Elliott
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
| | - Prashanthan Sanders
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
| | - Olesja Zamyatkin
- Klinik für Innere Medizin III, Universität des Saarlandes, Homburg/Saar, Germany
| | - Michael Böhm
- Klinik für Innere Medizin III, Universität des Saarlandes, Homburg/Saar, Germany
| | - Ulrich Schotten
- Department of Physiology, University of Maastricht, Maastricht, the Netherlands
| | | | - Dominik Linz
- Klinik für Innere Medizin III, Universität des Saarlandes, Homburg/Saar, Germany
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
| |
Collapse
|
19
|
Affiliation(s)
- Randy T. Cowling
- Department of Medicine, Division of Cardiovascular Medicine, University of California-San Diego, La Jolla, California
| |
Collapse
|
20
|
Pshezhetsky AV, Ashmarina M. Keeping it trim: roles of neuraminidases in CNS function. Glycoconj J 2018; 35:375-386. [PMID: 30088207 PMCID: PMC6182584 DOI: 10.1007/s10719-018-9837-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 12/11/2022]
Abstract
The sialylated glyconjugates (SGC) are found in abundance on the surface of brain cells, where they form a dense array of glycans mediating cell/cell and cell/protein recognition in numerous physiological and pathological processes. Metabolic genetic blocks in processing and catabolism of SGC result in development of severe storage disorders, dominated by CNS involvement including marked neuroinflammation and neurodegeneration, the pathophysiological mechanisms of which are still discussed. SGC patterns in the brain are cell and organelle-specific, dynamic and maintained by highly coordinated processes of their biosynthesis, trafficking, processing and catabolism. The changes in the composition of SGC during development and aging of the brain cannot be explained based solely on the regulation of the SGC-synthesizing enzymes, sialyltransferases, suggesting that neuraminidases (sialidases) hydrolysing the removal of terminal sialic acid residues also play an essential role. In the current review we summarize the roles of three mammalian neuraminidases: neuraminidase 1, neuraminidase 3 and neuraminidase 4 in processing brain SGC. Emerging data demonstrate that these enzymes with different, yet overlapping expression patterns, intracellular localization and substrate specificity play essential roles in the physiology of the CNS.
Collapse
Affiliation(s)
- Alexey V Pshezhetsky
- Sainte-Justine Hospital Research Center, Department of Paediatrics, University of Montreal, CHU Ste-Justine, Centre de recherche, 3175 Côte-Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada.
- Department of Anatomy and Cell Biology, McGill University, Montreal, H3A0C7, Canada.
| | - Mila Ashmarina
- Sainte-Justine Hospital Research Center, Department of Paediatrics, University of Montreal, CHU Ste-Justine, Centre de recherche, 3175 Côte-Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada
| |
Collapse
|
21
|
van Rooden EJ, van Esbroeck ACM, Baggelaar MP, Deng H, Florea BI, Marques ARA, Ottenhoff R, Boot RG, Overkleeft HS, Aerts JMFG, van der Stelt M. Chemical Proteomic Analysis of Serine Hydrolase Activity in Niemann-Pick Type C Mouse Brain. Front Neurosci 2018; 12:440. [PMID: 30018533 PMCID: PMC6037894 DOI: 10.3389/fnins.2018.00440] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/11/2018] [Indexed: 12/27/2022] Open
Abstract
The endocannabinoid system (ECS) is considered to be an endogenous protective system in various neurodegenerative diseases. Niemann-Pick type C (NPC) is a neurodegenerative disease in which the role of the ECS has not been studied yet. Most of the endocannabinoid enzymes are serine hydrolases, which can be studied using activity-based protein profiling (ABPP). Here, we report the serine hydrolase activity in brain proteomes of a NPC mouse model as measured by ABPP. Two ABPP methods are used: a gel-based method and a chemical proteomics method. The activities of the following endocannabinoid enzymes were quantified: diacylglycerol lipase (DAGL) α, α/β-hydrolase domain-containing protein 4, α/β-hydrolase domain-containing protein 6, α/β-hydrolase domain-containing protein 12, fatty acid amide hydrolase, and monoacylglycerol lipase. Using the gel-based method, two bands were observed for DAGL α. Only the upper band corresponding to this enzyme was significantly decreased in the NPC mouse model. Chemical proteomics showed that three lysosomal serine hydrolase activities (retinoid-inducible serine carboxypeptidase, cathepsin A, and palmitoyl-protein thioesterase 1) were increased in Niemann-Pick C1 protein knockout mouse brain compared to wild-type brain, whereas no difference in endocannabinoid hydrolase activity was observed. We conclude that these targets might be interesting therapeutic targets for future validation studies.
Collapse
Affiliation(s)
- Eva J van Rooden
- Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | | | - Marc P Baggelaar
- Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Hui Deng
- Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Bogdan I Florea
- Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - André R A Marques
- Institute of Biochemistry, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Roelof Ottenhoff
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Rolf G Boot
- Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Herman S Overkleeft
- Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Johannes M F G Aerts
- Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Mario van der Stelt
- Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| |
Collapse
|
22
|
Fougerat A, Pan X, Smutova V, Heveker N, Cairo CW, Issad T, Larrivée B, Medin JA, Pshezhetsky AV. Neuraminidase 1 activates insulin receptor and reverses insulin resistance in obese mice. Mol Metab 2018; 12:76-88. [PMID: 29735266 PMCID: PMC6001920 DOI: 10.1016/j.molmet.2018.03.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/23/2018] [Accepted: 03/30/2018] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Neuraminidase 1 (NEU1) cleaves terminal sialic acids of glycoconjugates during lysosomal catabolism. It also modulates the structure and activity of cellular surface receptors affecting diverse pathways. Previously we demonstrated that NEU1 activates the insulin receptor (IR) and that NEU1-deficient CathAS190A-Neo mice (hypomorph of the NEU1 activator protein, cathepsin A/CathA) on a high-fat diet (HFD) develop hyperglycaemia and insulin resistance faster than wild-type animals. The major objective of the current work was to reveal the molecular mechanism by which NEU1 desialylation activates the IR and to test if increase of NEU1 activity in insulin target tissues reverses insulin resistance and glucose intolerance. METHODS To test if desialylation causes a conformational change in the IR dimer we measured interaction between the receptor subunits by Bioluminescence Resonance Energy Transfer in the HEK293T cells either overexpressing NEU1 or treated with the NEU1 inhibitor. The influence of NEU1 overexpression on insulin resistance was studied in vitro in palmitate-treated HepG2 cells transduced with NEU1-expressing lentivirus and in vivo in C57Bl6 mice treated with HFD and either pharmacological inducer of NEU1, Ambroxol or NEU1-expressing adenovirus. NEU1-deficient CathAS190A-Neo mice were used as a control. RESULTS By desialylation of IR, NEU1 induced formation of its active dimer leading to insulin signaling. Overexpression of NEU1 in palmitate-treated HepG2 cells restored insulin signaling, suggesting that increased NEU1 levels may reverse insulin resistance. Five-day treatment of glycemic C57Bl6 mice receiving HFD with the activator of the lysosomal gene network, Ambroxol, increased NEU1 expression and activity in muscle tissue, normalized fasting glucose levels, and improved physiological and molecular responses to glucose and insulin. Ambroxol did not improve insulin sensitivity in obese insulin-resistant CathAS190A-Neo mice indicating that the Ambroxol effect is mediated through NEU1 induction. Sustained increase of liver NEU1 activity through adenovirus-based gene transfer failed to attenuate insulin resistance most probably due to negative feedback regulation of IR expression. CONCLUSION Together our results demonstrate that increase of NEU1 activity in insulin target tissues reverses insulin resistance and glucose intolerance suggesting that a pharmacological modulation of NEU1 activity may be potentially explored for restoring insulin sensitivity and resolving hyperglycemia associated with T2DM.
