1
|
Yakovleva E, Zhang B. Clinical, Laboratory, Molecular, and Reproductive Aspects of Combined Deficiency of Factors V and VIII. Semin Thromb Hemost 2025; 51:116-127. [PMID: 39209292 PMCID: PMC11839339 DOI: 10.1055/s-0044-1789019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [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, Moscow, Russia
| | - Bin Zhang
- Genomic Medicine Institute, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| |
Collapse
|
2
|
Tavasoli B, Zangooie A, Safdari SM, Hoseinnezhad T, Shabannezhad A, Alikhani A, Salehi Z, Dorgalaleh A. Correlation between Phenotype and Coagulation Factor Activity Level in Rare Bleeding Disorders: A Systematic Review. Semin Thromb Hemost 2025; 51:180-195. [PMID: 39638319 DOI: 10.1055/s-0044-1800832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Rare bleeding disorders (RBDs) represent 3 to 5% of congenital bleeding disorders and are primarily inherited in an autosomal recessive manner, with increased prevalence in consanguineous populations. Clinically, RBDs can be accompanied by mild to severe bleeding episodes, often assessed using bleeding assessment tools (BATs) such as the International Society on Thrombosis and Hemostasis (ISTH)-BAT. However, the correlation between bleeding severity and coagulation factor activity levels remains inconsistent. This systematic review investigates this relationship to enhance understanding and improve management strategies for patients with RBD. This review adhered to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and was registered with the International Prospective Register for Systematic Reviews (PROSPERO) (CRD42024504537). Using the PICO (Population, Intervention, Comparator, and Outcomes) framework, the study focused on RBD patients to explore the correlation between coagulation factor activity levels and bleeding severity. A comprehensive search was conducted across PubMed, Scopus, and Web of Science until April 1, 2024, with data extracted on bleeding severity, phenotype, and coagulation factor activity levels. The analysis highlights complex and often inconsistent relationships between coagulation factor levels and the severity of bleeding. In cases of fibrinogen deficiency, three out of four studies (n = 73 of 111 cases, 66%) demonstrated a moderate to strong correlation between fibrinogen levels and bleeding severity. In prothrombin deficiency, one of two studies (n = 16 of 29 cases, 55%) found a strong correlation between FII levels and bleeding severity. Four of six studies (n = 106 of 139 cases, 76%) in FV deficiency found a weak or no correlation between factor activity and bleeding severity. In combined FV and FVIII deficiency, two of three studies (n = 26 of 60 cases, 43%) found a significant correlation between factor activity and bleeding severity. In FVII deficiency, four (of nine) studies with a study population of 325 patients (65%) found a weak correlation between factor activity and severity of bleeding. Almost all studies (five of six studies, n = 114 of 118 patients, 97%) in FX deficiency revealed a strong correlation between FX levels and bleeding severity. In FXI deficiency, most studies (five of seven studies, n = 254 patients, 93%) found a weak or no correlation between factor activity and bleeding severity or symptoms. For FXIII deficiency, there was a moderate to strong correlation between FXIII activity and bleeding severity in all three studies (n = 61 patients). In conclusion, despite current controversies, this review highlights a moderate or strong correlation between factor activity and bleeding severity in fibrinogen, FX, and FXIII deficiencies, but no correlation or weak correlation for FV, FVII, and FXI deficiencies. Further prospective studies with standardized BATs on a large number of patients are needed to better understand these relationships and optimize patient management.
Collapse
Affiliation(s)
- Behnaz Tavasoli
- Department of Hematology and Blood Transfusion, School of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Alireza Zangooie
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Seyed Mehrab Safdari
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Taraneh Hoseinnezhad
- Department of Hematology and Blood Transfusion, School of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Ashkan Shabannezhad
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Amirreza Alikhani
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
3
|
Franchini M, Focosi D. Clinical, Laboratory, and Molecular Aspects of Factor V Deficiency. Semin Thromb Hemost 2025; 51:111-115. [PMID: 39191405 DOI: 10.1055/s-0044-1789021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Factor V (FV) is a glycoprotein that plays a pivotal role in hemostasis, being involved in coagulant and anticoagulant pathways. Congenital FV deficiency is a rare bleeding disorder with an incidence of 1 per million live births, considering the most severe homozygous form. FV deficiency is diagnosed using routine coagulation tests and FV activity assays. Several mutations, including missense, nonsense, and frameshift, have been detected in the F5 gene. Clinical symptoms are variable, ranging from mild ecchymoses and mucosal bleeding to life-threatening intracranial hemorrhage. The mainstay of treatment includes fresh-frozen plasma, preferentially virus-inactivated. In this narrative review, we provide an update of the main laboratory, molecular, clinical, and therapeutic features of inherited FV deficiency.
