1
|
Ma S, Liu B, Du H, Yang F, Han J, Huang X, Zhang M, Ji S, Jiang M. RNAi targeting LMAN1-MCFD2 complex promotes anticoagulation in mice. J Thromb Thrombolysis 2024:10.1007/s11239-024-03034-6. [PMID: 39222205 DOI: 10.1007/s11239-024-03034-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/11/2024] [Indexed: 09/04/2024]
Abstract
Combined deficiency of coagulation factor V (FV) and factor VIII (FVIII) is a rare bleeding disease caused by variants in either lectin mannose binding 1 (LMAN1) or multiple coagulation factor deficiency 2 (MCFD2) gene. Reducing the level of FVIII by inhibiting the LMAN1-MCFD2 complex may become a new anticoagulant approach. We aimed to find a new therapeutic option for anticoagulation by RNA interference (RNAi) targeting LMAN1 and MCFD2. siRNA sequences with cross-homology between mice and humans were designed based on LMAN1 or MCFD2 transcripts in NCBI and were screened with the Dual-Luciferase reporter assay. The optimal siRNAs were chemically modified and conjugated with three N-acetylgalactosamine molecules (GalNAc-siRNA), promoting their targeted delivery to the liver. The expression of LMAN1 and MCFD2 in cell lines or mice was examined by RT-qPCR and western blotting. For the mice administered with siRNA, we assessed their coagulation function by measuring APTT and the activity of FVIII factor. After administration, siRNAs GalNAc-LMAN1 and GalNAc-MCFD2 demonstrated effective and persistent LMAN1 and MCFD2 inhibition. 7 days after injection of 3mg/kg GalNAc-LMAN1, the LMAN1 mRNA levels reduced to 19.97% ± 3.78%. MCFD2 mRNA levels reduced to 32.22% ± 13.14% with injection of 3mg/kg GalNAc-MCFD2. After repeated administration, APTT was prolonged and the FVIII activity was remarkably decreased. The tail bleeding test of mice showed that the amount of bleeding in the treated group did not significantly increase compared with the control group. Our study confirms that therapy with RNAi targeting LMAN1-MCFD2 complex is effective and can be considered a viable option for anticoagulation drugs. However, the benefits and potential risk of bleeding in thrombophilic mice model needs to be evaluated.
Collapse
Affiliation(s)
- Siqian Ma
- Hematology Department, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215021, China
| | - Boyan Liu
- Hematology Department, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215021, China
| | - Hong Du
- Suzhou Genephama Co., Ltd, , Suzhou, 215123, China
| | - Fei Yang
- National Clinical Medical Research Center of Blood Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215007, China
- Key Laboratory of Thrombosis and Hemostasis of National Health Commission of People's Republic of China, Suzhou, 215006, China
| | - Jingjing Han
- National Clinical Medical Research Center of Blood Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215007, China
- Key Laboratory of Thrombosis and Hemostasis of National Health Commission of People's Republic of China, Suzhou, 215006, China
| | - Xinqi Huang
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, 215123, China
| | - Minyang Zhang
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, 215123, China
| | - Shundong Ji
- National Clinical Medical Research Center of Blood Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215007, China
- Key Laboratory of Thrombosis and Hemostasis of National Health Commission of People's Republic of China, Suzhou, 215006, China
| | - Miao Jiang
- Hematology Department, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215021, China.
- National Clinical Medical Research Center of Blood Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215007, China.
- Key Laboratory of Thrombosis and Hemostasis of National Health Commission of People's Republic of China, Suzhou, 215006, China.
| |
Collapse
|
2
|
Yakovleva E, Zhang B. Clinical, Laboratory, Molecular, and Reproductive Aspects of Combined Deficiency of Factors V and VIII. Semin Thromb Hemost 2024. [PMID: 39209292 DOI: 10.1055/s-0044-1789019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Congenital combined deficiency of factor V (FV) and factor VIII (FVIII; F5F8D, OMIM 227300) is a rare hereditary coagulopathy and accounts for approximately 3% of cases of rare coagulation disorders. The prevalence of this disease in the general population is estimated to be 1:1,000,000 and is significantly higher in regions where consanguineous marriages are permitted, such as the Mideast and South Asia. The disease has an autosomal recessive mode of inheritance and therefore occurs with an equal incidence among males and females. Heterozygous mutation carriers usually do not have clinical manifestations. The molecular basis of this disease differs from that of stand-alone congenital deficiencies of FVIII and FV. F5F8D is caused by mutations in either LMAN1 or MCFD2, which encode components of a cargo receptor complex for endoplasmic reticulum to Golgi transport of FV and FVIII, leading to defects in an intracellular transport pathway shared by these two coagulation factors. Congenital combined deficiency of FV and FVIII is characterized by decreased activities of both FV and FVIII in plasma, usually to 5 to 30% of normal. Clinical manifestations in most cases are represented by mild or moderate hemorrhagic syndrome. The simultaneous decreases of two coagulation factors present complications in the diagnosis and management of the disease. In female patients, the disease requires a special approach for family planning, pregnancy management, and parturition. This review summarizes recent progress in clinical, laboratory, and molecular understanding of this disorder.
Collapse
Affiliation(s)
- Elena Yakovleva
- Clinical and Diagnostic Department of Hematology and Hemostasis Disorders, National Medical Research Center for Hematology, Novy Zykovsky, Russia
| | - Bin Zhang
- Genomic Medicine Institute, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| |
Collapse
|
3
|
Yang W, Mu H, Zhou F, Wang J, Bi H, Zhou Z. Successful hip arthroplasty in a patient with combined deficiency of factor V and factor FVIII. Haemophilia 2024; 30:558-560. [PMID: 38146236 DOI: 10.1111/hae.14925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/10/2023] [Accepted: 12/11/2023] [Indexed: 12/27/2023]
Affiliation(s)
- Wen Yang
- Department of Hematology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hongli Mu
- Department of Hematology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Fan Zhou
- Department of Hematology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jia Wang
- Department of Hematology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hui Bi
- Department of Hematology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zeping Zhou
- Department of Hematology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
4
|
Al Khudari R, Batesh D, Habash R, Hamdn O. Hemophilia A and factor V deficiency in a girl with Turner syndrome: a case report. J Med Case Rep 2023; 17:480. [PMID: 37978530 PMCID: PMC10656841 DOI: 10.1186/s13256-023-04215-2] [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: 03/13/2023] [Accepted: 10/13/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Hemophilia is an X-linked, recessive inherited disease caused by a defect or deficiency of one of the coagulation factors (VIII or IX). It is considered a rare disease in females. One of the reasons that hemophilia affects females is Turner syndrome. Hemophilia with Turner syndrome is a very rare case, but the combination of Turner syndrome, hemophilia, and factor V deficiency is an isolated case that has never been recorded in the medical literature. CASE PRESENTATION In our case, a 5-year-old Syrian girl presented with hemorrhage of gum, epistaxis, and short stature. The lab tests showed: prolonged activated partial thromboplastin time and prothrombin time with deficiency of factor V (1%) and factor VIII (1%). We diagnosed hemophilia A with factor V deficiency. In addition to short stature, the patient was noted to have spaced nipples and winged neck. We performed karyotyping that showed deletion of one X chromosome (45X0), Turner syndrome. There is no family history of hemophilia or any other genetic disease. CONCLUSIONS In females affected with hemophilia, karyotyping should be performed. It is very important not to exclude the possibility of a combination of deficiency of more than one clotting factor, and to note that deficiency of more than one factor does not necessarily increase the severity of bleeding compared with deficiency of a single factor.
Collapse
Affiliation(s)
- Rawan Al Khudari
- Department of Paediatrics, Faculty of Medicine, Damascus University, Damascus, Syria.
| | - Duaa Batesh
- Faculty of Medicine, Damascus University, Damascus, Syria
| | - Roaa Habash
- Faculty of Medicine, Damascus University, Damascus, Syria
| | - Othman Hamdn
- Division of Hematology and Oncology, Department of Paediatrics, Faculty of Medicine, Damascus University, Damascus, Syria
| |
Collapse
|
5
|
Zhang Y, Srivastava V, Zhang B. Mammalian cargo receptors for endoplasmic reticulum-to-Golgi transport: mechanisms and interactions. Biochem Soc Trans 2023:BST20220713. [PMID: 37334845 DOI: 10.1042/bst20220713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/22/2023] [Accepted: 05/30/2023] [Indexed: 06/21/2023]
Abstract
Proteins that are destined to enter the secretory pathway are synthesized on the rough endoplasmic reticulum (ER) and then translocated into the ER lumen, where they undergo posttranslational modifications, folding, and assembly. After passing a quality control system, the cargo proteins are packaged into coat protein complex II (COPII) vesicles to exit the ER. In metazoans, most COPII subunits have multiple paralogs, enabling COPII vesicles the flexibility to transport a diverse range of cargo. The cytoplasmic domains of transmembrane proteins can interact with SEC24 subunits of COPII to enter the ER exit sites. Some transmembrane proteins may also act as cargo receptors that bind soluble secretory proteins within the ER lumen, enabling them to enter COPII vesicles. The cytoplasmic domains of cargo receptors also contain coat protein complex I binding motifs that allow for their cycling back to the ER after unloading their cargo in the ER-Golgi intermediate compartment and cis-Golgi. Once unloaded, the soluble cargo proteins continue maturation through the Golgi before reaching their final destinations. This review provides an overview of receptor-mediated transport of secretory proteins from the ER to the Golgi, with a focus on the current understanding of two mammalian cargo receptors: the LMAN1-MCFD2 complex and SURF4, and their roles in human health and disease.