Collapse
Affiliation(s)
- Anne Fougerat
- CHU Sainte-Justine Research Centre, Departments of Biochemistry and Pediatrics, University of Montreal, Montreal, Canada
| | - Xuefang Pan
- CHU Sainte-Justine Research Centre, Departments of Biochemistry and Pediatrics, University of Montreal, Montreal, Canada
| | - Victoria Smutova
- CHU Sainte-Justine Research Centre, Departments of Biochemistry and Pediatrics, University of Montreal, Montreal, Canada
| | - Nikolaus Heveker
- CHU Sainte-Justine Research Centre, Departments of Biochemistry and Pediatrics, University of Montreal, Montreal, Canada
| | - Christopher W Cairo
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada
| | - Tarik Issad
- INSERM U1016, CNRS UMR8104, Université Paris Descartes Sorbonne Paris Cité, Institut Cochin, Paris, France
| | - Bruno Larrivée
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Canada
| | | | - Alexey V Pshezhetsky
- CHU Sainte-Justine Research Centre, Departments of Biochemistry and Pediatrics, University of Montreal, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada.
| |
Collapse
|
23
|
Guo T, Dätwyler P, Demina E, Richards MR, Ge P, Zou C, Zheng R, Fougerat A, Pshezhetsky AV, Ernst B, Cairo CW. Selective Inhibitors of Human Neuraminidase 3. J Med Chem 2018; 61:1990-2008. [PMID: 29425031 DOI: 10.1021/acs.jmedchem.7b01574] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human neuraminidases (NEU) are associated with human diseases including cancer, atherosclerosis, and diabetes. To obtain small molecule inhibitors as research tools for the study of their biological functions, we designed a library of 2-deoxy-2,3-didehydro- N-acetylneuraminic acid (DANA) analogues with modifications at C4 and C9 positions. This library allowed us to discover selective inhibitors targeting the human NEU3 isoenzyme. Our most selective inhibitor for NEU3 has a Ki of 320 ± 40 nM and a 15-fold selectivity over other human neuraminidase isoenzymes. This inhibitor blocks glycolipid processing by NEU3 in vitro. To improve their pharmacokinetic properties, various esters of the best inhibitors were synthesized and evaluated. Finally, we confirmed that our best compounds exhibited selective inhibition of NEU orthologues from murine brain.
Collapse
Affiliation(s)
- Tianlin Guo
- Alberta Glycomics Centre and Department of Chemistry , University of Alberta , Edmonton Alberta T6G 2G2 , Canada
| | - Philipp Dätwyler
- Department of Pharmaceutical Sciences, Pharmacenter , University of Basel , Klingelbergstrasse 50 , CH-4056 Basel , Switzerland
| | - Ekaterina Demina
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center , University of Montreal , Montréal , Quebec H3T 1C5 , Canada
| | - Michele R Richards
- Alberta Glycomics Centre and Department of Chemistry , University of Alberta , Edmonton Alberta T6G 2G2 , Canada
| | - Peng Ge
- Alberta Glycomics Centre and Department of Chemistry , University of Alberta , Edmonton Alberta T6G 2G2 , Canada
| | - Chunxia Zou
- Alberta Glycomics Centre and Department of Chemistry , University of Alberta , Edmonton Alberta T6G 2G2 , Canada
| | - Ruixiang Zheng
- Alberta Glycomics Centre and Department of Chemistry , University of Alberta , Edmonton Alberta T6G 2G2 , Canada
| | - Anne Fougerat
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center , University of Montreal , Montréal , Quebec H3T 1C5 , Canada
| | - Alexey V Pshezhetsky
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center , University of Montreal , Montréal , Quebec H3T 1C5 , Canada
| | - Beat Ernst
- Department of Pharmaceutical Sciences, Pharmacenter , University of Basel , Klingelbergstrasse 50 , CH-4056 Basel , Switzerland
| | - Christopher W Cairo
- Alberta Glycomics Centre and Department of Chemistry , University of Alberta , Edmonton Alberta T6G 2G2 , Canada
| |
Collapse
|
24
|
Pan X, Wang Y, Lübke T, Hinek A, Pshezhetsky AV. Mice, double deficient in lysosomal serine carboxypeptidases Scpep1 and Cathepsin A develop the hyperproliferative vesicular corneal dystrophy and hypertrophic skin thickenings. PLoS One 2017; 12:e0172854. [PMID: 28234994 PMCID: PMC5325571 DOI: 10.1371/journal.pone.0172854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/17/2017] [Indexed: 01/04/2023] Open
Abstract
Vasoactive and mitogenic peptide, endothelin-1 (ET-1) plays an important role in physiology of the ocular tissues by regulating the growth of corneal epithelial cells and maintaining the hemodynamics of intraocular fluids. We have previously established that ET-1 can be degraded in vivo by two lysosomal/secreted serine carboxypeptidases, Cathepsin A (CathA) and Serine Carboxypeptidase 1 (Scpep1) and that gene-targeted CathAS190A/Scpep1-/- mice, deficient in CathA and Scpep1 have a prolonged half-life of circulating ET-1 associated with systemic hypertension. In the current work we report that starting from 6 months of age, ~43% of CathAS190A/Scpep1-/- mice developed corneal clouding that eventually caused vision impairment. Histological evaluation of these mice demonstrated a selective fibrotic thickening and vacuolization of the corneas, resembling human hyperproliferative vesicular corneal stromal dystrophy and coexisting with a peculiar thickening of the skin epidermis. Moreover, we found that cultured corneal epithelial cells, skin fibroblasts and vascular smooth muscle cells derived from CathA/Scpep1-deficient mice, demonstrated a significantly higher proliferative response to treatment with exogenous ET-1, as compared with cells from wild type mice. We also detected increased activation level of ERK1/2 and AKT kinases involved in cell proliferation in the ET-1-treated cultured cells from CathA/Scpep1 deficient mice. Together, results from our experimental model suggest that; in normal tissues the tandem of serine carboxypeptidases, Scpep1 and CathA likely constitutes an important part of the physiological mechanism responsible for the balanced elimination of heightened levels of ET-1 that otherwise would accumulate in tissues and consequently contribute to development of the hyper-proliferative corneal dystrophy and abnormal skin thickening.
