1
|
Liu XH, Liu XT, Wu Y, Li SA, Ren KD, Cheng M, Huang B, Yang Y, Liu PP. Broadening Horizons: Exploring the Cathepsin Family as Therapeutic Targets for Alzheimer's Disease. Aging Dis 2024:AD.2024.0456. [PMID: 39122455 DOI: 10.14336/ad.2024.0456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/02/2024] [Indexed: 08/12/2024] Open
Abstract
Alzheimer's disease (AD) is an intricate neurodegenerative disorder characterized by the accumulation of misfolded proteins, including beta-amyloid (Aβ) and tau, leading to cognitive decline. Despite decades of research, the precise mechanisms underlying its onset and progression remain elusive. Cathepsins are a family of lysosomal enzymes that play vital roles in cellular processes, including protein degradation and regulation of immune responses. Emerging evidence suggests that cathepsins may be involved in AD pathogenesis. Cathepsins can influence the activation of microglia and astrocytes, the resident immune cells in the brain. However, cathepsin dysfunction may lead to the accumulation of misfolded proteins, notably Aβ and tau. In addition, dysregulated cathepsin activity may induce an exaggerated immune response, promoting chronic inflammation and neuronal dysfunction in patients with AD. By unraveling the classification, functions, and roles of cathepsins in AD's pathogenesis, this review sheds light on their intricate involvement in this devastating disease. Targeting cathepsin activity could be a promising and novel approach for mitigating the pathological processes that contribute to AD, providing new avenues for its treatment and prevention.
Collapse
Affiliation(s)
- Xiao-Hui Liu
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiao-Tong Liu
- Clinical Laboratory, the First Hospital of Yongnian District, Yongnian, Hebei, China
| | - Yue Wu
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shu-Ang Li
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kai-Di Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Meng Cheng
- Translational Medical Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bing Huang
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, China
| | - Yang Yang
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pei-Pei Liu
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
2
|
He S, Lv X, He X, Guo J, Pan R, Jin Y, Tian Z, Pan L, Zhang S. Drug Repositioning for Amyloid Transthyretin Amyloidosis by Interactome Network Corrected by Graph Neural Networks and Transcriptome Analysis. Hum Gene Ther 2024; 35:70-79. [PMID: 37756369 DOI: 10.1089/hum.2021.222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023] Open
Abstract
Amyloid transthyretin (ATTR) amyloidosis caused by transthyretin misfolded into amyloid deposits in nerve and heart is a progressive rare disease. The unknown pathogenesis and the lack of therapy make the 5-year survival prognosis extremely poor. Currently available ATTR drugs can only relieve symptoms and slow down progression, but no drug has demonstrated curable effect for this disease. The growing volume of pharmacological data and large-scale genome and transcriptome data bring new opportunities to find potential new ATTR drugs through computational drug repositioning. We collected the ATTR-related in the disease pathogenesis and differentially expressed (DE) genes from five public databases and Gene Expression Omnibus expression profiles, respectively, then screened drug candidates by a corrected protein-protein network analysis of the ATTR-related genes as well as the drug targets from DrugBank database, and then filtered the drug candidates on the basis of gene expression data perturbed by compounds. We collected 139 and 56 ATTR-related genes from five public databases and transcriptome data, respectively, and performed functional enrichment analysis. We screened out 355 drug candidates based on the proximity to ATTR-related genes in the corrected interactome network, refined by graph neural networks. An Inverted Gene Set Enrichment analysis was further applied to estimate the effect of perturbations on ATTR-related and DE genes. High probability drug candidates were discussed. Drug repositioning using systematic computational processes on an interactome network with transcriptome data were performed to screen out several potential new drug candidates for ATTR.