Collapse
Affiliation(s)
- Yuan Zhang
- Genomic Medicine Institute, Lerner Research Institute of Cleveland Clinic, Cleveland, OH, U.S.A
| | - Vishal Srivastava
- Genomic Medicine Institute, Lerner Research Institute of Cleveland Clinic, Cleveland, OH, U.S.A
| | - Bin Zhang
- Genomic Medicine Institute, Lerner Research Institute of Cleveland Clinic, Cleveland, OH, U.S.A
| |
Collapse
|
6
|
Miller MH, Swaby LG, Vailoces VS, LaFratta M, Zhang Y, Zhu X, Hitchcock DJ, Jewett TJ, Zhang B, Tigno-Aranjuez JT. LMAN1 is a receptor for house dust mite allergens. Cell Rep 2023; 42:112208. [PMID: 36870056 PMCID: PMC10105285 DOI: 10.1016/j.celrep.2023.112208] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 12/01/2022] [Accepted: 02/15/2023] [Indexed: 03/05/2023] Open
Abstract
Development of therapies with the potential to change the allergic asthmatic disease course will require the discovery of targets that play a central role during the initiation of an allergic response, such as those involved in the process of allergen recognition. We use a receptor glycocapture technique to screen for house dust mite (HDM) receptors and identify LMAN1 as a candidate. We verify the ability of LMAN1 to directly bind HDM allergens and demonstrate that LMAN1 is expressed on the surface of dendritic cells (DCs) and airway epithelial cells (AECs) in vivo. Overexpression of LMAN1 downregulates NF-κB signaling in response to inflammatory cytokines or HDM. HDM promotes binding of LMAN1 to the FcRγ and recruitment of SHP1. Last, peripheral DCs of asthmatic individuals show a significant reduction in the expression of LMAN1 compared with healthy controls. These findings have potential implications for the development of therapeutic interventions for atopic disease.
Collapse
Affiliation(s)
- Madelyn H Miller
- Biotechnology and Immunology Research, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Lindsay G Swaby
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Vanessa S Vailoces
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Maggie LaFratta
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Yuan Zhang
- Genomic Medicine Institute, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Xiang Zhu
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Dorilyn J Hitchcock
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Travis J Jewett
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Bin Zhang
- Genomic Medicine Institute, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Justine T Tigno-Aranjuez
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA.
| |
Collapse
|
7
|
Bioinformatics analysis of LMAN1 expression, clinical characteristics, and its effects on cell proliferation and invasion in glioma. Brain Res 2022; 1789:147952. [PMID: 35623391 DOI: 10.1016/j.brainres.2022.147952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/11/2022] [Accepted: 05/21/2022] [Indexed: 11/20/2022]
Abstract
Glioma is the most common primary central nervous system malignant tumor with high heterogeneity and poor prognosis. So far, the complex pathological process of glioma has not been fully elucidated, and there is a lack of effective biomarkers for the diagnosis and molecular targeted therapy of glioma. Using bioinformatics methods, 77 upregulated and 89 downregulated differentially expressed genes (DEGs) were detected by intersection analysis in different gene expression datasets of glioma cases from public databases. Then, GO and KEGG pathway analysis revealed that the biological functions of these upregulated DEGs were mainly focused on immune response, and the signaling pathways were mainly enriched in integrin family cell surface interactions. The overexpression of the LMAN1 gene of interest was then confirmed using the TCGA dataset and further verified by qRT-PCR in 29 clinical samples and 5 glioma cell lines. Furthermore, high expression of LMAN1 was found to be associated with higher WHO grade, IDH status, and 1p/19q co-deletion. Survival analysis showed that high expression of LMAN1 was associated with poor prognosis in glioma. Gene set enrichment analysis (GSEA) indicated that many cancer-related pathways were associated with LMAN1-high phenotype. Protein-protein interaction (PPI) analysis revealed significant interaction between LMAN1 and MCFD2, F8, and TMED10. Finally, cell experiments showed that LMAN1 knockdown significantly inhibited the proliferation, migration and invasion and promoted apoptosis in glioma cells. This study highlighted the malignant role of LMAN1 in gliomas and provided a potentially valuable biomarker for prognosis evaluation and molecular targeted therapy of glioma.
Collapse
|
8
|
Abstract
INTRODUCTION Hemophilia A (HA) or B (HB) is an X-linked recessive disorder caused by a defect in the factor VIII (FVIII) or factor IX (FIX) gene which leads to the dysfunction of blood coagulation. Protein replacement therapy (PRT) uses recombinant proteins and plasma-derived products, which incurs high cost and inconvenience requiring routine intravenous infusions and life-time treatment. Understanding of detailed molecular mechanisms on FVIII gene function could provide innovative solutions to amend this disorder. In recent decades, gene therapeutics have advanced rapidly and a one-time cure solution has been proposed. AREAS COVERED This review summarizes current understanding of molecular pathways involved in blood coagulation, with emphasis on FVIII's functional role. The existing knowledge and challenges on FVIII gene expression, from transcription, translation, post-translational modification including glycosylation to protein processing and secretion, and co-factor interactions are deciphered and potential molecular interventions discussed. EXPERT OPINION This article reviews the potential treatment targets for HA and HB, including antibodies, small molecules and gene therapeutics, based on molecular mechanisms of FVIII biosynthesis, and further, assessing the pros and cons of these various treatment strategies. Understanding detailed FVIII protein synthesis and secretory pathways could provide exciting opportunities in identifying novel therapeutics to ameliorate hemophilia state.
Collapse
Affiliation(s)
- Jie Gong
- School of Medicine, University of Electronic Science and Technology of China, Sichuan, China
| | - Hao-Lin Wang
- School of Medicine, University of Electronic Science and Technology of China, Sichuan, China
| | - Lung-Ji Chang
- School of Medicine, University of Electronic Science and Technology of China, Sichuan, China.,Geno-Immune Medical Institute, Shenzhen, China
| |
Collapse
|
9
|
LMAN1-MCFD2 complex is a cargo receptor for the ER-Golgi transport of α1-antitrypsin. Biochem J 2022; 479:839-855. [PMID: 35322856 PMCID: PMC9022998 DOI: 10.1042/bcj20220055] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 11/17/2022]
Abstract
α1-antitrypsin (AAT) is a serine protease inhibitor synthesized in hepatocytes and protects the lung from damage by neutrophil elastase. AAT gene mutations result in AAT deficiency (AATD), which leads to lung and liver diseases. The AAT Z variant forms polymer within the endoplasmic reticulum (ER) of hepatocytes and results in reduction of AAT secretion and severe disease. Previous studies demonstrated a secretion defect of AAT in LMAN1 deficient cells, and mild decreases in AAT levels in male LMAN1 and MCFD2 deficient mice. LMAN1 is a transmembrane lectin that forms a complex with a small soluble protein MCFD2. The LMAN1-MCFD2 protein complex cycles between the ER and the Golgi. Here we report that LMAN1 and MCFD2 knockout (KO) HepG2 and HEK293T cells display reduced AAT secretion and elevated intracellular AAT levels due to a delayed ER-to-Golgi transport of AAT. Secretion defects in KO cells were rescued by wild-type LMAN1 or MCFD2, but not by mutant proteins. Elimination of the second glycosylation site of AAT abolished LMAN1 dependent secretion. Co-immunoprecipitation experiment in MCFD2 KO cells suggested that AAT interaction with LMAN1 is independent of MCFD2. Furthermore, our results suggest that secretion of the Z variant, both monomers and polymers, is also LMAN1-dependent. Results provide direct evidence supporting that the LMAN1-MCFD2 complex is a cargo receptor for the ER-to-Golgi transport of AAT and that interactions of LMAN1 with an N-glycan of AAT is critical for this process. These results have implications in production of recombinant AAT and in developing treatments for AATD patients.
Collapse
|
10
|
Abstract
Constitutive vesicle trafficking is the default pathway used by all cells for movement of intracellular cargoes between subcellular compartments and in and out of the cell. Classically, constitutive trafficking was thought to be continuous and unregulated, in contrast to regulated secretion, wherein vesicles are stored intracellularly until undergoing synchronous membrane fusion following a Ca2+ signal. However, as shown in the literature reviewed here, many continuous trafficking steps can be up- or down-regulated by Ca2+, including several steps associated with human pathologies. Notably, we describe a series of Ca2+ pumps, channels, Ca2+-binding effector proteins, and their trafficking machinery targets that together regulate the flux of cargo in response to genetic alterations as well as baseline and agonist-dependent Ca2+ signals. Here, we review the most recent advances, organized by organellar location, that establish the importance of these components in trafficking steps. Ultimately, we conclude that Ca2+ regulates an expanding series of distinct mechanistic steps. Furthermore, the involvement of Ca2+ in trafficking is complex. For example, in some cases, the same Ca2+ effectors regulate surprisingly distinct trafficking steps, or even the same trafficking step with opposing influences, through binding to different target proteins.
Collapse
Affiliation(s)
- John Sargeant
- Division of Biological Sciences & Center for Structural & Functional Neuroscience, University of Montana, 32 Campus Drive, Missoula, MT 59812, USA
| | - Jesse C Hay
- Division of Biological Sciences & Center for Structural & Functional Neuroscience, University of Montana, 32 Campus Drive, Missoula, MT 59812, USA
| |
Collapse
|
11
|
Baliarsingh S, Sahoo S, Jo YH, Han YS, Sarkar A, Lee YS, Mohanty J, Patnaik BB. Molecular cloning, sequence characterization, and expression analysis of C-type lectin (CTL) and ER-Golgi intermediate compartment 53-kDa protein (ERGIC-53) homologs from the freshwater prawn, Macrobrachium rosenbergii. AQUACULTURE INTERNATIONAL : JOURNAL OF THE EUROPEAN AQUACULTURE SOCIETY 2022; 30:1011-1035. [PMID: 35153391 PMCID: PMC8816683 DOI: 10.1007/s10499-022-00845-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 12/20/2021] [Indexed: 06/14/2023]
Abstract
UNLABELLED Lectin protein families are diverse and multi-functional in crustaceans. The carbohydrate-binding domains (CRDs) of lectins recognize the molecular patterns associated with pathogens and orchestrate important roles in crustacean defense. In this study, two lectin homologs, a single CRD containing C-type lectin (CTL) and an L-type lectin (LTL) domain containing endoplasmic reticulum Golgi intermediate compartment 53 kDa protein (ERGIC-53) were identified from the freshwater prawn, Macrobrachium rosenbergii. The open reading frames of MrCTL and MrERGIC-53 were 654 and 1,515 bp, encoding polypeptides of 217 and 504 amino acids, respectively. Further, MrCTL showed a 20-amino acid transmembrane helix region and 10 carbohydrate-binding residues within the CRD. MrERGIC-53 showed a signal peptide region, a type-I transmembrane region, and a coiled-coil region at the C-terminus. Phylogenetic analysis revealed a close relationship between MrCTL and MrLectin and M. nipponense CTL (MnCTL), whereas MrERGIC-53 shared high sequence identity with Eriocheir sinensis ERGIC-53 and Penaeus vannamei MBL-1. A homology-based model predicted small carbohydrate-combining sites with a metal-binding site for ligand binding (Ca2+ binding site) in MrCTL and beta-sheets connected by short loops and beta-bends forming a dome-shaped beta-barrel structure representing the LTL domain of MrERGIC-53. Quantitative real-time polymerase chain reaction detected MrCTL and MrERGIC-53 transcripts in all examined tissues, with particularly high levels observed in hemocytes, hepatopancreas, and mucosal-associated tissues, such as the stomach and intestine. Further, the expression levels of MrCTL and MrERGIC-53 transcripts were remarkably altered after V. harveyi challenge, suggesting putative function in host innate immunity. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s10499-022-00845-3.