Collapse
Affiliation(s)
- Xuefang Pan
- Department of Medical Genetics, CHU Sainte-Justine Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Yanting Wang
- Cardiovascular Research Program, the Hospital for Sick Children and Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | - Torben Lübke
- Department of Chemistry, Biochemistry I, Bielefeld University, Bielefeld, Germany
| | - Aleksander Hinek
- Cardiovascular Research Program, the Hospital for Sick Children and Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | - Alexey V. Pshezhetsky
- Department of Medical Genetics, CHU Sainte-Justine Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
25
|
Mice with Catalytically Inactive Cathepsin A Display Neurobehavioral Alterations. Behav Neurol 2017; 2017:4261873. [PMID: 28133419 PMCID: PMC5241486 DOI: 10.1155/2017/4261873] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 12/05/2016] [Indexed: 11/18/2022] Open
Abstract
The lysosomal carboxypeptidase A, Cathepsin A (CathA), is a serine protease with two distinct functions. CathA protects β-galactosidase and sialidase Neu1 against proteolytic degradation by forming a multienzyme complex and activates sialidase Neu1. CathA deficiency causes the lysosomal storage disease, galactosialidosis. These patients present with a broad range of clinical phenotypes, including growth retardation, and neurological deterioration along with the accumulation of the vasoactive peptide, endothelin-1, in the brain. Previous in vitro studies have shown that CathA has specific activity against vasoactive peptides and neuropeptides, including endothelin-1 and oxytocin. A mutant mouse with catalytically inactive CathA enzyme (CathAS190A) shows increased levels of endothelin-1. In the present study, we elucidated the involvement of CathA in learning and long-term memory in 3-, 6-, and 12-month-old mice. Hippocampal endothelin-1 and oxytocin accumulated in CathAS190A mice, which showed learning impairments as well as long-term and spatial memory deficits compared with wild-type littermates, suggesting that CathA plays a significant role in learning and in memory consolidation through its regulatory role in vasoactive peptide processing.
Collapse
|
26
|
Annunziata I, d'Azzo A. Galactosialidosis: historic aspects and overview of investigated and emerging treatment options. Expert Opin Orphan Drugs 2016; 5:131-141. [PMID: 28603679 DOI: 10.1080/21678707.2016.1266933] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Galactosialidosis is a glycoprotein storage disease caused by mutations in the CTSA gene, encoding lysosomal protective protein/cathepsin A (PPCA). The enzyme's catalytic activity is distinct from its protective function towards β-galactosidase (β-GAL) and neuraminidase 1 (NEU1), with which PPCA forms a complex. In this configuration the two glycosidases acquire their full activity and stability in lysosomes. Deficiency of PPCA results in combined NEU1/β-GAL deficiency. Because of its low incidence, galactosialidosis is considered an orphan disorder with no therapy yet available. AREAS COVERED This review gives a historic overview on the discovery of PPCA, which defined galactosialidosis as a new clinical entity; the evidence for the existence of the PPCA/NEU1/β-GAL complex; the clinical forms of galactosialidosis and disease-causing CTSA mutations. Ppca-/- mice have proven to be a suitable model to test different therapeutic approaches, paving the way for the development of clinical trials for patients with galactosialidosis. EXPERT OPINION Improved understanding of the molecular bases of disease has sparked renewed incentive from clinicians and scientists alike to develop therapies for rare conditions, like GS, and has increased the willingness of biotech companies to invest in the manufacturing of new therapeutics. Both ERT and gene therapy may become available to patients in the near future.
Collapse
Affiliation(s)
- Ida Annunziata
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Alessandra d'Azzo
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
27
|
Timur ZK, Akyildiz Demir S, Seyrantepe V. Lysosomal Cathepsin A Plays a Significant Role in the Processing of Endogenous Bioactive Peptides. Front Mol Biosci 2016; 3:68. [PMID: 27826550 PMCID: PMC5078471 DOI: 10.3389/fmolb.2016.00068] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/07/2016] [Indexed: 11/13/2022] Open
Abstract
Lysosomal serine carboxypeptidase Cathepsin A (CTSA) is a multifunctional enzyme with distinct protective and catalytic function. CTSA present in the lysosomal multienzyme complex to facilitate the correct lysosomal routing, stability and activation of with beta-galactosidase and alpha-neuraminidase. Beside CTSA has role in inactivation of bioactive peptides including bradykinin, substances P, oxytocin, angiotensin I and endothelin-I by cleavage of 1 or 2 amino acid(s) from C-terminal ends. In this study, we aimed to elucidate the regulatory role of CTSA on bioactive peptides in knock-in mice model of CTSAS190A . We investigated the level of bradykinin, substances P, oxytocin, angiotensin I and endothelin-I in the kidney, liver, lung, brain and serum from CTSAS190A mouse model at 3- and 6-months of age. Our results suggest CTSA selectively contributes to processing of bioactive peptides in different tissues from CTSAS190A mice compared to age matched WT mice.
Collapse
Affiliation(s)
- Zehra Kevser Timur
- Izmir Institute of Technology, Molecular Biology and Genetics Izmir, Turkey
| | | | - Volkan Seyrantepe
- Izmir Institute of Technology, Molecular Biology and Genetics Izmir, Turkey
| |
Collapse
|
28
|
Petrera A, Kern U, Linz D, Gomez-Auli A, Hohl M, Gassenhuber J, Sadowski T, Schilling O. Proteomic Profiling of Cardiomyocyte-Specific Cathepsin A Overexpression Links Cathepsin A to the Oxidative Stress Response. J Proteome Res 2016; 15:3188-95. [PMID: 27432266 DOI: 10.1021/acs.jproteome.6b00413] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cathepsin A (CTSA) is a lysosomal carboxypeptidase present at the cell surface and secreted outside the cell. Additionally, CTSA binds to β-galactosidase and neuraminidase 1 to protect them from degradation. CTSA has gained attention as a drug target for the treatment of cardiac hypertrophy and heart failure. Here, we investigated the impact of CTSA on the murine cardiac proteome in a mouse model of cardiomyocyte-specific human CTSA overexpression using liquid chromatography-tandem mass spectrometry in conjunction with an isotopic dimethyl labeling strategy. We identified up to 2000 proteins in each of three biological replicates. Statistical analysis by linear models for microarray data (limma) found >300 significantly affected proteins (moderated p-value ≤0.01), thus establishing CTSA as a key modulator of the cardiac proteome. CTSA strongly impaired the balance of the proteolytic system by upregulating several proteases such as cathepsin B, cathepsin D, and cathepsin Z while down-regulating numerous protease inhibitors. Moreover, cardiomyocyte-specific human CTSA overexpression strongly reduced the levels of numerous antioxidative stress proteins, i.e., peroxiredoxins and protein deglycase DJ-1. In vitro, using cultured rat cardiomyocytes, ectopic overexpression of CTSA resulted in accumulation of reactive oxygen species. Collectively, our proteomic and functional data strengthen an association of CTSA with the cellular oxidative stress response.