Collapse
Affiliation(s)
- Shan He
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - XiaoYing Lv
- Global Health Drug Discovery Institute, Beijing, China
| | - XinYue He
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - JinJiang Guo
- Global Health Drug Discovery Institute, Beijing, China
| | - RuoKai Pan
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - YuTong Jin
- Global Health Drug Discovery Institute, Beijing, China
| | - Zhuang Tian
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - LuRong Pan
- Global Health Drug Discovery Institute, Beijing, China
| | - ShuYang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Zglejc-Waszak K, Mukherjee K, Korytko A, Lewczuk B, Pomianowski A, Wojtkiewicz J, Banach M, Załęcki M, Nowicka N, Jarosławska J, Kordas B, Wąsowicz K, Juranek JK. Novel insights into the nervous system affected by prolonged hyperglycemia. J Mol Med (Berl) 2023; 101:1015-1028. [PMID: 37462767 PMCID: PMC10400689 DOI: 10.1007/s00109-023-02347-y] [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: 11/25/2022] [Revised: 06/15/2023] [Accepted: 06/28/2023] [Indexed: 08/05/2023]
Abstract
Multiple molecular pathways including the receptor for advanced glycation end-products-diaphanous related formin 1 (RAGE-Diaph1) signaling are known to play a role in diabetic peripheral neuropathy (DPN). Evidence suggests that neuropathological alterations in type 1 diabetic spinal cord may occur at the same time as or following peripheral nerve abnormalities. We demonstrated that DPN was associated with perturbations of RAGE-Diaph1 signaling pathway in peripheral nerve accompanied by widespread spinal cord molecular changes. More than 500 differentially expressed genes (DEGs) belonging to multiple functional pathways were identified in diabetic spinal cord and of those the most enriched was RAGE-Diaph1 related PI3K-Akt pathway. Only seven of spinal cord DEGs overlapped with DEGs from type 1 diabetic sciatic nerve and only a single gene cathepsin E (CTSE) was common for both type 1 and type 2 diabetic mice. In silico analysis suggests that molecular changes in spinal cord may act synergistically with RAGE-Diaph1 signaling axis in the peripheral nerve. KEY MESSAGES: Molecular perturbations in spinal cord may be involved in the progression of diabetic peripheral neuropathy. Diabetic peripheral neuropathy was associated with perturbations of RAGE-Diaph1 signaling pathway in peripheral nerve accompanied by widespread spinal cord molecular changes. In silico analysis revealed that PI3K-Akt signaling axis related to RAGE-Diaph1 was the most enriched biological pathway in diabetic spinal cord. Cathepsin E may be the target molecular hub for intervention against diabetic peripheral neuropathy.
Collapse
Affiliation(s)
- Kamila Zglejc-Waszak
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-085, Olsztyn, Poland.
| | - Konark Mukherjee
- Fralin Biomedical Research Institute at VTC, Virginia Tech, VA, 24016, USA
| | - Agnieszka Korytko
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-085, Olsztyn, Poland
| | - Bogdan Lewczuk
- Department of Histology and Embryology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719, Olsztyn, Poland
| | - Andrzej Pomianowski
- Internal Medicine Department, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719, Olsztyn, Poland
| | - Joanna Wojtkiewicz
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-085, Olsztyn, Poland
| | - Marta Banach
- Department of Neurology, Collegium Medicum, Jagiellonian University, 31-008, Krakow, Poland
| | - Michał Załęcki
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719, Olsztyn, Poland
| | - Natalia Nowicka
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-085, Olsztyn, Poland
| | - Julia Jarosławska
- Department of Biological Functions of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748, Olsztyn, Poland
| | - Bernard Kordas
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-085, Olsztyn, Poland
| | - Krzysztof Wąsowicz
- Department of Pathophysiology, Forensic Veterinary Medicine and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719, Olsztyn, Poland
| | - Judyta K Juranek
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-085, Olsztyn, Poland.
| |
Collapse
|
4
|
Moreira J, Martins H, Saraiva M, Saraiva MJ. TLR2 and 4 signaling pathways are altered in macrophages from V30M TTR mice with down-regulated expression of chemokines. Clin Sci (Lond) 2023; 137:355-366. [PMID: 36852978 DOI: 10.1042/cs20220656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/15/2023] [Accepted: 02/28/2023] [Indexed: 03/01/2023]
Abstract
Hereditary amyloid transthyretin (ATTRv) amyloidosis is a fatal neurodegenerative disorder, first identified in Portugal. The most common transthyretin (TTR) mutation in ATTRv results from an exchange of a methionine for a valine at position 30 (V30M). ATTRv is characterized by the extracellular deposition of aggregates and fibrils of mutant forms of TTR, particularly in the nerves and ganglia of the peripheral nervous system (PNS). This phenotype is often accompanied by the lack of inflammatory infiltrates, despite the importance of macrophages in removal of TTR deposits in ATTRv patients. The mechanisms underlying this impairment of inflammatory responses in ATTRv patients are poorly understood. Here, we show a significant down-regulation in the expression of several chemokines by bone marrow-derived macrophages (BMDM) generated from V30M TTR mice upon stimulation with toll-like receptor 4 (TLR4) and TLR2 agonists. The phosphorylation of the MAP kinase p38, important for TLR4 and TLR2 signaling pathways, was also down-regulated in V30M macrophages, as compared with wild-type (WT) ones. The present study contributes with new insights to unravel the molecular mechanisms underlying the lack of inflammatory immune responses observed in ATTRv patients and may help in the development of new immune therapeutic strategies for the disease.