Collapse
Affiliation(s)
- Snigdha Baliarsingh
- PG Department of Biosciences and Biotechnology, Fakir Mohan University, Vyasa Vihar, Nuapadhi, Balasore, 756089 Odisha India
| | - Sonalina Sahoo
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Kausalyaganga, Bhubaneswar, 751002 Odisha India
| | - Yong Hun Jo
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture, School of Agriculture and Life Sciences, Chonnam National University, Gwangju, South Korea
| | - Yeon Soo Han
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture, School of Agriculture and Life Sciences, Chonnam National University, Gwangju, South Korea
| | - Arup Sarkar
- School of Biotech Sciences, Trident Academy of Creative Technology, Chandaka Industrial Estate, Chandrasekharpur, Bhubaneswar, 751024 Odisha India
| | - Yong Seok Lee
- School of Life Sciences and Biotechnology, College of Natural Sciences, Soonchunhyang University, Asan City, Asan, South Korea
| | - Jyotirmaya Mohanty
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Kausalyaganga, Bhubaneswar, 751002 Odisha India
| | - Bharat Bhusan Patnaik
- PG Department of Biosciences and Biotechnology, Fakir Mohan University, Vyasa Vihar, Nuapadhi, Balasore, 756089 Odisha India
| |
Collapse
|
12
|
Pérez-Rodriguez S, Wulff T, Voldborg BG, Altamirano C, Trujillo-Roldán MA, Valdez-Cruz NA. Compartmentalized Proteomic Profiling Outlines the Crucial Role of the Classical Secretory Pathway during Recombinant Protein Production in Chinese Hamster Ovary Cells. ACS OMEGA 2021; 6:12439-12458. [PMID: 34056395 PMCID: PMC8154153 DOI: 10.1021/acsomega.0c06030] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/24/2021] [Indexed: 05/11/2023]
Abstract
Different cellular processes that contribute to protein production in Chinese hamster ovary (CHO) cells have been previously investigated by proteomics. However, although the classical secretory pathway (CSP) has been well documented as a bottleneck during recombinant protein (RP) production, it has not been well represented in previous proteomic studies. Hence, the significance of this pathway for production of RP was assessed by identifying its own proteins that were associated to changes in RP production, through subcellular fractionation coupled to shot-gun proteomics. Two CHO cell lines producing a monoclonal antibody with different specific productivities were used as cellular models, from which 4952 protein groups were identified, which represent a coverage of 59% of the Chinese hamster proteome. Data are available via ProteomeXchange with identifier PXD021014. By using SAM and ROTS algorithms, 493 proteins were classified as differentially expressed, of which about 80% was proposed as novel targets and one-third were assigned to the CSP. Endoplasmic reticulum (ER) stress, unfolded protein response, calcium homeostasis, vesicle traffic, glycosylation, autophagy, proteasomal activity, protein synthesis and translocation into ER lumen, and secretion of extracellular matrix components were some of the affected processes that occurred in the secretory pathway. Processes from other cellular compartments, such as DNA replication, transcription, cytoskeleton organization, signaling, and metabolism, were also modified. This study gives new insights into the molecular traits of higher producer cells and provides novel targets for development of new sub-lines with improved phenotypes for RP production.
Collapse
Affiliation(s)
- Saumel Pérez-Rodriguez
- Programa
de Investigación de Producción de Biomoléculas,
Departamento de Biología Molecular y Biotecnología,
Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán 04510 Ciudad de
México, México
| | - Tune Wulff
- The
Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby 2800, Denmark
| | - Bjørn G. Voldborg
- The
Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby 2800, Denmark
| | - Claudia Altamirano
- Laboratorio
de Cultivos Celulares, Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Avenida Brasil 2085 Valparaíso, Chile
| | - Mauricio A. Trujillo-Roldán
- Programa
de Investigación de Producción de Biomoléculas,
Departamento de Biología Molecular y Biotecnología,
Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán 04510 Ciudad de
México, México
| | - Norma A. Valdez-Cruz
- Programa
de Investigación de Producción de Biomoléculas,
Departamento de Biología Molecular y Biotecnología,
Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán 04510 Ciudad de
México, México
| |
Collapse
|
13
|
Qiu L, Xie M, Zhou M, Liu X, Hu Z, Wu L. Restoration of FVIII Function and Phenotypic Rescue in Hemophilia A Mice by Transplantation of MSCs Derived From F8-Modified iPSCs. Front Cell Dev Biol 2021; 9:630353. [PMID: 33644070 PMCID: PMC7905062 DOI: 10.3389/fcell.2021.630353] [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: 11/17/2020] [Accepted: 01/25/2021] [Indexed: 01/19/2023] Open
Abstract
Hemophilia A (HA), an X-linked recessive congenital bleeding disorder, affects 80%–85% of patients with hemophilia. Nearly half of severe cases of hemophilia are caused by a 0.6-Mb genomic inversion (Inv22) that disrupts F8. Although viral-based gene therapy has shown therapeutic effects for hemophilia B (HB), this promising approach is not applicable for HA at the present stage; this limitation is mainly due to the large size of F8 cDNA, which far exceeds the adeno-associated virus (AAV) packaging capacity. We previously reported an in situ genetic correction of Inv22 in HA patient-specific induced pluripotent stem cells (HA-iPSCs) by using TALENs. We also investigated an alternative strategy for targeted gene addition, in which cDNA of the B-domain deleted F8 (BDDF8) was targeted at the rDNA locus of HA-iPSCs using TALENickases to restore FVIII function. Mesenchymal stem cells (MSCs) have low immunogenicity and can secrete FVIII under physiological conditions; in this study, MSCs were differentiated from F8-corrected iPSCs, BDDF8-iPSCs, and HA-iPSCs. Differentiated MSCs were characterized, and FVIII expression efficacy in MSCs was verified in vitro. The three types of MSCs were introduced into HA mice via intravenous injection. Long-term engraftment with restoration of FVIII function and phenotypic rescue was observed in HA mice transplanted with F8-corrected iMSCs and BDDF8-iMSCs. Our findings suggest that ex vivo gene therapy using iMSCs derived from F8-modified iPSCs can be feasible, effective, and promising for the clinical translation of therapeutic gene editing of HA and other genetic birth defects, particularly those that involve large sequence variants.
Collapse
Affiliation(s)
- Liyan Qiu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetic, School of Life Sciences, Central South University, Changsha, China
| | - Mi Xie
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetic, School of Life Sciences, Central South University, Changsha, China
| | - Miaojin Zhou
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetic, School of Life Sciences, Central South University, Changsha, China
| | - Xionghao Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetic, School of Life Sciences, Central South University, Changsha, China
| | - Zhiqing Hu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetic, School of Life Sciences, Central South University, Changsha, China
| | - Lingqian Wu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetic, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
14
|
Viinikangas T, Khosrowabadi E, Kellokumpu S. N-Glycan Biosynthesis: Basic Principles and Factors Affecting Its Outcome. EXPERIENTIA SUPPLEMENTUM (2012) 2021; 112:237-257. [PMID: 34687012 DOI: 10.1007/978-3-030-76912-3_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Carbohydrate chains are the most abundant and diverse of nature's biopolymers and represent one of the four fundamental macromolecular building blocks of life together with proteins, nucleic acids, and lipids. Indicative of their essential roles in cells and in multicellular organisms, genes encoding proteins associated with glycosylation account for approximately 2% of the human genome. It has been estimated that 50-80% of all human proteins carry carbohydrate chains-glycans-as part of their structure. Despite cells utilize only nine different monosaccharides for making their glycans, their order and conformational variation in glycan chains together with chain branching differences and frequent post-synthetic modifications can give rise to an enormous repertoire of different glycan structures of which few thousand is estimated to carry important structural or functional information for a cell. Thus, glycans are immensely versatile encoders of multicellular life. Yet, glycans do not represent a random collection of unpredictable structures but rather, a collection of predetermined but still dynamic entities that are present at defined quantities in each glycosylation site of a given protein in a cell, tissue, or organism.In this chapter, we will give an overview of what is currently known about N-glycan synthesis in higher eukaryotes, focusing not only on the processes themselves but also on factors that will affect or can affect the final outcome-the dynamicity and heterogeneity of the N-glycome. We hope that this review will help understand the molecular details underneath this diversity, and in addition, be helpful for those who plan to produce optimally glycosylated antibody-based therapeutics.