Collapse
Affiliation(s)
- Agnese Petrera
- Institute of Molecular Medicine and Cell Research, University of Freiburg , Stefan Meier Strasse 17, 79104 Freiburg, Germany
| | - Ursula Kern
- Institute of Molecular Medicine and Cell Research, University of Freiburg , Stefan Meier Strasse 17, 79104 Freiburg, Germany
| | - Dominik Linz
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes , 66424 Homburg/Saar, Germany
| | - Alejandro Gomez-Auli
- Institute of Molecular Medicine and Cell Research, University of Freiburg , Stefan Meier Strasse 17, 79104 Freiburg, Germany
| | - Mathias Hohl
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes , 66424 Homburg/Saar, Germany
| | - Johann Gassenhuber
- Diabetes Division, Sanofi-Aventis Deutschland GmbH , 65926 Frankfurt, Germany
| | - Thorsten Sadowski
- Diabetes Division, Sanofi-Aventis Deutschland GmbH , 65926 Frankfurt, Germany
| | - Oliver Schilling
- Institute of Molecular Medicine and Cell Research, University of Freiburg , Stefan Meier Strasse 17, 79104 Freiburg, Germany
| |
Collapse
|
29
|
Petrera A, Gassenhuber J, Ruf S, Gunasekaran D, Esser J, Shahinian JH, Hübschle T, Rütten H, Sadowski T, Schilling O. Cathepsin A inhibition attenuates myocardial infarction-induced heart failure on the functional and proteomic levels. J Transl Med 2016; 14:153. [PMID: 27246731 PMCID: PMC4888645 DOI: 10.1186/s12967-016-0907-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 05/13/2016] [Indexed: 01/05/2023] Open
Abstract
Background Myocardial infarction (MI) is a major cause of heart failure. The carboxypeptidase cathepsin A is a novel target in the treatment of cardiac failure. We aim to show that recently developed inhibitors of the protease cathepsin A attenuate post-MI heart failure. Methods Mice were subjected to permanent left anterior descending artery (LAD) ligation or sham operation. 24 h post–surgery, LAD-ligated animals were treated with daily doses of the cathepsin A inhibitor SAR1 or placebo. After 4 weeks, the three groups (sham, MI-placebo, MI-SAR1) were evaluated. Results Compared to sham-operated animals, placebo-treated mice showed significantly impaired cardiac function and increased plasma BNP levels. Cathepsin A inhibition prevented the increase of plasma BNP levels and displayed a trend towards improved cardiac functionality. Proteomic profiling was performed for the three groups (sham, MI-placebo, MI-SAR1). More than 100 proteins were significantly altered in placebo-treated LAD ligation compared to the sham operation, including known markers of cardiac failure as well as extracellular/matricellular proteins. This ensemble constitutes a proteome fingerprint of myocardial infarction induced by LAD ligation in mice. Cathepsin A inhibitor treatment normalized the marked increase of the muscle stress marker CA3 as well as of Igγ 2b and fatty acid synthase. For numerous further proteins, cathepsin A inhibition partially dampened the LAD ligation-induced proteome alterations. Conclusions Our proteomic and functional data suggest that cathepsin A inhibition has cardioprotective properties and support a beneficial effect of cathepsin A inhibition in the treatment of heart failure after myocardial infarction. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0907-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Agnese Petrera
- Institute for Molecular Medicine and Cell Research, University of Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany
| | - Johann Gassenhuber
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926, Frankfurt Am Main, Germany
| | - Sven Ruf
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926, Frankfurt Am Main, Germany
| | - Deepika Gunasekaran
- Institute for Molecular Medicine and Cell Research, University of Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany
| | - Jennifer Esser
- Department of Cardiology and Angiology, University Heart Center Freiburg, University of Freiburg, Breisacher Strasse 33, 79106, Freiburg, Germany
| | - Jasmin Hasmik Shahinian
- Department of Cardiac Surgery, University Hospital Basel, Spitalstrasse 21, Basel, Switzerland
| | - Thomas Hübschle
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926, Frankfurt Am Main, Germany
| | - Hartmut Rütten
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926, Frankfurt Am Main, Germany
| | - Thorsten Sadowski
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926, Frankfurt Am Main, Germany
| | - Oliver Schilling
- Institute for Molecular Medicine and Cell Research, University of Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany. .,BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79104, Freiburg, Germany. .,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
30
|
Linz D, Hohl M, Dhein S, Ruf S, Reil JC, Kabiri M, Wohlfart P, Verheule S, Böhm M, Sadowski T, Schotten U. Cathepsin A mediates susceptibility to atrial tachyarrhythmia and impairment of atrial emptying function in Zucker diabetic fatty rats. Cardiovasc Res 2016; 110:371-80. [DOI: 10.1093/cvr/cvw071] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/25/2016] [Indexed: 02/01/2023] Open
|
31
|
Duca L, Blaise S, Romier B, Laffargue M, Gayral S, El Btaouri H, Kawecki C, Guillot A, Martiny L, Debelle L, Maurice P. Matrix ageing and vascular impacts: focus on elastin fragmentation. Cardiovasc Res 2016; 110:298-308. [DOI: 10.1093/cvr/cvw061] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/07/2016] [Indexed: 12/17/2022] Open
|
32
|
Tillner J, Lehmann A, Paehler T, Lukacs Z, Ruf S, Sadowski T, Pinquier JL, Ruetten H. Tolerability, safety, and pharmacokinetics of the novel cathepsin A inhibitor SAR164653 in healthy subjects. Clin Pharmacol Drug Dev 2015; 5:57-68. [PMID: 27119579 DOI: 10.1002/cpdd.201] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 05/06/2015] [Indexed: 11/11/2022]
Abstract
Cathepsin A (CathA) is a lysosomal protein where it forms a stable complex with neuraminidase and ß-galactosidase. CathA also has enzymatic activity and is involved in the degradation of many peptides. CathA was recently discovered as a target for heart failure, fostering the development of CathA inhibitors with SAR164653 as a frontrunner. The first-in-man study investigated single oral doses from 20 to 800 mg of SAR164653 followed by repeat dose studies at doses up to 800 mg in healthy young and elderly subjects. SAR164653 was safe and well tolerated at doses up to 800 mg in healthy subjects, and a maximum tolerated dose could not be determined from the study. Activity of ß-galactosidase measured in leukocytes did not show any abnormalities. The tmax was 1.0 to 2.5 hours, and the t1/2 was ∼5-11 after single dosing; exposure increased less than dose proportional. Following multiple dosing, accumulation was not observed, Cmax and AUC0-24 increased in a dose-proportional manner, and t1/2 was around 14-20 hours. The novel CathA inhibitor SAR164653 was found to have a favorable safety profile in these early phase 1 studies, but further studies are required to confirm if SAR164653 is equally safe in patients undergoing long-term treatment.
Collapse
Affiliation(s)
| | - Anne Lehmann
- Sanofi-Aventis Deutschland, Frankfurt a.M., Germany
| | | | - Zoltan Lukacs
- Hamburg University Medical Center, Metabolic Laboratory, Hamburg, Germany
| | - Sven Ruf
- Sanofi-Aventis Deutschland, Frankfurt a.M., Germany
| | | | | | | |
Collapse
|
33
|
Timur ZK, Akyildiz Demir S, Marsching C, Sandhoff R, Seyrantepe V. Neuraminidase-1 contributes significantly to the degradation of neuronal B-series gangliosides but not to the bypass of the catabolic block in Tay-Sachs mouse models. Mol Genet Metab Rep 2015; 4:72-82. [PMID: 26937414 PMCID: PMC4750590 DOI: 10.1016/j.ymgmr.2015.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 07/23/2015] [Accepted: 07/23/2015] [Indexed: 11/17/2022] Open
Affiliation(s)
- Z K Timur
- Izmir Institute of Technology, Department of Molecular Biology and Genetics, Izmir, Turkey
| | - S Akyildiz Demir
- Izmir Institute of Technology, Department of Molecular Biology and Genetics, Izmir, Turkey
| | - C Marsching
- Lipid Biochemistry Lab, Cancer Research Center, Heidelberg, Germany
| | - R Sandhoff
- Lipid Biochemistry Lab, Cancer Research Center, Heidelberg, Germany
| | - V Seyrantepe
- Izmir Institute of Technology, Department of Molecular Biology and Genetics, Izmir, Turkey
| |
Collapse
|
34
|
Qin Z. Soluble elastin peptides in cardiovascular homeostasis: Foe or ally. Peptides 2015; 67:64-73. [PMID: 25794852 DOI: 10.1016/j.peptides.2015.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 03/04/2015] [Accepted: 03/09/2015] [Indexed: 12/23/2022]
Abstract
Elastin peptides, also known as elastin-derived peptides or elastokines, are soluble polypeptides in blood and tissue. The blood levels of elastin peptides are usually low but can increase during cardiovascular diseases, such as atherosclerosis, aortic aneurysm and diabetes with vascular complications. Generally, elastin peptides are derived from the degradation of insoluble elastic polymers. The biological activities of elastin peptides are bidirectional, e.g., a pro-inflammatory effect on monocyte migration induction vs. a protective effect on vasodilation promotion. However, recent in vivo studies have demonstrated that elastin peptides promote the formation of atherosclerotic plaques in hypercholesterolemic mice and induce hyperglycemia and elevations in plasma lipid levels in fasted mice. More important, the detrimental effects induced by elastin peptides can be largely inhibited by genetic or pharmacological blockade of the elastin receptor complex or by neutralization of an antibody against elastin peptides. These studies indicate new therapeutic strategies for the treatment of cardiovascular diseases by targeting elastin peptide metabolism. Therefore, the goal of this review is to summarize current knowledge about elastin peptides relevant to cardiovascular pathologies to further delineate their potential application in cardiovascular disease.