Collapse
Affiliation(s)
- João Moreira
- Molecular Neurobiology Group, i3S - Instituto de Investigação e Inovação em Saúde, Department of Neurobiology and Neurologic Disorders, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Department of Molecular Biology, Universidade do Porto, 4050-313 Porto, Portugal
| | - Helena Martins
- Molecular Neurobiology Group, i3S - Instituto de Investigação e Inovação em Saúde, Department of Neurobiology and Neurologic Disorders, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Margarida Saraiva
- Immune Regulation Group, i3S - Instituto de Investigação e Inovação em Saúde, Department of Infection, Immunity,and Regeneration, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Maria João Saraiva
- Molecular Neurobiology Group, i3S - Instituto de Investigação e Inovação em Saúde, Department of Neurobiology and Neurologic Disorders, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
5
|
Chandrasekhar G, Rajasekaran R. Theoretical investigations of TTR derived aggregation-prone peptides’ potential to biochemically attenuate the amyloidogenic propensities of V30 M TTR amyloid fibrils. J INDIAN CHEM SOC 2023. [DOI: 10.1016/j.jics.2023.100892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
6
|
Tanner SM, Lorenz RG. FVB/N mouse strain regulatory T cells differ in phenotype and function from the C57BL/6 and BALB/C strains. FASEB Bioadv 2022; 4:648-661. [PMID: 36238362 PMCID: PMC9536134 DOI: 10.1096/fba.2021-00161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/29/2022] [Accepted: 06/16/2022] [Indexed: 11/18/2022] Open
Abstract
Regulatory T cells (Treg) are vital to the maintenance of immune homeostasis. The genetic background of an inbred mouse strain can have a profound effect on the immune response in the animal, including Treg responses. Most Treg studies focus on animals created on the C57BL/6 or BALB/c background. Recent studies have demonstrated a difference in the phenotype and behavior of C57BL/6 and BALB/c Tregs. In this study, we have investigated the function of FVB/N Tregs compared to C57BL/6 and BALB/c. We observed that while FVB/N Tregs appear to suppress normally in a cell contact-dependent system, FVB/N Tregs are less capable of suppressing when regulation depends on the secretion of a soluble factor. FVB/N Tregs produce IL-10; however, TGF-β was not detected in any culture from C57BL/6 or FVB/N. C57BL/6 Foxp3+ Tregs expressed more of the TGF-β-related proteins glycoprotein-A repetitions predominant (GARP) and latency-associated peptide (LAP) on the cell surface than both FVB/N and BALB/c, but C57BL/6 Tregs expressed significantly less Ctse (Cathepsin E) mRNA. Each strain displayed different abilities of thymic Tregs (tTreg) to maintain Foxp3 expression and had a varying generation of induced Tregs (iTregs). In vitro generated FVB/N iTregs expressed significantly less GARP and LAP. These results suggest Tregs of different strains have varying phenotypes and dominant mechanisms of action for the suppression of an immune response. This information should be taken into consideration when Tregs are examined in future studies, particularly for therapeutic purposes in a genetically diverse population.