Collapse
Affiliation(s)
- Teemu Viinikangas
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Elham Khosrowabadi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Sakari Kellokumpu
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| |
Collapse
|
15
|
Altered phenotype in LMAN1-deficient mice with low levels of residual LMAN1 expression. Blood Adv 2020; 4:5635-5643. [PMID: 33196840 PMCID: PMC7686883 DOI: 10.1182/bloodadvances.2020002523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/13/2020] [Indexed: 01/28/2023] Open
Abstract
Combined deficiency of coagulation factors V and VIII (F5F8D) is an autosomal recessive bleeding disorder caused by loss-of-function mutations in either LMAN1 or MCFD2. The latter genes encode 2 components of a mammalian cargo receptor that facilitates secretion of coagulation factor V (FV) and factor VIII (FVIII) from the endoplasmic reticulum (ER) to the Golgi via coat protein complex II vesicles. F5F8D patients exhibit FV and FVIII levels that are ∼10% to 15% of normal. We report herein a comparative analysis for a series of murine Lman1 alleles. Consistent with previous reports, mice completely deficient in LMAN1 (Lman1-/-) exhibit ∼50% FV and FVIII levels. In contrast, mice carrying a hypomorphic Lman1 allele (Lman1cgt/cgt) that expresses ∼6% to 8% of wild-type Lman1 mRNA levels exhibit intermediate plasma FV and FVIII reductions (∼70% of wild-type levels). Lman1-/- mice exhibit ER accumulation of another LMAN1 cargo, alpha-1 antitrypsin (A1AT), with an intermediate level of A1AT ER retention observed in Lman1cgt/cgt mice. Finally, the previously reported strain-specific, partially penetrant, perinatal lethality of LMAN1-deficient mice (Lman1gt1/gt1) was confirmed in Lman1-/- mice, although it was not observed in Lman1cgt/cgt mice. Taken together, these results show a dose-dependent effect of residual LMAN1 on the secretion of its cargo proteins. The results also suggest that human subjects with hypomorphic LMAN1 mutations might present with mild bleeding phenotypes resulting from more modest reductions in FV and FVIII, which could be missed by routine clinical evaluation. Finally, these findings suggest that therapeutic targeting of LMAN1 to reduce FV and FVIII as an anticoagulant strategy may only require partial inhibition of LMAN1 function.
Collapse
|
16
|
Elnaggar M, Al-Mohannadi A, Kizhakayil D, Raynaud CM, Al-Mannai S, Gentilcore G, Pavlovski I, Sathappan A, Van Panhuys N, Borsotti C, Follenzi A, Grivel JC, Deola S. Flow-Cytometry Platform for Intracellular Detection of FVIII in Blood Cells: A New Tool to Assess Gene Therapy Efficiency for Hemophilia A. Mol Ther Methods Clin Dev 2020; 17:1-12. [PMID: 31886317 PMCID: PMC6920166 DOI: 10.1016/j.omtm.2019.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 11/03/2019] [Indexed: 01/08/2023]
Abstract
Detection of factor VIII (FVIII) in cells by flow cytometry is controversial, and no monoclonal fluorescent antibody is commercially available. In this study, we optimized such an assay and successfully used it as a platform to study the functional properties of phosphoglycerate kinase (PGK)-FVIII lentiviral vector-transduced cells by directly visualizing FVIII in cells after different gene transfer conditions. We could measure cellular stress parameters after transduction by correlating gene expression and protein accumulation data. Flow cytometry performed on transduced cell lines showed that increasing MOI rates resulted in increased protein levels, plateauing after an MOI of 30. We speculated that, at higher MOI, FVIII production could be impaired by a limiting factor required for proper folding. To test this hypothesis, we interfered with the unfolded protein response by blocking proteasomal degradation and measured the accumulation of intracellular misfolded protein. Interestingly, at higher MOIs the cells displayed signs of toxicity with reactive oxygen species accumulation. This suggests the need for identifying a safe window of transduction dose to avoid consequent cell toxicity. Herein, we show that our flow cytometry platform for intracytoplasmic FVIII protein detection is a reliable method for optimizing gene therapy protocols in hemophilia A by shedding light on the functional status of cells after gene transfer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Igor Pavlovski
- Research Department, Sidra Medicine, PO Box 26999, Doha, Qatar
| | | | | | - Chiara Borsotti
- Department of Health Sciences, Università del Piemonte Orientale “A. Avogadro,” 28100 Novara, Italy
| | - Antonia Follenzi
- Department of Health Sciences, Università del Piemonte Orientale “A. Avogadro,” 28100 Novara, Italy
| | | | - Sara Deola
- Research Department, Sidra Medicine, PO Box 26999, Doha, Qatar
| |
Collapse
|
17
|
Anelli T, Panina-Bordignon P. How to Avoid a No-Deal ER Exit. Cells 2019; 8:cells8091051. [PMID: 31500301 PMCID: PMC6769657 DOI: 10.3390/cells8091051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 08/28/2019] [Accepted: 09/06/2019] [Indexed: 01/01/2023] Open
Abstract
Efficiency and fidelity of protein secretion are achieved thanks to the presence of different steps, located sequentially in time and space along the secretory compartment, controlling protein folding and maturation. After entering into the endoplasmic reticulum (ER), secretory proteins attain their native structure thanks to specific chaperones and enzymes. Only correctly folded molecules are allowed by quality control (QC) mechanisms to leave the ER and proceed to downstream compartments. Proteins that cannot fold properly are instead retained in the ER to be finally destined to proteasomal degradation. Exiting from the ER requires, in most cases, the use of coated vesicles, departing at the ER exit sites, which will fuse with the Golgi compartment, thus releasing their cargoes. Protein accumulation in the ER can be caused by a too stringent QC or by ineffective transport: these situations could be deleterious for the organism, due to the loss of the secreted protein, and to the cell itself, because of abnormal increase of protein concentration in the ER. In both cases, diseases can arise. In this review, we will describe the pathophysiology of protein folding and transport between the ER and the Golgi compartment.
Collapse
Affiliation(s)
- Tiziana Anelli
- Vita-Salute San Raffaele University, 20132 Milan, Italy.
- Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Paola Panina-Bordignon
- Vita-Salute San Raffaele University, 20132 Milan, Italy.
- Division of Neuroscience, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| |
Collapse
|
18
|
ssODN-Mediated In-Frame Deletion with CRISPR/Cas9 Restores FVIII Function in Hemophilia A-Patient-Derived iPSCs and ECs. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:198-209. [PMID: 31261034 PMCID: PMC6610636 DOI: 10.1016/j.omtn.2019.05.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 05/25/2019] [Accepted: 05/27/2019] [Indexed: 12/31/2022]
Abstract
Given that the cDNA of F8 is too large to be packaged into adeno-associated virus (AAV) capsids, gene transfer of some versions of B-domain-deleted F8 (BDD-F8) for hemophilia A (HA) treatment has been attempted with promising results. Here, we describe an efficient gene correction via single-stranded-oligodeoxynucleotide (ssODN)-mediated in-frame deletion within the B domain of F8 with CRISPR/Cas9 in HA-patient-derived induced pluripotent stem cells (HA-iPSCs). The expression and activity of FVIII was restored in corrected HA-iPSC-derived induced endothelial progenitor cells (C-iEPCs) in vitro and in vivo. The bleeding phenotype was rescued in HA mice after C-iEPC infusion. Our results demonstrate an efficient approach for in situ gene correction via introduction of a tiny deletion using ssODN and CRISPR/Cas9 to reframe the F8 transcript and restore FVIII function in HA-iPSC-derived EPCs with potential clinical impact in HA gene therapy. For the first time, we demonstrated in vitro and in vivo the FVIII function that is encoded by the endogenous F8 gene with a partially deleted B domain. This work also suggests an applicable strategy for genetic correction of other gene frameshift mutations.
Collapse
|
19
|
Analysis of MCFD2- and LMAN1-deficient mice demonstrates distinct functions in vivo. Blood Adv 2019; 2:1014-1021. [PMID: 29735583 DOI: 10.1182/bloodadvances.2018018317] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 04/14/2018] [Indexed: 12/23/2022] Open
Abstract
The LMAN1-MCFD2 complex serves as a cargo receptor for efficient transport of factor V (FV) and FVIII from the endoplasmic reticulum (ER) to the Golgi. Genetic deficiency of LMAN1 or MCFD2 in humans results in the moderate bleeding disorder combined FV and FVIII deficiency, with a similar phenotype previously observed in LMAN1-deficient mice. We now report that MCFD2-deficient mice generated by gene targeting also demonstrate reduced plasma FV and FVIII, with levels lower than those in LMAN1-deficient mice, similar to previous observations in LMAN1- and MCDF2-deficient humans. Surprisingly, FV and FVIII levels in doubly deficient mice match the higher levels observed in LMAN1-deficient mice. In contrast to the strain-specific partial lethality previously observed in LMAN1-null mice, MCFD2-null mice demonstrate normal survival in different genetic backgrounds, although doubly deficient mice exhibit partial embryonic lethality comparable to LMAN1-deficient mice. These results suggest that an alternative pathway is responsible for FV/FVIII secretion in doubly deficient mice and distinct cargo-specific functions for LMAN1 and MCFD2 within the ER-to-Golgi secretory pathway. We also observed decreased plasma levels of α1-antitrypsin (AAT) in male mice for all 3 groups of deficient mice. Comparable accumulation of AAT was observed in hepatocyte ER of singly and doubly deficient mice, demonstrating a role for LMAN1 and MCFD2 in efficient ER exit of AAT.
Collapse
|
20
|
Fu YL, Zhang B, Mu TW. LMAN1 (ERGIC-53) promotes trafficking of neuroreceptors. Biochem Biophys Res Commun 2019; 511:356-362. [PMID: 30791981 DOI: 10.1016/j.bbrc.2019.02.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 02/10/2019] [Indexed: 12/15/2022]
Abstract
The endoplasmic reticulum-Golgi intermediate compartment protein-53 (ERGIC-53, aka LMAN1), which cycles between the endoplasmic reticulum (ER) and Golgi, is a known cargo receptor for a number of soluble proteins. However, whether LMAN1 plays a role as a trafficking factor in the central nervous system is largely unknown. Here, we determined the role of LMAN1 on endogenous protein levels of the Cys-loop superfamily of neuroreceptors, including gamma-aminobutyric acid type A receptors (GABAARs), 5-hydroxytryptamine (serotonin) type 3 (5-HT3) receptors, and nicotinic acetylcholine receptors (nAChRs). Knockdown of LMAN1 reduces the surface trafficking of endogenous β3 subunits of GABAARs in mouse hypothalamic GT1-7 neurons. Furthermore, Western blot analysis of brain homogenates from LMAN1 knockout mice demonstrated that loss of LMAN1 decreases the total protein levels of 5HT3A receptors and γ2 subunits of GABAARs. LMAN1 knockout regulates the ER proteostasis network by upregulating ERP44 without changing calnexin levels. Interestingly, despite the critical role of the glycan-binding function of LMAN1 in its other known cargo clients, LMAN1 interacts with GABAARs in a glycan-independent manner. In summary, LMAN1 is a trafficking factor for certain neuroreceptors in the central nervous system. This is the first report of LMAN1 function in membrane protein trafficking.