Collapse
Affiliation(s)
- Zhenyu Qin
- Division of Vascular Surgery, Department of Surgery, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.
| |
Collapse
|
35
|
Abstract
INTRODUCTION Sialidosis is a neurosomatic, lysosomal storage disease (LSD) caused by mutations in the NEU1 gene, encoding the lysosomal sialidase NEU1. Deficient enzyme activity results in impaired processing/degradation of sialo-glycoproteins, and accumulation of oversialylated metabolites. Sialidosis is considered an orphan disorder for which no therapy is currently available. AREAS COVERED The review describes the clinical forms of sialidosis and the NEU1 mutations so far identified; NEU1 requirement to complex with the protective protein/cathepsin A for stability and activation; and the pathogenic effects of NEU1 deficiency. Studies of the molecular mechanisms of pathogenesis in animal models uncovered basic cellular pathways downstream of NEU1 and its substrates, which may be implicated in more common adult (neurodegenerative) diseases. The development of a Phase I/II clinical trial for patients with galactosialidosis may prove suitable for sialidosis patients with the attenuated form of the disease. EXPERT OPINION Recently, there has been a renewed interest in the development of therapies for orphan LSDs, like sialidosis. Given the small number of potentially eligible patients, the way to treat sialidosis would be through the coordinated effort of clinical centers, which provide diagnosis and care for these patients, and the basic research labs that work towards understanding the disease pathogenesis.
Collapse
Affiliation(s)
- Alessandra d'Azzo
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Eda Machado
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ida Annunziata
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
36
|
Abstract
All three members of the endothelin (ET) family of peptides, ET-1, ET-2, and ET-3, are expressed in the human kidney, with ET-1 being the predominant isoform. ET-1 and ET-2 bind to two G-protein-coupled receptors, ETA and ETB, whereas at physiological concentrations ET-3 has little affinity for the ET(A) receptor. The human kidney is unusual among the peripheral organs in expressing a high density of ET(B). The renal vascular endothelium only expresses the ET(B) subtype and ET-1 acts in an autocrine or paracrine manner to release vasodilators. Endothelial ETB in kidney, as well as liver and lungs, also has a critical role in scavenging ET-1 from the plasma. The third major function is ET-1 activation of ET(B) in in the nephron to reduce salt and water re-absorption. In contrast, ET(A) predominate on smooth muscle, causing vasoconstriction and mediating many of the pathophysiological actions of ET-1. The role of the two receptors has been delineated using highly selective ET(A) (BQ123, TAK-044) and ET(B) (BQ788) peptide antagonists. Nonpeptide antagonists, bosentan, macitentan, and ambrisentan, that are either mixed ET(A)/ET(B) antagonists or display ET(A) selectivity, have been approved for clinical use but to date are limited to pulmonary hypertension. Ambrisentan is in clinical trials in patients with type 2 diabetic nephropathy. This review summarizes ET-receptor antagonism in the human kidney, and considers the relative merits of selective versus nonselective antagonism in renal disease.
Collapse
Affiliation(s)
- Janet J Maguire
- Clinical Pharmacology Unit, University of Cambridge, Addenbrooke׳s Hospital, Cambridge, United Kingdom
| | - Anthony P Davenport
- Clinical Pharmacology Unit, University of Cambridge, Addenbrooke׳s Hospital, Cambridge, United Kingdom.
| |
Collapse
|
37
|
Maguire JJ, Davenport AP. Endothelin@25 - new agonists, antagonists, inhibitors and emerging research frontiers: IUPHAR Review 12. Br J Pharmacol 2014; 171:5555-72. [PMID: 25131455 PMCID: PMC4290702 DOI: 10.1111/bph.12874] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 07/22/2014] [Accepted: 07/25/2014] [Indexed: 12/16/2022] Open
Abstract
Since the discovery of endothelin (ET)-1 in 1988, the main components of the signalling pathway have become established, comprising three structurally similar endogenous 21-amino acid peptides, ET-1, ET-2 and ET-3, that activate two GPCRs, ETA and ETB . Our aim in this review is to highlight the recent progress in ET research. The ET-like domain peptide, corresponding to prepro-ET-193-166 , has been proposed to be co-synthesized and released with ET-1, to modulate the actions of the peptide. ET-1 remains the most potent vasoconstrictor in the human cardiovascular system with a particularly long-lasting action. To date, the major therapeutic strategy to block the unwanted actions of ET in disease, principally in pulmonary arterial hypertension, has been to use antagonists that are selective for the ETA receptor (ambrisentan) or that block both receptor subtypes (bosentan). Macitentan represents the next generation of antagonists, being more potent than bosentan, with longer receptor occupancy and it is converted to an active metabolite; properties contributing to greater pharmacodynamic and pharmacokinetic efficacy. A second strategy is now being more widely tested in clinical trials and uses combined inhibitors of ET-converting enzyme and neutral endopeptidase such as SLV306 (daglutril). A third strategy based on activating the ETB receptor, has led to the renaissance of the modified peptide agonist IRL1620 as a clinical candidate in delivering anti-tumour drugs and as a pharmacological tool to investigate experimental pathophysiological conditions. Finally, we discuss biased signalling, epigenetic regulation and targeting with monoclonal antibodies as prospective new areas for ET research.
Collapse
Affiliation(s)
- J J Maguire
- Clinical Pharmacology Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | | |
Collapse
|
38
|
Petrera A, Lai ZW, Schilling O. Carboxyterminal protein processing in health and disease: key actors and emerging technologies. J Proteome Res 2014; 13:4497-504. [PMID: 25204196 DOI: 10.1021/pr5005746] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Carboxypeptidases are important mediators of cellular behavior. Through C-terminal truncations, they alter protein functionality and participate in proteome turnover. Similarly, carboxypeptidases shape the human peptidome by targeting neuroendocrine and vasoactive peptides, thereby regulating signaling pathways in the nervous and cardiovascular systems as well as in embryonic development. Carboxypeptidases are widely connected to various pathological processes such as carcinogenesis and neurodegenerative and cardiovascular diseases. The repertoire of carboxypeptidase in vivo substrates still remains poorly defined, largely due to the lack of suitable experimental approaches. Understanding the precise role of carboxypeptidases is pivotal in the future development of diagnostic/prognostic markers in such diseases. To date, very little attention has been paid to the implication of carboxypeptidases in shaping the proteome as well as the peptidome. This review focuses on the patho-physiological function of carboxypeptidases and highlights the approaches by which proteomics-based technologies can be applied to characterize carboxypeptidases and to quantify the differential regulation of proteins by carboxypeptidases in a proteome-wide manner.