Collapse
Affiliation(s)
- Scott M. Tanner
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Division of Natural Sciences and EngineeringUniversity of South Carolina UpstateSpartanburgSouth CarolinaUSA
| | - Robin G. Lorenz
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Department of PathologyGenentechSouth San FransiscoCaliforniaUSA
| |
Collapse
|
7
|
Xie Z, Meng J, Kong W, Wu Z, Lan F, Narengaowa, Hayashi Y, Yang Q, Bai Z, Nakanishi H, Qing H, Ni J. Microglial cathepsin E plays a role in neuroinflammation and amyloid β production in Alzheimer's disease. Aging Cell 2022; 21:e13565. [PMID: 35181976 PMCID: PMC8920437 DOI: 10.1111/acel.13565] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/03/2022] [Accepted: 01/25/2022] [Indexed: 02/05/2023] Open
Abstract
Regulation of neuroinflammation and β‐amyloid (Aβ) production are critical factors in the pathogenesis of Alzheimer's disease (AD). Cathepsin E (CatE), an aspartic protease, is widely studied as an inducer of growth arrest and apoptosis in several types of cancer cells. However, the function of CatE in AD is unknown. In this study, we demonstrated that the ablation of CatE in human amyloid precursor protein knock‐in mice, called APPNL−G−F mice, significantly reduced Aβ accumulation, neuroinflammation, and cognitive impairments. Mechanistically, microglial CatE is involved in the secretion of soluble TNF‐related apoptosis‐inducing ligand, which plays an important role in microglia‐mediated NF‐κB‐dependent neuroinflammation and neuronal Aβ production by beta‐site APP cleaving enzyme 1. Furthermore, cannula‐delivered CatE inhibitors improved memory function and reduced Aβ accumulation and neuroinflammation in AD mice. Our findings reveal that CatE as a modulator of microglial activation and neurodegeneration in AD and suggest CatE as a therapeutic target for AD by targeting neuroinflammation and Aβ pathology.
Collapse
Affiliation(s)
- Zhen Xie
- Key Laboratory of Molecular Medicine and Biotherapy Department of Biology School of Life Science Beijing Institute of Technology Beijing China
| | - Jie Meng
- Department of Neurology and State Key Laboratory of Biotherapy Collaborative Innovation Center for Biotherapy West China Hospital Sichuan University Chengdu China
- Department of Aging Science and Pharmacology Faculty of Dental Science Kyushu University Fukuoka Japan
| | - Wei Kong
- Key Laboratory of Molecular Medicine and Biotherapy Department of Biology School of Life Science Beijing Institute of Technology Beijing China
| | - Zhou Wu
- Department of Aging Science and Pharmacology Faculty of Dental Science Kyushu University Fukuoka Japan
| | - Fei Lan
- Key Laboratory of Molecular Medicine and Biotherapy Department of Biology School of Life Science Beijing Institute of Technology Beijing China
| | - Narengaowa
- Key Laboratory of Molecular Medicine and Biotherapy Department of Biology School of Life Science Beijing Institute of Technology Beijing China
| | - Yoshinori Hayashi
- Department of Physiology Nihon University School of Dentistry Tokyo Japan
| | - Qinghu Yang
- Research Center for Resource Peptide Drugs Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources Yan’an University Yan’an China
| | - Zhantao Bai
- Research Center for Resource Peptide Drugs Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources Yan’an University Yan’an China
| | - Hiroshi Nakanishi
- Department of Pharmacology Faculty of Pharmacy Yasuda Women’s University Hiroshima Japan
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy Department of Biology School of Life Science Beijing Institute of Technology Beijing China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy Department of Biology School of Life Science Beijing Institute of Technology Beijing China
- Department of Aging Science and Pharmacology Faculty of Dental Science Kyushu University Fukuoka Japan
| |
Collapse
|
8
|
Wu Y, Zhou W, Yang Z, Li J, Jin Y. miR-185-5p Represses Cells Growth and Metastasis of Osteosarcoma via Targeting Cathepsin E. Int J Toxicol 2022; 41:115-125. [PMID: 35213250 DOI: 10.1177/10915818211069270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Osteosarcoma (OS) is a malignant bone tumor characterized by poor prognosis due to its regional invasion and early metastasis. In this study, we aimed to find the role and the underlying mechanism of Cathepsin E (CTSE) in OS growth and metastasis. We found CTSE is upregulated in metastatic OS, rather than in the primary lesion, as confirmed by RT-qPCR and western blot analysis of clinical OS samples. Furthermore, both in vitro and in vivo experiments illustrated that CTSE promoted both growth and metastasis of OS cells, partially mediated through the modulation of Epithelial-Mesenchymal Transition (EMT). Bioinformatics analysis predicted that miR-185-5p downregulates CTSE via directly binding to the 3'UTR of CTSE, which was verified by luciferase reporter assay and rescue assays. This study reported for the first time that CTSE is a potential biomarker in OS tumorigenesis and metastasis, providing a promising therapeutic target for OS treatment.