Collapse
Affiliation(s)
- Yan-Lin Fu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Bin Zhang
- Genomic Medicine Institute, Lerner Research Institute of Cleveland Clinic, Cleveland, OH, USA
| | - Ting-Wei Mu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
21
|
Abstract
The flow of cargo vesicles along the secretory pathway requires concerted action among various regulators. The COPII complex, assembled by the activated SAR1 GTPases on the surface of the endoplasmic reticulum, orchestrates protein interactions to package cargos and generate transport vesicles en route to the Golgi. The dynamic nature of COPII, however, hinders analysis with conventional biochemical assays. Here we apply proximity-dependent biotinylation labeling to capture the dynamics of COPII transport in cells. When SAR1B was fused with a promiscuous biotin ligase, BirA*, the fusion protein SAR1B-BirA* biotinylates and thus enables the capture of COPII machinery and cargos in a GTP-dependent manner. Biochemical and pulse-chase imaging experiments demonstrate that the COPII coat undergoes a dynamic cycle of engagement-disengagement with the transmembrane cargo receptor LMAN1/ERGIC53. LMAN1 undergoes a process of concentrative sorting by the COPII coat, via a dimeric sorting code generated by oligomerization of the cargo receptor. Similar oligomerization events have been observed with other COPII sorting signals, suggesting that dimeric/multimeric sorting codes may serve as a general mechanism to generate selectivity of cargo sorting.
Collapse
|
22
|
Molecular mechanisms of missense mutations that generate ectopic N-glycosylation sites in coagulation factor VIII. Biochem J 2018; 475:873-886. [PMID: 29444815 DOI: 10.1042/bcj20170884] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/07/2018] [Accepted: 02/14/2018] [Indexed: 11/17/2022]
Abstract
N-glycosylation is a common posttranslational modification of secreted and membrane proteins, catalyzed by the two enzymatic isoforms of the oligosaccharyltransferase, STT3A and STT3B. Missense mutations are the most common mutations in inherited diseases; however, missense mutations that generate extra, non-native N-glycosylation sites have not been well characterized. Coagulation factor VIII (FVIII) contains five consensus N-glycosylation sites outside its functionally dispensable B domain. We developed a computer program that identified hemophilia A mutations in FVIII that can potentially create ectopic glycosylation sites. We determined that 18 of these ectopic sites indeed become N-glycosylated. These sites span the domains of FVIII and are primarily associated with a severe disease phenotype. Using STT3A and STT3B knockout cells, we determined that ectopic glycosylation exhibited different degrees of dependence on STT3A and STT3B. By separating the effects of ectopic N-glycosylation from those due to underlying amino acid changes, we showed that ectopic glycans promote the secretion of some mutants, but impair the secretion of others. However, ectopic glycans that enhanced secretion could not functionally replace a native N-glycan in the same domain. Secretion-deficient mutants, but not mutants with elevated secretion levels, show increased association with the endoplasmic reticulum chaperones BiP (immunoglobulin heavy chain-binding protein) and calreticulin. Though secreted to different extents, all studied mutants exhibited lower relative activity than wild-type FVIII. Our results reveal differential impacts of ectopic N-glycosylation on FVIII folding, trafficking and activity, which highlight complex disease-causing mechanisms of FVIII missense mutations. Our findings are relevant to other secreted and membrane proteins with mutations that generate ectopic N-glycans.
Collapse
|
23
|
Suzuki S, Nakamura Y, Suzuki N, Yamazaki T, Takagi Y, Tamura S, Takagi A, Kanematsu T, Matsushita T, Kojima T. Combined deficiency of factors V and VIII by chance coinheritance of parahaemophilia and haemophilia A, but not by mutations of either LMAN1 or MCFD2, in a Japanese family. Haemophilia 2017; 24:e13-e16. [PMID: 29082580 DOI: 10.1111/hae.13360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2017] [Indexed: 11/26/2022]
Affiliation(s)
- S Suzuki
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Y Nakamura
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - N Suzuki
- Department of Transfusion Medicine, Nagoya University Hospital, Nagoya, Japan
| | | | - Y Takagi
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - S Tamura
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - A Takagi
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - T Kanematsu
- Department of Clinical Laboratory, Nagoya University Hospital, Nagoya, Japan
| | - T Matsushita
- Department of Transfusion Medicine, Nagoya University Hospital, Nagoya, Japan.,Department of Clinical Laboratory, Nagoya University Hospital, Nagoya, Japan
| | - T Kojima
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
24
|
Missense mutations near the N-glycosylation site of the A2 domain lead to various intracellular trafficking defects in coagulation factor VIII. Sci Rep 2017; 7:45033. [PMID: 28327546 PMCID: PMC5361195 DOI: 10.1038/srep45033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 02/20/2017] [Indexed: 11/14/2022] Open
Abstract
Missense mutation is the most common mutation type in hemophilia. However, the majority of missense mutations remain uncharacterized. Here we characterize how hemophilia mutations near the unused N-glycosylation site of the A2 domain (N582) of FVIII affect protein conformation and intracellular trafficking. N582 is located in the middle of a short 310-helical turn (D580-S584), in which most amino acids have multiple hemophilia mutations. All 14 missense mutations found in this 310-helix reduced secretion levels of the A2 domain and full-length FVIII. Secreted mutants have decreased activities relative to WT FVIII. Selected mutations also lead to partial glycosylation of N582, suggesting that rapid folding of local conformation prevents glycosylation of this site in wild-type FVIII. Protease sensitivity, stability and degradation of the A2 domain vary among mutants, and between non-glycosylated and glycosylated species of the same mutant. Most of the mutants interact with the ER chaperone BiP, while only mutants with aberrant glycosylation interact with calreticulin. Our results show that the short 310-helix from D580 to S584 is critical for proper biogenesis of the A2 domain and FVIII, and reveal a range of molecular mechanisms by which FVIII missense mutations lead to moderate to severe hemophilia A.
Collapse
|
25
|
Brown L, Tilzer L, Plapp F. Factor V and VIII deficiency treated with therapeutic plasma exchange prior to redo mitral valve replacement. J Clin Apher 2016; 32:196-199. [DOI: 10.1002/jca.21478] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 05/24/2016] [Indexed: 11/10/2022]
Affiliation(s)
- Laura Brown
- Department of Pathology and Laboratory Medicine; Kansas University Medical Center; Kansas City Kansas
| | - Lowell Tilzer
- Department of Pathology and Laboratory Medicine; Kansas University Medical Center; Kansas City Kansas
| | - Fred Plapp
- Department of Pathology and Laboratory Medicine; Kansas University Medical Center; Kansas City Kansas
| |
Collapse
|
26
|
Aldiri I, Ajioka I, Xu B, Zhang J, Chen X, Benavente C, Finkelstein D, Johnson D, Akiyama J, Pennacchio LA, Dyer MA. Brg1 coordinates multiple processes during retinogenesis and is a tumor suppressor in retinoblastoma. Development 2016; 142:4092-106. [PMID: 26628093 PMCID: PMC4712833 DOI: 10.1242/dev.124800] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Retinal development requires precise temporal and spatial coordination of cell cycle exit, cell fate specification, cell migration and differentiation. When this process is disrupted, retinoblastoma, a developmental tumor of the retina, can form. Epigenetic modulators are central to precisely coordinating developmental events, and many epigenetic processes have been implicated in cancer. Studying epigenetic mechanisms in development is challenging because they often regulate multiple cellular processes; therefore, elucidating the primary molecular mechanisms involved can be difficult. Here we explore the role of Brg1 (Smarca4) in retinal development and retinoblastoma in mice using molecular and cellular approaches. Brg1 was found to regulate retinal size by controlling cell cycle length, cell cycle exit and cell survival during development. Brg1 was not required for cell fate specification but was required for photoreceptor differentiation and cell adhesion/polarity programs that contribute to proper retinal lamination during development. The combination of defective cell differentiation and lamination led to retinal degeneration in Brg1-deficient retinae. Despite the hypocellularity, premature cell cycle exit, increased cell death and extended cell cycle length, retinal progenitor cells persisted in Brg1-deficient retinae, making them more susceptible to retinoblastoma. ChIP-Seq analysis suggests that Brg1 might regulate gene expression through multiple mechanisms. Summary: The SWI/SNF protein Brg1 controls cell cycle length, cell cycle exit and cell survival, and is required for cell differentiation and retinal lamination, in the developing mouse retina.
Collapse
Affiliation(s)
- Issam Aldiri
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Itsuki Ajioka
- Center for Brain Integration Research (CBIR), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Beisi Xu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jiakun Zhang
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Claudia Benavente
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Dianna Johnson
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jennifer Akiyama
- Lawrence Berkeley National Laboratory, Genomics Division, Berkeley, CA 94701, USA Department of Energy, Joint Genome Institute, Walnut Creek, CA 94598, USA
| | - Len A Pennacchio
- Lawrence Berkeley National Laboratory, Genomics Division, Berkeley, CA 94701, USA Department of Energy, Joint Genome Institute, Walnut Creek, CA 94598, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN 38163, USA Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
27
|
Lai JD, Georgescu MT, Hough C, Lillicrap D. To clear or to fear: An innate perspective on factor VIII immunity. Cell Immunol 2015; 301:82-9. [PMID: 26547364 PMCID: PMC7124272 DOI: 10.1016/j.cellimm.2015.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/26/2015] [Accepted: 10/26/2015] [Indexed: 12/12/2022]
Abstract
FVIII inhibitor development involves a combination of innate immune modulators. Clearance and immunity is influenced at 3 levels: the protein, cell, and location. Cells associated with FVIII half-life may influence the immune response against FVIII.