Collapse
Affiliation(s)
- Agnese Petrera
- Institute of Molecular Medicine and Cell Research, ‡BIOSS Centre for Biological Signaling Studies, University of Freiburg , D-79104 Freiburg, Germany
| | | | | |
Collapse
|
39
|
Smutova V, Albohy A, Pan X, Korchagina E, Miyagi T, Bovin N, Cairo CW, Pshezhetsky AV. Structural basis for substrate specificity of mammalian neuraminidases. PLoS One 2014; 9:e106320. [PMID: 25222608 PMCID: PMC4164519 DOI: 10.1371/journal.pone.0106320] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 07/24/2014] [Indexed: 12/21/2022] Open
Abstract
The removal of sialic acid (Sia) residues from glycoconjugates in vertebrates is mediated by a family of neuraminidases (sialidases) consisting of Neu1, Neu2, Neu3 and Neu4 enzymes. The enzymes play distinct physiological roles, but their ability to discriminate between the types of linkages connecting Sia and adjacent residues and between the identity and arrangement of the underlying sugars has never been systematically studied. Here we analyzed the specificity of neuraminidases by studying the kinetics of hydrolysis of BODIPY-labeled substrates containing common mammalian sialylated oligosaccharides: 3′Sia-LacNAc, 3′SiaLac, SiaLex, SiaLea, SiaLec, 6′SiaLac, and 6′SiaLacNAc. We found significant differences in substrate specificity of the enzymes towards the substrates containing α2,6-linked Sia, which were readily cleaved by Neu3 and Neu1 but not by Neu4 and Neu2. The presence of a branching 2-Fuc inhibited Neu2 and Neu4, but had almost no effect on Neu1 or Neu3. The nature of the sugar residue at the reducing end, either glucose (Glc) or N-acetyl-D-glucosamine (GlcNAc) had only a minor effect on all neuraminidases, whereas core structure (1,3 or 1,4 bond between D-galactose (Gal) and GlcNAc) was found to be important for Neu4 strongly preferring β3 (core 1) to β4 (core 2) isomer. Neu3 and Neu4 were in general more active than Neu1 and Neu2, likely due to their preference for hydrophobic substrates. Neu2 and Neu3 were examined by molecular dynamics to identify favorable substrate orientations in the binding sites and interpret the differences in their specificities. Finally, using knockout mouse models, we confirmed that the substrate specificities observed in vitro were recapitulated in enzymes found in mouse brain tissues. Our data for the first time provide evidence for the characteristic substrate preferences of neuraminidases and their ability to discriminate between distinct sialoside targets.
Collapse
Affiliation(s)
- Victoria Smutova
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montreal, Montréal, Canada
| | - Amgad Albohy
- Alberta Glycomics Center, Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Xuefang Pan
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montreal, Montréal, Canada
| | - Elena Korchagina
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Taeko Miyagi
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan
| | - Nicolai Bovin
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Christopher W. Cairo
- Alberta Glycomics Center, Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Alexey V. Pshezhetsky
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montreal, Montréal, Canada
- * E-mail:
| |
Collapse
|
40
|
Halvorsen AR, Helland A, Fleischer T, Haug KM, Grenaker Alnaes GI, Nebdal D, Syljuåsen RG, Touleimat N, Busato F, Tost J, Saetersdal AB, Børresen-Dale AL, Kristensen V, Edvardsen H. Differential DNA methylation analysis of breast cancer reveals the impact of immune signaling in radiation therapy. Int J Cancer 2014; 135:2085-95. [PMID: 24658971 PMCID: PMC4298788 DOI: 10.1002/ijc.28862] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 02/27/2014] [Accepted: 03/12/2014] [Indexed: 01/26/2023]
Abstract
Radiotherapy (RT) is a central treatment modality for breast cancer patients. The purpose of our study was to investigate the DNA methylation changes in tumors following RT, and to identify epigenetic markers predicting treatment outcome. Paired biopsies from patients with inoperable breast cancer were collected both before irradiation (n = 20) and after receiving 10-24 Gray (Gy) (n = 19). DNA methylation analysis was performed by using Illumina Infinium 27K arrays. Fourteen genes were selected for technical validation by pyrosequencing. Eighty-two differentially methylated genes were identified in irradiated (n = 11) versus nonirradiated (n = 19) samples (false discovery rate, FDR = 1.1%). Methylation levels in pathways belonging to the immune system were most altered after RT. Based on methylation levels before irradiation, a panel of five genes (H2AFY, CTSA, LTC4S, IL5RA and RB1) were significantly associated with clinical response (p = 0.041). Furthermore, the degree of methylation changes for 2,516 probes correlated with the given radiation dose. Within the 2,516 probes, an enrichment for pathways involved in cellular immune response, proliferation and apoptosis was identified (FDR < 5%). Here, we observed clear differences in methylation levels induced by radiation, some associated with response to treatment. Our study adds knowledge on the molecular mechanisms behind radiation response.
Collapse
Affiliation(s)
- Ann Rita Halvorsen
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Pshezhetsky AV, Ashmarina LI. Desialylation of surface receptors as a new dimension in cell signaling. BIOCHEMISTRY (MOSCOW) 2014; 78:736-45. [PMID: 24010837 DOI: 10.1134/s0006297913070067] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Terminal sialic acid residues are found in abundance in glycan chains of glycoproteins and glycolipids on the surface of all live cells forming an outer layer of the cell originally known as glycocalyx. Their presence affects the molecular properties and structure of glycoconjugates, modifying their function and interactions with other molecules. Consequently, the sialylation state of glycoproteins and glycolipids has been recognized as a critical factor modulating molecular recognitions inside the cell, between the cells, between the cells and the extracellular matrix, and between the cells and certain exogenous pathogens. Until recently sialyltransferases that catalyze transfer of sialic acid residues to the glycan chains in the process of their biosynthesis were thought to be mainly responsible for the creation and maintenance of a temporal and spatial diversity of sialylated moieties. However, the growing evidence suggests that in mammalian cells, at least equally important roles belong to sialidases/neuraminidases, which are located on the cell surface and in intracellular compartments, and may either initiate the catabolism of sialoglycoconjugates or just cleave their sialic acid residues, and thereby contribute to temporal changes in their structure and functions. The current review summarizes emerging data demonstrating that mammalian neuraminidase 1, well known for its lysosomal catabolic function, is also targeted to the cell surface and assumes the previously unrecognized role as a structural and functional modulator of cellular receptors.
Collapse
Affiliation(s)
- A V Pshezhetsky
- Department of Medical Genetics, CHU Sainte-Justine Research Center, Montreal, Qc, H3T1C5, Canada.
| | | |
Collapse
|
42
|
Serine carboxypeptidase SCPEP1 and Cathepsin A play complementary roles in regulation of vasoconstriction via inactivation of endothelin-1. PLoS Genet 2014; 10:e1004146. [PMID: 24586188 PMCID: PMC3937211 DOI: 10.1371/journal.pgen.1004146] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 12/13/2013] [Indexed: 11/19/2022] Open
Abstract
The potent vasoconstrictor peptides, endothelin 1 (ET-1) and angiotensin II control adaptation of blood vessels to fluctuations of blood pressure. Previously we have shown that the circulating level of ET-1 is regulated through its proteolytic cleavage by secreted serine carboxypeptidase, cathepsin A (CathA). However, genetically-modified mouse expressing catalytically inactive CathA S190A mutant retained about 10-15% of the carboxypeptidase activity against ET-1 in its tissues suggesting a presence of parallel/redundant catabolic pathway(s). In the current work we provide direct evidence that the enzyme, which complements CathA action towards ET-1 is a retinoid-inducible lysosomal serine carboxypeptidase 1 (Scpep1), a CathA homolog with previously unknown biological function. We generated a mouse strain devoid of both CathA and Scpep1 activities (DD mice) and found that in response to high-salt diet and systemic injections of ET-1 these animals showed significantly increased blood pressure as compared to wild type mice or those with single deficiencies of CathA or Scpep1. We also found that the reactivity of mesenteric arteries from DD mice towards ET-1 was significantly higher than that for all other groups of mice. The DD mice had a reduced degradation rate of ET-1 in the blood whereas their cultured arterial vascular smooth muscle cells showed increased ET-1-dependent phosphorylation of myosin light chain 2. Together, our results define the biological role of mammalian serine carboxypeptidase Scpep1 and suggest that Scpep1 and CathA together participate in the control of ET-1 regulation of vascular tone and hemodynamics.