Collapse
Affiliation(s)
- Yue Wu
- Department of Orthopaedics, Beijing United Family Healthcare, Beijing, China
| | - Weili Zhou
- Joint Surgery Department of Orthopaedics, The Third Hospital of Changsha, Changsha, China
| | - Zhijun Yang
- Trauma Department of Orthopaedics, The First Affiliated Hospital of South China University, Hengyang, China
| | - Jinping Li
- Joint Surgery Department of Orthopaedics, Changsha Central Hospital, Changsha, China
| | - Yi Jin
- Joint Surgery Department of Orthopaedics, Changsha Central Hospital, Changsha, China
| |
Collapse
|
9
|
In Vitro and In Vivo Effects of SerpinA1 on the Modulation of Transthyretin Proteolysis. Int J Mol Sci 2021; 22:ijms22179488. [PMID: 34502397 PMCID: PMC8430710 DOI: 10.3390/ijms22179488] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 12/14/2022] Open
Abstract
Transthyretin (TTR) proteolysis has been recognized as a complementary mechanism contributing to transthyretin-related amyloidosis (ATTR amyloidosis). Accordingly, amyloid deposits can be composed mainly of full-length TTR or contain a mixture of both cleaved and full-length TTR, particularly in the heart. The fragmentation pattern at Lys48 suggests the involvement of a serine protease, such as plasmin. The most common TTR variant, TTR V30M, is susceptible to plasmin-mediated proteolysis, and the presence of TTR fragments facilitates TTR amyloidogenesis. Recent studies revealed that the serine protease inhibitor, SerpinA1, was differentially expressed in hepatocyte-like cells (HLCs) from ATTR patients. In this work, we evaluated the effects of SerpinA1 on in vitro and in vivo modulation of TTR V30M proteolysis, aggregation, and deposition. We found that plasmin-mediated TTR proteolysis and aggregation are partially inhibited by SerpinA1. Furthermore, in vivo downregulation of SerpinA1 increased TTR levels in mice plasma and deposition in the cardiac tissue of older animals. The presence of TTR fragments was observed in the heart of young and old mice but not in other tissues following SerpinA1 knockdown. Increased proteolytic activity, particularly plasmin activity, was detected in mice plasmas. Overall, our results indicate that SerpinA1 modulates TTR proteolysis and aggregation in vitro and in vivo.
Collapse
|
10
|
Hein S, Furkel J, Knoll M, Aus dem Siepen F, Schönland S, Hegenbart U, Katus HA, Kristen AV, Konstandin MH. Impaired in vitro growth response of plasma-treated cardiomyocytes predicts poor outcome in patients with transthyretin amyloidosis. Clin Res Cardiol 2021; 110:579-590. [PMID: 33481097 PMCID: PMC8055573 DOI: 10.1007/s00392-020-01801-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/23/2020] [Indexed: 12/26/2022]
Abstract
OBJECTIVES Direct toxic effects of transthyretin amyloid in patient plasma upon cardiomyocytes are discussed. However, no data regarding the relevance of this putative effect for clinical outcome are available. In this monocentric prospective study, we analyzed cellular hypertrophy after phenylephrine stimulation in vitro in the presence of patient plasma and correlated the cellular growth response with phenotype and prognosis. METHODS AND RESULTS Progress in automated microscopy and image analysis allows high-throughput analysis of cell morphology. Using the InCell microscopy system, changes in cardiomyocyte's size after treatment with patient plasma from 89 patients suffering from transthyretin amyloidosis and 16 controls were quantified. For this purpose, we propose a novel metric that we named Hypertrophic Index, defined as difference in cell size after phenylephrine stimulation normalized to the unstimulated cell size. Its prognostic value was assessed for multiple endpoints (HTX: death/heart transplantation; DMP: cardiac decompensation; MACE: combined) using Cox proportional hazard models. Cells treated with plasma from healthy controls and hereditary transthyretin amyloidosis with polyneuropathy showed an increase in Hypertrophic Index after phenylephrine stimulation, whereas stimulation after treatment with hereditary cardiac amyloidosis or wild-type transthyretin patient plasma showed a significantly attenuated response. Hypertrophic Index was associated in univariate analyses with HTX (hazard ratio (HR) high vs low: 0.12 [0.02-0.58], p = 0.004), DMP: (HR 0.26 [0.11-0.62], p = 0.003) and MACE (HR 0.24 [0.11-0.55], p < 0.001). Its prognostic value was independent of established risk factors, cardiac TroponinT or N-terminal prohormone brain natriuretic peptide (NTproBNP). CONCLUSIONS Attenuated cardiomyocyte growth response after stimulation with patient plasma in vitro is an independent risk factor for adverse cardiac events in ATTR patients.