The enigma that is factor VIII immunogenicity remains ever pertinent in the treatment of hemophilia A. Development of neutralizing antibodies against the therapeutic protein in 25–30% of patients likely depends on the appropriate activation of the innate immune response shortly following antigen encounter. Our understanding of this important immunological synapse remains ill-defined. In this review, we examine the three distinct factors contributing to the fate of factor VIII almost immediately after infusion: the characteristics of the protein, the cell, and the microenvironment. We propose a continuum between clearance and antigen presentation that facilitates removal of FVIII from circulation leading to either tolerance or immunity.
Collapse
Affiliation(s)
- Jesse Derek Lai
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, Canada
| | | | - Christine Hough
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, Canada
| | - David Lillicrap
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, Canada.
| |
Collapse
|
28
|
Wang A, Duan Q, Ding K, Liu X, Wu J, Sun Z. Successful abdominal operation without replacement therapy in a patient with combined factor V (FV) and FVIII deficiency due to novel homozygous mutation in LMAN1. Haemophilia 2015; 21:e492-4. [PMID: 26193913 DOI: 10.1111/hae.12756] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2015] [Indexed: 11/27/2022]
Affiliation(s)
- A. Wang
- Department of Hematology; Anhui Medical University Affiliated Anhui Provincial Hospital; Hefei China
- Anhui Provincial Hemophilia Treatment Center; Anhui Provincial Hospital; Hefei China
| | - Q. Duan
- Department of Hematology; Anhui Medical University Affiliated Anhui Provincial Hospital; Hefei China
- Anhui Provincial Hemophilia Treatment Center; Anhui Provincial Hospital; Hefei China
| | - K. Ding
- Department of Hematology; Anhui Medical University Affiliated Anhui Provincial Hospital; Hefei China
- Anhui Provincial Hemophilia Treatment Center; Anhui Provincial Hospital; Hefei China
| | - X. Liu
- Department of Hematology; Anhui Medical University Affiliated Anhui Provincial Hospital; Hefei China
- Anhui Provincial Hemophilia Treatment Center; Anhui Provincial Hospital; Hefei China
| | - J. Wu
- Department of Hematology; Anhui Medical University Affiliated Anhui Provincial Hospital; Hefei China
- Anhui Provincial Hemophilia Treatment Center; Anhui Provincial Hospital; Hefei China
| | - Z. Sun
- Department of Hematology; Anhui Medical University Affiliated Anhui Provincial Hospital; Hefei China
- Anhui Provincial Hemophilia Treatment Center; Anhui Provincial Hospital; Hefei China
| |
Collapse
|
29
|
Principles of treatment and update of recommendations for the management of haemophilia and congenital bleeding disorders in Italy. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2015; 12:575-98. [PMID: 25350962 DOI: 10.2450/2014.0223-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
30
|
Fang J, Liu M, Zhang X, Sakamoto T, Taatjes DJ, Jena BP, Sun F, Woods J, Bryson T, Kowluru A, Zhang K, Chen X. COPII-Dependent ER Export: A Critical Component of Insulin Biogenesis and β-Cell ER Homeostasis. Mol Endocrinol 2015; 29:1156-69. [PMID: 26083833 DOI: 10.1210/me.2015-1012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic β-cells possess a highly active protein synthetic and export machinery in the endoplasmic reticulum (ER) to accommodate the massive production of proinsulin. ER homeostasis is vital for β-cell functions and is maintained by the delicate balance between protein synthesis, folding, export, and degradation. Disruption of ER homeostasis by diabetes-causing factors leads to β-cell death. Among the 4 components to maintain ER homeostasis in β-cells, the role of ER export in insulin biogenesis is the least understood. To address this knowledge gap, the present study investigated the molecular mechanism of proinsulin ER export in MIN6 cells and primary islets. Two inhibitory mutants of the secretion-associated RAS-related protein (Sar)1 small GTPase, known to specifically block coat protein complex II (COPII)-dependent ER export, were overexpressed in β-cells using recombinant adenoviruses. Results from this approach, as well as small interfering RNA-mediated Sar1 knockdown, demonstrated that defective Sar1 function blocked proinsulin ER export and abolished its conversion to mature insulin in MIN6 cells, isolated mouse, and human islets. It is further revealed, using an in vitro vesicle formation assay, that proinsulin was packaged into COPII vesicles in a GTP- and Sar1-dependent manner. Blockage of COPII-dependent ER exit by Sar1 mutants strongly induced ER morphology change, ER stress response, and β-cell apoptosis. These responses were mediated by the PKR (double-stranded RNA-dependent kinase)-like ER kinase (PERK)/eukaryotic translation initiation factor 2α (p-eIF2α) and inositol-requiring protein 1 (IRE1)/x-box binding protein 1 (Xbp1) pathways but not via activating transcription factor 6 (ATF6). Collectively, results from the study demonstrate that COPII-dependent ER export plays a vital role in insulin biogenesis, ER homeostasis, and β-cell survival.
Collapse
Affiliation(s)
- Jingye Fang
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Ming Liu
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Xuebao Zhang
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Takeshi Sakamoto
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Douglas J Taatjes
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Bhanu P Jena
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Fei Sun
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - James Woods
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Tim Bryson
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Anjaneyulu Kowluru
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Kezhong Zhang
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Xuequn Chen
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| |
Collapse
|
31
|
Mumford AD, Ackroyd S, Alikhan R, Bowles L, Chowdary P, Grainger J, Mainwaring J, Mathias M, O'Connell N. Guideline for the diagnosis and management of the rare coagulation disorders: a United Kingdom Haemophilia Centre Doctors' Organization guideline on behalf of the British Committee for Standards in Haematology. Br J Haematol 2014; 167:304-26. [PMID: 25100430 DOI: 10.1111/bjh.13058] [Citation(s) in RCA: 210] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Andrew D Mumford
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Klaus J, Eisenhauer P, Russo J, Mason AB, Do D, King B, Taatjes D, Cornillez-Ty C, Boyson J, Thali M, Zheng C, Liao L, Yates J, Zhang B, Ballif B, Botten J. The intracellular cargo receptor ERGIC-53 is required for the production of infectious arenavirus, coronavirus, and filovirus particles. Cell Host Microbe 2014; 14:522-34. [PMID: 24237698 PMCID: PMC3999090 DOI: 10.1016/j.chom.2013.10.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 08/02/2013] [Accepted: 09/18/2013] [Indexed: 12/21/2022]
Abstract
Arenaviruses and hantaviruses cause severe human disease. Little is known regarding host proteins required for their propagation. We identified human proteins that interact with the glycoproteins (GPs) of a prototypic arenavirus and hantavirus and show that the lectin endoplasmic reticulum (ER)-Golgi intermediate compartment 53 kDa protein (ERGIC-53), a cargo receptor required for glycoprotein trafficking within the early exocytic pathway, associates with arenavirus, hantavirus, coronavirus, orthomyxovirus, and filovirus GPs. ERGIC-53 binds to arenavirus GPs through a lectin-independent mechanism, traffics to arenavirus budding sites, and is incorporated into virions. ERGIC-53 is required for arenavirus, coronavirus, and filovirus propagation; in its absence, GP-containing virus particles form but are noninfectious, due in part to their inability to attach to host cells. Thus, we have identified a class of pathogen-derived ERGIC-53 ligands, a lectin-independent basis for their association with ERGIC-53, and a role for ERGIC-53 in the propagation of several highly pathogenic RNA virus families.
Collapse
Affiliation(s)
- Joseph P. Klaus
- Department of Medicine, Division of Immunobiology, University of Vermont, Burlington, VT 05405, USA
- Cellular, Molecular and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, USA
| | - Philip Eisenhauer
- Department of Medicine, Division of Immunobiology, University of Vermont, Burlington, VT 05405, USA
| | - Joanne Russo
- Department of Medicine, Division of Immunobiology, University of Vermont, Burlington, VT 05405, USA
| | - Anne B. Mason
- Department of Biochemistry, University of Vermont, Burlington, VT 05405, USA
| | - Danh Do
- Department of Medicine, Division of Immunobiology, University of Vermont, Burlington, VT 05405, USA
| | - Benjamin King
- Department of Medicine, Division of Immunobiology, University of Vermont, Burlington, VT 05405, USA
- Cellular, Molecular and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, USA
| | - Douglas Taatjes
- Department of Pathology, University of Vermont, Burlington, VT 05405, USA
| | | | | | - Markus Thali
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405, USA
| | - Chunlei Zheng
- Genomic Medicine Institute, Lerner Research Institute of Cleveland Clinic, Cleveland, OH 44195, USA
| | - Lujian Liao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - John R. Yates
- The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bin Zhang
- Genomic Medicine Institute, Lerner Research Institute of Cleveland Clinic, Cleveland, OH 44195, USA
| | - Bryan A. Ballif
- Department of Biology, University of Vermont, Burlington, VT 05405, USA
| | - Jason W. Botten
- Department of Medicine, Division of Immunobiology, University of Vermont, Burlington, VT 05405, USA
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405, USA
- Corresponding author
| |
Collapse
|
33
|
Springer S, Malkus P, Borchert B, Wellbrock U, Duden R, Schekman R. Regulated Oligomerization Induces Uptake of a Membrane Protein into COPII Vesicles Independent of Its Cytosolic Tail. Traffic 2014; 15:531-45. [DOI: 10.1111/tra.12157] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 01/07/2014] [Accepted: 01/30/2014] [Indexed: 12/22/2022]
Affiliation(s)
| | - Per Malkus
- Department of Systems Biology; Harvard Medical School; Boston MA 02115 USA
| | - Britta Borchert
- Biochemistry and Cell Biology; Jacobs University Bremen; Bremen Germany
| | - Ursula Wellbrock
- Biochemistry and Cell Biology; Jacobs University Bremen; Bremen Germany
| | - Rainer Duden
- Centre for Structural and Cell Biology in Medicine, Institute of Biology; University of Lübeck; Lübeck Germany
| | - Randy Schekman
- Howard Hughes Medical Institute and Department of Molecular and Cell Biology; University of California, Berkeley; Berkeley CA 94720 USA
| |
Collapse
|
34
|
YAMAMOTO K. Intracellular lectins are involved in quality control of glycoproteins. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2014; 90:67-82. [PMID: 24522156 PMCID: PMC3948941 DOI: 10.2183/pjab.90.67] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Glycoprotein quality control is categorized into three kinds of reactions; the folding of nascent glycoproteins, ER-associated degradation of misfolded or unassembled glycoproteins, and transport and sorting of correctly folded glycoproteins. In all three processes, N-glycans on the glycoproteins are used as tags that are recognized by intracellular lectins. We analyzed the functions of these intracellular lectins and their sugar-binding specificities. The results clearly showed that the A, B, and C-arms of high mannose-type glycans participate in the folding, transport and sorting, and degradation, respectively, of newly synthesized peptides. After correctly folded glycoproteins are transported to the Golgi apparatus, N-glycans are trimmed into Man3GlcNAc2 and then rebuilt into various complex-type glycans in the Golgi, resulting in the addition of diverse sugar structures that allow glycoproteins to play various roles outside of the cells.