Collapse
|
43
|
Schreuder HA, Liesum A, Kroll K, Böhnisch B, Buning C, Ruf S, Sadowski T. Crystal structure of cathepsin A, a novel target for the treatment of cardiovascular diseases. Biochem Biophys Res Commun 2014; 445:451-6. [PMID: 24530914 DOI: 10.1016/j.bbrc.2014.02.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 02/05/2014] [Indexed: 10/25/2022]
Abstract
The lysosomal serine carboxypeptidase cathepsin A is involved in the breakdown of peptide hormones like endothelin and bradykinin. Recent pharmacological studies with cathepsin A inhibitors in rodents showed a remarkable reduction in cardiac hypertrophy and atrial fibrillation, making cathepsin A a promising target for the treatment of heart failure. Here we describe the crystal structures of activated cathepsin A without inhibitor and with two compounds that mimic the tetrahedral intermediate and the reaction product, respectively. The structure of activated cathepsin A turned out to be very similar to the structure of the inactive precursor. The only difference was the removal of a 40 residue activation domain, partially due to proteolytic removal of the activation peptide, and partially by an order-disorder transition of the peptides flanking the removed activation peptide. The termini of the catalytic core are held together by the Cys253-Cys303 disulfide bond, just before and after the activation domain. One of the compounds we soaked in our crystals reacted covalently with the catalytic Ser150 and formed a tetrahedral intermediate. The other compound got cleaved by the enzyme and a fragment, resembling one of the natural reaction products, was found in the active site. These studies establish cathepsin A as a classical serine proteinase with a well-defined oxyanion hole. The carboxylate group of the cleavage product is bound by a hydrogen-bonding network involving one aspartate and two glutamate side chains. This network can only form if at least half of the carboxylate groups involved are protonated, which explains the acidic pH optimum of the enzyme.
Collapse
Affiliation(s)
- Herman A Schreuder
- Sanofi-Aventis Pharma Deutschland GmbH, Industriepark Höchst, 65926 Frankfurt am Main, Germany.
| | - Alexander Liesum
- Sanofi-Aventis Pharma Deutschland GmbH, Industriepark Höchst, 65926 Frankfurt am Main, Germany.
| | - Katja Kroll
- Sanofi-Aventis Pharma Deutschland GmbH, Industriepark Höchst, 65926 Frankfurt am Main, Germany.
| | - Britta Böhnisch
- Sanofi-Aventis Pharma Deutschland GmbH, Industriepark Höchst, 65926 Frankfurt am Main, Germany.
| | - Christian Buning
- Sanofi-Aventis Pharma Deutschland GmbH, Industriepark Höchst, 65926 Frankfurt am Main, Germany.
| | - Sven Ruf
- Sanofi-Aventis Pharma Deutschland GmbH, Industriepark Höchst, 65926 Frankfurt am Main, Germany.
| | - Thorsten Sadowski
- Sanofi-Aventis Pharma Deutschland GmbH, Industriepark Höchst, 65926 Frankfurt am Main, Germany.
| |
Collapse
|
44
|
Gayral S, Garnotel R, Castaing-Berthou A, Blaise S, Fougerat A, Berge E, Montheil A, Malet N, Wymann MP, Maurice P, Debelle L, Martiny L, Martinez LO, Pshezhetsky AV, Duca L, Laffargue M. Elastin-derived peptides potentiate atherosclerosis through the immune Neu1-PI3Kγ pathway. Cardiovasc Res 2013; 102:118-27. [PMID: 24357053 DOI: 10.1093/cvr/cvt336] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Elastin is degraded during vascular ageing and its products, elastin-derived peptides (EP), are present in the human blood circulation. EP binds to the elastin receptor complex (ERC) at the cell surface, composed of elastin-binding protein (EBP), a cathepsin A and a neuraminidase 1. Some in vitro functions have clearly been attributed to this binding, but the in vivo implications for arterial diseases have never been clearly investigated. METHODS AND RESULTS Here, we demonstrate that chronic doses of EP injected into mouse models of atherosclerosis increase atherosclerotic plaque size formation. Similar effects were observed following an injection of a VGVAPG peptide, suggesting that the ERC mediates these effects. The absence of phosphoinositide 3-kinase γ (PI3Kγ) in bone marrow-derived cells prevented EP-induced atherosclerosis development, demonstrating that PI3Kγ drive EP-induced arterial lesions. Accordingly, in vitro studies showed that PI3Kγ was required for EP-induced monocyte migration and ROS production and that this effect was dependent upon neuraminidase activity. Finally, we showed that degradation of elastic lamellae in LDLR(-/-) mice fed an atherogenic diet correlated with atherosclerotic plaque formation. At the same time, the absence of the cathepsin A-neuraminidase 1 complex in cells of the haematopoietic lineage abolished atheroma plaque size progression and decreased leucocytes infiltration, clearly demonstrating the role of this complex in atherogenesis and suggesting the involvement of endogenous EP. CONCLUSION Altogether, this work identifies EP as an enhancer of atherogenesis and defines the Neuraminidase 1/PI3Kγ signalling pathway as a key mediator of this function in vitro and in vivo.
Collapse
Affiliation(s)
- Stephanie Gayral
- INSERM UMR 1048, I2MC, Bât. L3, 1 av Jean-Poulhès, BP 84225, 31432 Toulouse Cedex 4, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Lysosomal multienzyme complex: pros and cons of working together. Cell Mol Life Sci 2013; 71:2017-32. [PMID: 24337808 DOI: 10.1007/s00018-013-1538-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 11/29/2013] [Accepted: 12/02/2013] [Indexed: 12/31/2022]
Abstract
The ubiquitous distribution of lysosomes and their heterogeneous protein composition reflects the versatility of these organelles in maintaining cell homeostasis and their importance in tissue differentiation and remodeling. In lysosomes, the degradation of complex, macromolecular substrates requires the synergistic action of multiple hydrolases that usually work in a stepwise fashion. This catalytic machinery explains the existence of lysosomal enzyme complexes that can be dynamically assembled and disassembled to efficiently and quickly adapt to the pool of substrates to be processed or degraded, adding extra tiers to the regulation of the individual protein components. An example of such a complex is the one composed of three hydrolases that are ubiquitously but differentially expressed: the serine carboxypeptidase, protective protein/cathepsin A (PPCA), the sialidase, neuraminidase-1 (NEU1), and the glycosidase β-galactosidase (β-GAL). Next to this 'core' complex, the existence of sub-complexes, which may contain additional components, and function at the cell surface or extracellularly, suggests as yet unexplored functions of these enzymes. Here we review how studies of basic biological processes in the mouse models of three lysosomal storage disorders, galactosialidosis, sialidosis, and GM1-gangliosidosis, revealed new and unexpected roles for the three respective affected enzymes, Ppca, Neu1, and β-Gal, that go beyond their canonical degradative activities. These findings have broadened our perspective on their functions and may pave the way for the development of new therapies for these lysosomal storage disorders.