Collapse
Affiliation(s)
- Selina Hein
- Department of Cardiology, Angiology and Pulmonology, University Hospital Heidelberg, INF 410, 69120, Heidelberg, Germany.
| | - Jennifer Furkel
- Department of Cardiology, Angiology and Pulmonology, University Hospital Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Maximilian Knoll
- Department of Radiation Oncology, Heidelberg Ion-Beam Therapy Center (HIT), German Cancer Research Center, Heidelberg University Hospital (UKHD), Heidelberg, Germany
| | - Fabian Aus dem Siepen
- Department of Cardiology, Angiology and Pulmonology, University Hospital Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Stefan Schönland
- Department of Hematology, Oncology and Rheumatology, Heidelberg University, Heidelberg, Germany
| | - Ute Hegenbart
- Department of Hematology, Oncology and Rheumatology, Heidelberg University, Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, Angiology and Pulmonology, University Hospital Heidelberg, INF 410, 69120, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Arnt V Kristen
- Department of Cardiology, Angiology and Pulmonology, University Hospital Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Mathias H Konstandin
- Department of Cardiology, Angiology and Pulmonology, University Hospital Heidelberg, INF 410, 69120, Heidelberg, Germany.
- DZHK (German Center for Cardiovascular Research), Site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
11
|
Niemietz C, Bezerra F, Almeida MR, Guo S, Monia BP, Saraiva MJ, Schütz P, Schmidt HHJ, Zibert A. SERPINA1 modulates expression of amyloidogenic transthyretin. Exp Cell Res 2020; 395:112217. [PMID: 32768500 DOI: 10.1016/j.yexcr.2020.112217] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 07/31/2020] [Accepted: 08/01/2020] [Indexed: 12/14/2022]
Abstract
Hereditary transthyretin amyloidosis (ATTR) is caused by amyloid deposition of misfolded transthyretin (TTR) in various tissues. Recently, reduction of circulating serum TTR, achieved via silencing oligonucleotides, was introduced as therapy of ATTR amyloidosis. We explored the impact of Serpin Family A Member 1 (SERPINA1) on TTR mRNA and protein expression. Oncostatin M (OSM) induced SERPINA1 in hepatoma cells and mice, while concomitantly TTR expression was significantly reduced. SERPINA1 knockdown resulted in specific elevated TTR expression in hepatoma cells; however other genes belonging to the group of acute phase proteins were unaffected. In mice, serum TTR was elevated after mSERPINA1 knockdown throughout antisense treatment. Following SERPINA1 knockdown, TTR deposition in several tissues, including dorsal root ganglia and intestine, was found to be increased, however numbers did not exceed significance levels. The data suggest that SERPINA1 is a co-factor of TTR expression. Our findings provide novel insight in the regulation of TTR and reveal a role of SERPINA1 in the pathogenesis of ATTR amyloidosis.
Collapse
Affiliation(s)
- Christoph Niemietz
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Filipa Bezerra
- i3S - Instituto de Investigação e Inovação em Saúde, IBMC - Instituto de Biologia Molecular e Celular,and ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria Rosário Almeida
- i3S - Instituto de Investigação e Inovação em Saúde, IBMC - Instituto de Biologia Molecular e Celular,and ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | | | | | - Maria João Saraiva
- i3S - Instituto de Investigação e Inovação em Saúde, IBMC - Instituto de Biologia Molecular e Celular,and ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Paula Schütz
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Hartmut H-J Schmidt
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Andree Zibert
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany.
| |
Collapse
|
12
|
Srinivasan E, Natarajan N, Rajasekaran R. TTRMDB: A database for structural and functional analysis on the impact of SNPs over transthyretin (TTR) using bioinformatic tools. Comput Biol Chem 2020; 87:107290. [PMID: 32512488 DOI: 10.1016/j.compbiolchem.2020.107290] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/14/2020] [Accepted: 05/21/2020] [Indexed: 12/21/2022]
Abstract
Hereditary Transthyretin-associated amyloidosis (ATTR) is an autosomal dominant protein-folding disorder with adult-onset caused by mutation of transthyretin (TTR). TTR is characterized by extracellular deposition of amyloid, leading to loss of autonomy and finally, death. More than 100 distinct mutations in TTR gene have been reported from variable age of onset, clinical expression and penetrance data. Besides, the cure for the disease remains still obscure. Further, the prioritizing of mutations concerning the characteristic features governing the stability and pathogenicity of TTR mutant proteins remains unanswered, to date and thus, a complex state of study for researchers. Herein, we provide a full report encompassing the effects of every reported mutant model of TTR protein about the stability, functionality and pathogenicity using various computational tools. In addition, the results obtained from our study were used to create TTRMDB (Transthyretin mutant database), which could be easy access to researchers at http://vit.ac.in/ttrmdb.