Collapse
Affiliation(s)
- Kazuo YAMAMOTO
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
- Corresponding should be addressed: K. Yamamoto, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan (e-mail: )
| |
Collapse
|
35
|
Jickling GC, Ander BP, Stamova B, Zhan X, Liu D, Rothstein L, Verro P, Khoury J, Jauch EC, Pancioli AM, Broderick JP, Sharp FR. RNA in blood is altered prior to hemorrhagic transformation in ischemic stroke. Ann Neurol 2013; 74:232-40. [PMID: 23468366 DOI: 10.1002/ana.23883] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 02/19/2013] [Accepted: 03/01/2013] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Hemorrhagic transformation (HT) is a major complication of ischemic stroke that worsens outcomes and increases mortality. Disruption of the blood-brain barrier is a central feature of HT pathogenesis, and leukocytes may contribute to this process. We sought to determine whether ischemic strokes that develop HT have differences in RNA expression in blood within 3 hours of stroke onset prior to treatment with thrombolytic therapy. METHODS Stroke patient blood samples were obtained prior to treatment with thrombolysis, and leukocyte RNA was assessed by microarray analysis. Strokes that developed HT (n = 11) were compared to strokes without HT (n = 33) and controls (n = 14). Genes were identified (corrected p < 0.05, fold change ≥|1.2|), and functional analysis was performed. RNA prediction of HT in stroke was evaluated using cross-validation, and in a second stroke cohort (n = 52). RESULTS Ischemic strokes that developed HT had differential expression of 29 genes in circulating leukocytes prior to treatment with thrombolytic therapy. A panel of 6 genes could predict strokes that later developed HT with 80% sensitivity and 70.2% specificity. Key pathways involved in HT of human stroke are described, including amphiregulin, a growth factor that regulates matrix metalloproteinase-9; a shift in transforming growth factor-β signaling involving SMAD4, INPP5D, and IRAK3; and a disruption of coagulation factors V and VIII. INTERPRETATION Identified genes correspond to differences in inflammation and coagulation that may predispose to HT in ischemic stroke. Given the adverse impact of HT on stroke outcomes, further evaluation of the identified genes and pathways is warranted to determine their potential as therapeutic targets to reduce HT and as markers of HT risk.
Collapse
Affiliation(s)
- Glen C Jickling
- Department of Neurology and the MIND Institute, University of California, Davis, Sacramento, CA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Zheng C, Zhang B. Combined deficiency of coagulation factors V and VIII: an update. Semin Thromb Hemost 2013; 39:613-20. [PMID: 23852824 DOI: 10.1055/s-0033-1349223] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Combined deficiency of factor V (FV) and FVIII (F5F8D) is an autosomal recessive bleeding disorder characterized by simultaneous decreases of both coagulation factors. This review summarizes recent reports on the clinical presentations, treatments, and molecular mechanism of F5F8D. Genetic studies identified LMAN1 and MCFD2 as causative genes for this disorder, revealing a previously unknown intracellular transport pathway shared by the two important blood coagulation factors. LMAN1 and MCFD2 form a Ca2+-dependent cargo receptor complex that functions in the transport of FV/FVIII from the endoplasmic reticulum (ER) to the Golgi. Disrupting the LMAN1-MCFD2 receptor, complex formation is the primary molecular defect of missense mutations leading to F5F8D. The EF-hand domains of MCFD2 are necessary and sufficient for the interactions with both LMAN1 and FV/FVIII. Similarly, the carbohydrate recognition domain of LMAN1 contains distinct and separable binding sites for both MCFD2 and FV/FVIII. Therefore, FV and FVIII likely carry duel sorting signals that are separately recognized by LMAN1 and MCFD2 and necessary for the efficient ER-to-Golgi transport. FV and FVIII likely bind LMAN1 through the high-mannose N-linked glycans under the higher Ca2+ conditions in the ER and dissociate in the lower Ca2+ environment of the ER-Golgi intermediate compartment.
Collapse
Affiliation(s)
- Chunlei Zheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | |
Collapse
|
37
|
Zheng C, Page RC, Das V, Nix JC, Wigren E, Misra S, Zhang B. Structural characterization of carbohydrate binding by LMAN1 protein provides new insight into the endoplasmic reticulum export of factors V (FV) and VIII (FVIII). J Biol Chem 2013; 288:20499-509. [PMID: 23709226 DOI: 10.1074/jbc.m113.461434] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
LMAN1 (ERGIC-53) is a key mammalian cargo receptor responsible for the export of a subset of glycoproteins from the endoplasmic reticulum. Together with its soluble coreceptor MCFD2, LMAN1 transports coagulation factors V (FV) and VIII (FVIII). Mutations in LMAN1 or MCFD2 cause the genetic bleeding disorder combined deficiency of FV and FVIII (F5F8D). The LMAN1 carbohydrate recognition domain (CRD) binds to both glycoprotein cargo and MCFD2 in a Ca(2+)-dependent manner. To understand the biochemical basis and regulation of LMAN1 binding to glycoprotein cargo, we solved crystal structures of the LMAN1-CRD bound to Man-α-1,2-Man, the terminal carbohydrate moiety of high mannose glycans. Our structural data, combined with mutagenesis and in vitro binding assays, define the central mannose-binding site on LMAN1 and pinpoint histidine 178 and glycines 251/252 as critical residues for FV/FVIII binding. We also show that mannobiose binding is relatively independent of pH in the range relevant for endoplasmic reticulum-to-Golgi traffic, but is sensitive to lowered Ca(2+) concentrations. The distinct LMAN1/MCFD2 interaction is maintained at these lowered Ca(2+) concentrations. Our results suggest that compartmental changes in Ca(2+) concentration regulate glycoprotein cargo binding and release from the LMAN1·MCFD2 complex in the early secretory pathway.
Collapse
Affiliation(s)
- Chunlei Zheng
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Patel AJ, Liu HH, Lager RA, Malkovska V, Zhang B. Successful percutaneous coronary intervention in a patient with combined deficiency of FV and FVIII due to novel compound heterozygous mutations in LMAN1. Haemophilia 2013; 19:607-10. [PMID: 23557496 DOI: 10.1111/hae.12128] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2013] [Indexed: 11/29/2022]
Abstract
Percutaneous coronary intervention (PCI) in patients with congenital coagulation factor deficiencies presents a unique challenge. They are not only at increased risk of perioperative bleeding but can also suffer thrombosis of the stent as preventive anticoagulation and antiplatelet therapy is difficult. Several cases of successful PCI have been described in patients with haemophilia A and B, but there are no reports in patients with combined coagulation factor deficiencies. We used PCI to treat the coronary artery disease in a patient with the combined deficiency of factor V and factor VIII (F5F8D) and analysed the molecular basis of the disorder for this patient. A 68-year-old patient was admitted for urgent PCI with bare metal stent placement after the diagnosis of the F5F8D. Peripheral blood DNA was extracted for the sequence analysis of LMAN1 and MCFD2 genes. Mutations in LMAN1 was confirmed by molecular cloning of the PCR product and resequencing of the resulting clones. The patient underwent successful PCI with good long-term outcome. Our patient tolerated anticoagulation therapy well, with unfractionated heparin, and double antiplatelet therapy while he was initially supported with fresh frozen plasma and recombinant FVIII. Molecular analysis revealed that the patient carries unusual compound heterozygous frameshift mutations on the same microsatellite repeat region in exon 8 of LMAN1, one of which is a novel mutation (c.912delA). Our results suggest that patients with F5F8D can safely undergo PCI for coronary artery disease, with the treatment individualized to the specific patient.
Collapse
Affiliation(s)
- A J Patel
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL, USA
| | | | | | | | | |
Collapse
|
39
|
Chen Y, Hojo S, Matsumoto N, Yamamoto K. Regulation of Mac-2BP secretion is mediated by its N-glycan binding to ERGIC-53. Glycobiology 2013; 23:904-16. [PMID: 23550150 DOI: 10.1093/glycob/cwt027] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The leguminous-type (L-type) lectin ER-Golgi intermediate compartment (ERGIC)-53, a homo-oligomeric endoplasmic reticulum (ER)-Golgi recycling protein, functions as a transport receptor for newly synthesized glycoproteins in the early secretory pathway. Although a limited subset of cargo glycoproteins transported by ERGIC-53, such as the coagulation factors V and VIII, cathepsin C and Z and α1-antitrypsin, has been identified, the exact role of the N-glycan binding of ERGIC-53 in the transport of secretory glycoproteins for ER exit has yet to be clarified. By screening a cDNA library isolated from HepG2 cells via a green fluorescent protein fragment complementation assay, we assessed several candidate luminal ERGIC-53-interacting partners and identified Mac-2 binding protein (Mac-2BP) as a novel ERGIC-53-transported cargo glycoprotein. Using an N-glycan-binding-deficient mutant of ERGIC-53 (N156A) or treatment with N-glycosylation processing inhibitors, as well as the introduction of the ER-mis-targeting mutant (KKAA), we demonstrated that the high-mannose-type N-glycan binding of ERGIC-53 contributes to its interaction with Mac-2BP, which is essential for the ERGIC-53-mediated ER-Golgi transport of nascent proteins during early secretion. Furthermore, we also provide evidence that MCFD2 is involved in the secretion of Mac-2BP. These observations reveal a distinct role for the N-glycan binding of ERGIC-53 in the receptor-mediated ER exit of newly synthesized Mac-2BP in the early secretion pathway.