Collapse
|
46
|
Tan GJ, Peng ZK, Lu JP, Tang FQ. Cathepsins mediate tumor metastasis. World J Biol Chem 2013; 4:91-101. [PMID: 24340132 PMCID: PMC3856311 DOI: 10.4331/wjbc.v4.i4.91] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 09/06/2013] [Accepted: 09/17/2013] [Indexed: 02/05/2023] Open
Abstract
Cathepsins are highly expressed in various human cancers, associated with tumor metastasis. It is superfamily, concluding A, B, C, D, E, F, G, H, L, K, O, S, V, and W family members. As a group of lysosomal proteinases or endopeptidases, each member has a different function, playing different roles in distinct tumorigenic processes such as proliferation, angiogenesis, metastasis, and invasion. Cathepsins belong to a diverse number of enzyme subtypes, including cysteine proteases, serine proteases and aspartic proteases. The contribution of cathepsins to invasion in human cancers is well documented, although the precise mechanisms by which cathepsins exert their effects are still not clear. In the present review, the role of cathepsin family members in cancer is discussed.
Collapse
|
47
|
Galactosialidosis: review and analysis of CTSA gene mutations. Orphanet J Rare Dis 2013; 8:114. [PMID: 23915561 PMCID: PMC3737020 DOI: 10.1186/1750-1172-8-114] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 07/22/2013] [Indexed: 11/10/2022] Open
Abstract
Background Mutations in the CTSA gene, that encodes the protective protein/cathepsin A or PPCA, lead to the secondary deficiency of β-galactosidase (GLB1) and neuraminidase 1 (NEU1), causing the lysosomal storage disorder galactosialidosis (GS). Few clinical cases of GS have been reported in the literature, the majority of them belonging to the juvenile/adult group of patients. Methods The correct nomenclature of mutations for this gene is discussed through the analysis of the three PPCA/CTSA isoforms available in the GenBank database. Phenotype-genotype correlation has been assessed by computational analysis and review of previously reported single amino acid substitutions. Results We report the clinical and mutational analyses of four cases with the rare infantile form of GS. We identified three novel nucleotide changes, two of them resulting in the missense mutations, c.347A>G (p.His116Arg), c.775T>C (p.Cys259Arg), and the third, c.1216C>T, resulting in the p.Gln406* stop codon, a type of mutation identified for the first time in GS. An Italian founder effect of the c.114delG mutation can be suggested according to the origin of the only three patients carrying this mutation reported here and in the literature. Conclusions In early reports mutations nomenclature was selected according to all CTSA isoforms (three different isoforms), thus generating a lot of confusion. In order to assist physicians in the interpretation of detected mutations, we mark the correct nomenclature for CTSA mutations. The complexity of pathology caused by the multifunctions of CTSA, and the very low numbers of mutations (only 23 overall) in relation to the length of the CTSA gene are discussed. In addition, the in silico functional predictions of all reported missense mutations allowed us to closely predict the early infantile, late infantile and juvenile phenotypes, also disclosing different degrees of severity in the juvenile phenotype.
Collapse
|
48
|
Maurice P, Blaise S, Gayral S, Debelle L, Laffargue M, Hornebeck W, Duca L. Elastin fragmentation and atherosclerosis progression: The elastokine concept. Trends Cardiovasc Med 2013; 23:211-21. [DOI: 10.1016/j.tcm.2012.12.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 12/17/2012] [Accepted: 12/18/2012] [Indexed: 01/05/2023]
|
49
|
Dridi L, Seyrantepe V, Fougerat A, Pan X, Bonneil É, Thibault P, Moreau A, Mitchell GA, Heveker N, Cairo CW, Issad T, Hinek A, Pshezhetsky AV. Positive regulation of insulin signaling by neuraminidase 1. Diabetes 2013; 62:2338-46. [PMID: 23520133 PMCID: PMC3712076 DOI: 10.2337/db12-1825] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Neuraminidases (sialidases) catalyze the removal of sialic acid residues from sialylated glycoconjugates. We now report that mammalian neuraminidase 1 (Neu1), in addition to its catabolic function in lysosomes, is transported to the cell surface where it is involved in the regulation of insulin signaling. Insulin binding to its receptor rapidly induces interaction of the receptor with Neu1, which hydrolyzes sialic acid residues in the glycan chains of the receptor and, consequently, induces its activation. Cells from sialidosis patients with a genetic deficiency of Neu1 show impairment of insulin-induced phosphorylation of downstream protein kinase AKT, and treatment of these cells with purified Neu1 restores signaling. Genetically modified mice with ∼10% of the normal Neu1 activity exposed to a high-fat diet develop hyperglycemia and insulin resistance twice as fast as their wild-type counterparts. Together, these studies identify Neu1 as a novel component of the signaling pathways of energy metabolism and glucose uptake.
Collapse
Affiliation(s)
- Larbi Dridi
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montréal, Montréal, Québec, Canada
| | - Volkan Seyrantepe
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montréal, Montréal, Québec, Canada
| | - Anne Fougerat
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montréal, Montréal, Québec, Canada
| | - Xuefang Pan
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montréal, Montréal, Québec, Canada
| | - Éric Bonneil
- Institute of Research in Immunology and Cancer, University of Montréal, Montréal, Québec, Canada
| | - Pierre Thibault
- Institute of Research in Immunology and Cancer, University of Montréal, Montréal, Québec, Canada
| | - Allain Moreau
- Department of Stomatology, Faculty of Dentistry, University of Montréal, Montréal, Québec, Canada
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Sainte-Justine University Hospital Research Center, University of Montréal, Montréal, Québec, Canada
- Department of Biochemistry, University of Montréal, Montréal, Québec, Canada
| | - Grant A. Mitchell
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montréal, Montréal, Québec, Canada
| | - Nikolaus Heveker
- Department of Biochemistry, University of Montréal, Montréal, Québec, Canada
| | - Christopher W. Cairo
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Tarik Issad
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris, France
- INSERM, U1016, Paris, France
| | - Alexander Hinek
- Physiology and Experimental Medicine Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Alexey V. Pshezhetsky
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montréal, Montréal, Québec, Canada
- Department of Biochemistry, University of Montréal, Montréal, Québec, Canada
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montréal, Québec, Canada
- Corresponding author: Alexey V. Pshezhetsky,
| |
Collapse
|
50
|
Inhibition of CatA: an emerging strategy for the treatment of heart failure. Future Med Chem 2013; 5:399-409. [DOI: 10.4155/fmc.13.24] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The lysosomal serine carboxypeptidase CatA has a very important and well-known structural function as well as a, so far, less explored catalytic function. A complete loss of the CatA protein results in the lysosomal storage disease galactosialidosis caused by intralysosomal degradation of β-galactosidase and neuraminidase 1. However, mice with a catalytically inactive CatA enzyme show no signs of this disease. This observation establishes a clear distinction between structural and catalytic functions of the CatA enzyme. Recently, several classes of orally bioavailable synthetic inhibitors of CatA have been identified. Pharmacological studies in rodents indicate a remarkable influence of CatA inhibition on cardiovascular disease progression and identify CatA as a promising novel target for the treatment of heart failure.
Collapse
|