Collapse
Affiliation(s)
- E Srinivasan
- Bioinformatics Lab, Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology (Deemed to be University), Vellore 632014, Tamil Nadu, India
| | - Nandhini Natarajan
- Bioinformatics Lab, Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology (Deemed to be University), Vellore 632014, Tamil Nadu, India
| | - R Rajasekaran
- Bioinformatics Lab, Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology (Deemed to be University), Vellore 632014, Tamil Nadu, India.
| |
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: 21] [Impact Index Per Article: 5.3] [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
|
Dias A, Santos D, Coelho T, Alves-Ferreira M, Sequeiros J, Alonso I, Sousa A, Lemos C. C1QA and C1QC modify age-at-onset in familial amyloid polyneuropathy patients. Ann Clin Transl Neurol 2019; 6:748-754. [PMID: 31019999 PMCID: PMC6469251 DOI: 10.1002/acn3.748] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 11/24/2022] Open
Abstract
Objectives Transthyretin (TTR) familial amyloid polyneuropathy (FAP) (OMIM 176300) shows a variable age‐at‐onset (AO), including within families. We hypothesized that variants in C1QA and C1QC genes, might also act as genetic modifiers of AO in TTR‐FAP Val30Met Portuguese patients. Methods We analyzed DNA samples of 267 patients (117 families). To search for variants, all exons and flanking regions were genotyped by automated sequencing. We used generalized estimating equations (GEEs) to take into account the non‐independency of AO among relatives. Intensive in silico analyses were performed, using various software to assess miRNAs target sites, splicing sites, transcription factor binding sites alterations, and gene–gene interactions. Results Two variants for C1QA gene, GA genotype of rs201693493 (P < 0.001) and CT genotype of rs149050968 (P < 0.001), were significantly associated with later AO. In silico analysis demonstrated, that rs201693493 may alter splicing activity. Regarding C1QC, we found three statistically significant results: GA genotype of rs2935537 (P = 0.003), GA genotype of rs201241346 (P < 0.001) and GA genotype of rs200952686 (P < 0.001). The first two were associated with earlier AO, whereas the third was associated with later‐onset. Interpretation C1QA was associated with later onset, whereas C1QC may have a double role: variants may confer earlier or later AO. As found in a study in Cyprus, we confirmed the role of complement C1Q genes (and thus of inflammation) as modulator of AO in Portuguese patients with TTR‐FAP Val30Met.
Collapse
Affiliation(s)
- Andreia Dias
- i3S Instituto de Investigação e Inovação em Saúde Universidade do Porto Porto Portugal.,UnIGENe IBMC - Instituto de Biologia Molecular e Celular Universidade do Porto Porto Portugal.,FCUP Faculdade de Ciências da Universidade do Porto Porto Portugal
| | - Diana Santos
- i3S Instituto de Investigação e Inovação em Saúde Universidade do Porto Porto Portugal.,UnIGENe IBMC - Instituto de Biologia Molecular e Celular Universidade do Porto Porto Portugal.,ICBAS Instituto Ciências Biomédicas Abel Salazar Universidade do Porto Porto Portugal
| | - Teresa Coelho
- UCA Unidade Corino de Andrade Centro Hospitalar Universitário do Porto (CHUP) Porto Portugal
| | - Miguel Alves-Ferreira
- i3S Instituto de Investigação e Inovação em Saúde Universidade do Porto Porto Portugal.,UnIGENe IBMC - Instituto de Biologia Molecular e Celular Universidade do Porto Porto Portugal.,ICBAS Instituto Ciências Biomédicas Abel Salazar Universidade do Porto Porto Portugal
| | - Jorge Sequeiros
- i3S Instituto de Investigação e Inovação em Saúde Universidade do Porto Porto Portugal.,UnIGENe IBMC - Instituto de Biologia Molecular e Celular Universidade do Porto Porto Portugal.,ICBAS Instituto Ciências Biomédicas Abel Salazar Universidade do Porto Porto Portugal
| | - Isabel Alonso
- i3S Instituto de Investigação e Inovação em Saúde Universidade do Porto Porto Portugal.,UnIGENe IBMC - Instituto de Biologia Molecular e Celular Universidade do Porto Porto Portugal.,ICBAS Instituto Ciências Biomédicas Abel Salazar Universidade do Porto Porto Portugal