Collapse
Affiliation(s)
- Yang Chen
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo, Kashiwa, 277-8562 Chiba, Japan
| | | | | | | |
Collapse
|
40
|
Russo R, Esposito MR, Iolascon A. Inherited hematological disorders due to defects in coat protein (COP)II complex. Am J Hematol 2013; 88:135-40. [PMID: 22764119 DOI: 10.1002/ajh.23292] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 05/28/2012] [Accepted: 06/07/2012] [Indexed: 11/10/2022]
Abstract
Many diseases attributed to trafficking defects are primary disorders of protein folding and assembly. However, an increasing number of disease states are directly attributable to defects in trafficking machinery. In this context, the cytoplasmic coat protein (COP)II complex plays a pivotal role: it mediates the anterograde transport of correctly folded secretory cargo from the endoplasmic reticulum towards the Golgi apparatus. This review attempts to describe the involvement of COPII complex alteration in the pathogenesis of human genetic disorders; particularly, we will focus on two disorders, the Congenital Dyserythropoietic Anemia type II and the Combined Deficiency of Factor V and VIII.
Collapse
Affiliation(s)
- Roberta Russo
- CEINGE Biotecnologie Avanzate; University Federico II of Naples; Naples; Italy
| | | | | |
Collapse
|
41
|
Bolton-Maggs PHB. The rare inherited coagulation disorders. Pediatr Blood Cancer 2013; 60 Suppl 1:S37-40. [PMID: 23109366 DOI: 10.1002/pbc.24336] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 08/30/2012] [Indexed: 11/10/2022]
Abstract
The rare inherited coagulation disorders (RICD) are uncommon and thus not well-defined in terms of severity or management. Inheritance is autosomal; in some of these disorders in the heterozygote state affected individuals may be mildly symptomatic. Severe deficiencies are more common in association with consanguinity. Factor X and factor XIII deficiency have the most severe manifestations, while factor XI deficiency is the least severe. Factor VII and factor XI deficiencies show a poor relationship between the factor level and bleeding risk. Unlike hemophilia, women are equally affected by these RICD and can have problems related to menstruation and childbirth.
Collapse
|
42
|
Zhu M, Das V, Zheng C, Majumdar S, Zhang B. A synonymous mutation in LMAN1 creates an ectopic splice donor site and causes combined deficiency of FV and FVIII. J Thromb Haemost 2012; 10:2407-9. [PMID: 23006835 PMCID: PMC3563766 DOI: 10.1111/jth.12002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Min Zhu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Vaijyaanti Das
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Chunlei Zheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Suvankar Majumdar
- Pediatric Hematology/Oncology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Bin Zhang
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW By definition, rare factor deficiencies have a prevalence of less than 200,000 in the US population, or an incidence of less than one in 2000 in Europe. The very small numbers of patients with rare disorders present challenges in diagnosis, evaluation of bleeding risk and treatment. Use of new assays, full genome sequencing, and global clotting assays will significantly improve diagnosis of patients with rare bleeding disorders. RECENT FINDINGS In addition to new assays available for monitoring patients, new therapy, both recombinant and plasma derived, is now available. Registries and clinical trials have demonstrated decreased bleeding and improved outcomes when patients are treated with these agents. Expanding international registries have been initiated to correlate genotype and bleeding phenotype in conjunction with global assays. SUMMARY Ongoing research continues to expand our understanding of the pathophysiology of rare factor deficiencies. This work complements medical practice to incorporate early diagnosis and new treatment options for patients, resulting in safer and less sensitizing regimens and much improved clinical outcomes.
Collapse
|
44
|
Tao J, Zhu M, Wang H, Afelik S, Vasievich MP, Chen XW, Zhu G, Jensen J, Ginsburg D, Zhang B. SEC23B is required for the maintenance of murine professional secretory tissues. Proc Natl Acad Sci U S A 2012; 109:E2001-9. [PMID: 22745161 PMCID: PMC3406820 DOI: 10.1073/pnas.1209207109] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
In eukaryotic cells, newly synthesized secretory proteins require COPII (coat protein complex II) to exit the endoplasmic reticulum (ER). COPII contains five core components: SAR1, SEC23, SEC24, SEC13, and SEC31. SEC23 is a GTPase-activating protein that activates the SAR1 GTPase and also plays a role in cargo recognition. Missense mutations in the human COPII paralogues SEC23A and SEC23B result in craniolenticulosutural dysplasia and congenital dyserythropoietic anemia type II, respectively. We now report that mice completely deficient for SEC23B are born with no apparent anemia phenotype, but die shortly after birth, with degeneration of professional secretory tissues. In SEC23B-deficient embryonic pancreas, defects occur in exocrine and endocrine tissues shortly after differentiation. Pancreatic acini are completely devoid of zymogen granules, and the ER is severely distended. Similar ultrastructural alterations are also observed in salivary glands, but not in liver. Accumulation of proteins in the ER lumen activates the proapoptotic pathway of the unfolded protein response, suggesting a central role for apoptosis in the degeneration of these tissues in SEC23B-deficient embryos. Although maintenance of the secretory pathway should be required by all cells, our findings reveal a surprising tissue-specific dependence on SEC23B for the ER exit of highly abundant cargo, with high levels of SEC23B expression observed in professional secretory tissues. The disparate phenotypes in mouse and human could result from residual SEC23B function associated with the hypomorphic mutations observed in humans, or alternatively, might be explained by a species-specific shift in function between the closely related SEC23 paralogues.
Collapse
Affiliation(s)
- Jiayi Tao
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Min Zhu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - He Wang
- Departments of Internal Medicine and Human Genetics, Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Solomon Afelik
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Matthew P. Vasievich
- Departments of Internal Medicine and Human Genetics, Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Xiao-Wei Chen
- Departments of Internal Medicine and Human Genetics, Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Guojing Zhu
- Departments of Internal Medicine and Human Genetics, Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Jan Jensen
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - David Ginsburg
- Departments of Internal Medicine and Human Genetics, Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48109; and
| | - Bin Zhang
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| |
Collapse
|
45
|
Abstract
Multiple diseases, hematologic and nonhematologic, result from defects in the early secretory pathway. Congenital dyserythropoietic anemia type II (CDAII) and combined deficiency of coagulation factors V and VIII (F5F8D) are the 2 known hematologic diseases that result from defects in the endoplasmic reticulum (ER)-to-Golgi transport system. CDAII is caused by mutations in the SEC23B gene, which encodes a core component of the coat protein complex II (COPII). F5F8D results from mutations in either LMAN1 (lectin mannose-binding protein 1) or MCFD2 (multiple coagulation factor deficiency protein 2), which encode the ER cargo receptor complex LMAN1-MCFD2. These diseases and their molecular pathogenesis are the focus of this review.
Collapse
|
46
|
Mice deficient in LMAN1 exhibit FV and FVIII deficiencies and liver accumulation of α1-antitrypsin. Blood 2011; 118:3384-91. [PMID: 21795745 DOI: 10.1182/blood-2011-05-352815] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The type 1-transmembrane protein LMAN1 (ERGIC-53) forms a complex with the soluble protein MCFD2 and cycles between the endoplasmic reticulum (ER) and the ER-Golgi intermediate compartment (ERGIC). Mutations in either LMAN1 or MCFD2 cause the combined deficiency of factor V (FV) and factor VIII (FVIII; F5F8D), suggesting an ER-to-Golgi cargo receptor function for the LMAN1-MCFD2 complex. Here we report the analysis of LMAN1-deficient mice. Levels of plasma FV and FVIII, and platelet FV, are all reduced to ∼ 50% of wild-type in Lman1(-/-) mice, compared with the 5%-30% levels typically observed in human F5F8D patients. Despite previous reports identifying cathepsin C, cathepsin Z, and α1-antitrypsin as additional potential cargoes for LMAN1, no differences were observed between wild-type and Lman1(-/-) mice in the levels of cathepsin C and cathepsin Z in liver lysates or α1-antitrypsin levels in plasma. LMAN1 deficiency had no apparent effect on COPII-coated vesicle formation in an in vitro assay. However, the ER in Lman1(-/-) hepatocytes is slightly distended, with significant accumulation of α1-antitrypsin and GRP78. An unexpected, partially penetrant, perinatal lethality was observed for Lman1(-/-) mice, dependent on the specific inbred strain genetic background, suggesting a potential role for other, as yet unidentified LMAN1-dependent cargo proteins.
Collapse
|
47
|
Elmahmoudi H, Wigren E, Laatiri A, Jlizi A, Elgaaied A, Gouider E, Lindqvist Y. Analysis of newly detected mutations in the MCFD2 gene giving rise to combined deficiency of coagulation factors V and VIII. Haemophilia 2011; 17:e923-7. [PMID: 21492322 DOI: 10.1111/j.1365-2516.2011.02529.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Combined deficiency of coagulation factor V (FV) and factor VIII (FVIII) (F5F8D) is a rare autosomal recessive disorder characterized by mild-to-moderate bleeding and reduction in FV and FVIII levels in plasma. F5F8D is caused by mutations in one of two different genes, LMAN1 and MCFD2, which encode proteins that form a complex involved in the transport of FV and FVIII from the endoplasmic reticulum to the Golgi apparatus. Here, we report the identification of a novel mutation Asp89Asn in the MCFD2 gene in a Tunisian patient. In the encoded protein, this mutation causes substitution of a negatively charged aspartate, involved in several structurally important interactions, to an uncharged asparagine. To elucidate the structural effect of this mutation, we performed circular dichroism (CD) analysis of secondary structure and stability. In addition, CD analysis was performed on two missense mutations found in previously reported F5F8D patients. Our results show that all analysed mutant variants give rise to destabilized proteins and highlight the importance of a structurally intact and functional MCFD2 for the efficient secretion of coagulation factors V and VIII.
Collapse
Affiliation(s)
- H Elmahmoudi
- Laboratory of Genetics, Immunology and Human Pathologies, Tunis, Tunisia, Sweden.
| | | | | | | | | | | | | |
Collapse
|