| | - Alda Sousa
- i3S Instituto de Investigação e Inovação em Saúde Universidade do Porto Porto Portugal.,UnIGENe IBMC - Instituto de Biologia Molecular e Celular Universidade do Porto Porto Portugal.,ICBAS Instituto Ciências Biomédicas Abel Salazar Universidade do Porto Porto Portugal
| | - Carolina Lemos
- i3S Instituto de Investigação e Inovação em Saúde Universidade do Porto Porto Portugal.,UnIGENe IBMC - Instituto de Biologia Molecular e Celular Universidade do Porto Porto Portugal.,ICBAS Instituto Ciências Biomédicas Abel Salazar Universidade do Porto Porto Portugal
| |
Collapse
|
15
|
Moreira J, Saraiva M, Saraiva MJ. Downregulated Cathepsin E expression in bone marrow-derived macrophages from the pre-clinical familial amyloid polyneuropathy model. Amyloid 2019; 26:63-64. [PMID: 31343320 DOI: 10.1080/13506129.2019.1585342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- João Moreira
- a I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto , Portugal.,b IBMC - Instituto de Biologia Molecular e Celular, Grupo Neurobiologia Molecular, Universidade do Porto , Porto , Portugal
| | - Margarida Saraiva
- a I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto , Portugal.,c IBMC - Instituto de Biologia Molecular e Celular, Grupo Regulação Imune, Universidade do Porto , Porto , Portugal
| | - Maria João Saraiva
- a I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto , Portugal.,b IBMC - Instituto de Biologia Molecular e Celular, Grupo Neurobiologia Molecular, Universidade do Porto , Porto , Portugal
| |
Collapse
|
16
|
Sparse common component analysis for multiple high-dimensional datasets via noncentered principal component analysis. Stat Pap (Berl) 2018. [DOI: 10.1007/s00362-018-1045-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
17
|
Niemietz C, Fleischhauer L, Sandfort V, Guttmann S, Zibert A, Schmidt HHJ. Hepatocyte-like cells reveal novel role of SerpinA1 in transthyretin amyloidosis. J Cell Sci 2018; 131:jcs.219824. [DOI: 10.1242/jcs.219824] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 09/18/2018] [Indexed: 01/07/2023] Open
Abstract
Transthyretin (TTR)-related familial amyloid polyneuropathy (ATTR) results from aggregation and extracellular disposition of misfolded TTR variants. Growing evidence suggests the importance of hepatic chaperones for modulation of pathogenesis. We took advantage of iPSC-derived hepatocyte-like cells (HLCs) derived from ATTR patients (ATTR-HLCs) to compare chaperone gene expression to healthy individuals (H-HLCs). From the set of genes analyzed, chaperones that are predominantly located extracellularly were differently expressed. Expression of the chaperones showed a high correlation with TTR in both ATTR-HLCs and H-HLCs. In contrast, after TTR knockdown, the correlation was mainly affected in ATTR-HLCs suggesting that variant TTR expression triggers abberant chaperone expression. Serpin peptidase inhibitor clade A member 1 (SERPINA1/alpha-1 antitrypsin) was the only extracellular chaperone that was markedly upregulated after TTR knockdown in ATTR-HLCs. Co-immunoprecipitation revealed that SerpinA1 physically interacts with TTR. In vitro assays indicated that SerpinA1 can interfere with TTR aggregation. Taken together, our results suggest that extracellular chaperones play a crucial role in ATTR pathogenesis, in particular SerpinA1, which may affect amyloid formation.
Collapse
Affiliation(s)
- Christoph Niemietz
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Lutz Fleischhauer
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
- Present address: Fakultät für angewandte Naturwissenschaften und Mechatronik, Hochschule München, München, Germany
| | - Vanessa Sandfort
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Sarah Guttmann
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Andree Zibert
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Hartmut H.-J. Schmidt
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| |
Collapse
|