Collapse
Affiliation(s)
- Massimo Franchini
- Department of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| |
Collapse
|
4
|
Mitani S, Hosoda C, Onodera Y, Takabayashi Y, Sakata A, Shima M, Tatsumi K. Efficient generation of liver sinusoidal endothelial-like cells secreting coagulation factor VIII from human induced pluripotent stem cells. Mol Ther Methods Clin Dev 2024; 32:101355. [PMID: 39559558 PMCID: PMC11570519 DOI: 10.1016/j.omtm.2024.101355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 10/15/2024] [Indexed: 11/20/2024]
Abstract
Liver sinusoidal endothelial cells (LSECs) and LSEC progenitor cells (LPCs) derived from human pluripotent stem cells (PSCs) are expected as valuable cell sources for the development of cell therapy for hemophilia A, a congenital deficiency of coagulation factor VIII (FVIII), as LSECs are responsible for FVIII production. However, there is room for improvement in the efficiency of LSEC and LPC differentiation from human PSCs. In this study, we sought to optimize the method of mesoderm differentiation induction, the initial step of LSEC differentiation from human PSCs, to efficiently induce LSEC-like cells capable of secreting FVIII from human induced pluripotent stem cells (iPSCs). Following optimization of the concentration and stimulation period of CHIR99021 (glycogen synthase kinase 3β inhibitor), bone morphogenetic protein 4, fibroblast growth factor 2, and Activin A in the mesoderm induction step, approximately 65% and 54% of cells differentiated into LPCs and LSEC-like cells, respectively. Furthermore, we observed substantial FVIII protein secretion from LSEC-like cells in vitro. In conclusion, we established an efficient method for obtaining LPCs and functional LSEC-like cells from human iPSCs in vitro.
Collapse
Affiliation(s)
- Seiji Mitani
- Advanced Medical Science of Thrombosis and Hemostasis, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Chihiro Hosoda
- Advanced Medical Science of Thrombosis and Hemostasis, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Yu Onodera
- Advanced Medical Science of Thrombosis and Hemostasis, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Yoko Takabayashi
- Advanced Medical Science of Thrombosis and Hemostasis, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Asuka Sakata
- Medicinal Biology of Thrombosis and Hemostasis, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Midori Shima
- Medicinal Biology of Thrombosis and Hemostasis, Nara Medical University, Kashihara, Nara 634-8521, Japan
| | - Kohei Tatsumi
- Advanced Medical Science of Thrombosis and Hemostasis, Nara Medical University, Kashihara, Nara 634-8521, Japan
- Medicinal Biology of Thrombosis and Hemostasis, Nara Medical University, Kashihara, Nara 634-8521, Japan
| |
Collapse
|
5
|
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; 57:1349-1362. [PMID: 39222205 PMCID: PMC11645327 DOI: 10.1007/s11239-024-03034-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [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
|
6
|
Rezigue H, Chamouni P, Fretigny M, Barbay V, Le Cam-Duchez V, Bobee V, Lanne S, Dumesnil C, Vinciguerra C, Schneider P, Jourdy Y. Heterozygous large deletion mimicking homozygous substitution in MCFD2 in a patient with combined Factor V and Factor VIII deficiency. Haemophilia 2024; 30:1089-1091. [PMID: 38764219 DOI: 10.1111/hae.15038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 05/21/2024]
Affiliation(s)
- Hamdi Rezigue
- Service d'hématologie biologique, Hospices Civils de Lyon, Groupe Hospitalier Est, Bron, France
- Université Claude Bernard Lyon 1, UR4609 Hémostase et thrombose, Lyon, France
| | - Pierre Chamouni
- Université Claude Bernard Lyon 1, UR4609 Hémostase et thrombose, Lyon, France
| | - Mathilde Fretigny
- Service d'hématologie biologique, Hospices Civils de Lyon, Groupe Hospitalier Est, Bron, France
| | - Virginie Barbay
- CHU de Rouen, Hôpital Charles-Nicolle, Centre de ressources et de compétences - Maladies hémorragiques constitutionnelles, Rouen, France
- Service d'hématologie biologique, CHU de Rouen, Hôpital Charles Nicolle, Rouen, France
| | - Véronique Le Cam-Duchez
- CHU de Rouen, Hôpital Charles-Nicolle, Centre de ressources et de compétences - Maladies hémorragiques constitutionnelles, Rouen, France
- Service d'hématologie biologique, CHU de Rouen, Hôpital Charles Nicolle, Rouen, France
| | - Victor Bobee
- Service d'hématologie biologique, CHU de Rouen, Hôpital Charles Nicolle, Rouen, France
| | - Simon Lanne
- Laboratoire Defrance, Le Petit Quevilly, France
| | | | - Christine Vinciguerra
- Service d'hématologie biologique, Hospices Civils de Lyon, Groupe Hospitalier Est, Bron, France
- Université Claude Bernard Lyon 1, UR4609 Hémostase et thrombose, Lyon, France
| | - Pascale Schneider
- Service d'immuno-hémato-oncologie pédiatrique, CHU de Rouen, Hôpital Charles-Nicolle, Rouen, France
| | - Yohann Jourdy
- Service d'hématologie biologique, Hospices Civils de Lyon, Groupe Hospitalier Est, Bron, France
- Université Claude Bernard Lyon 1, UR4609 Hémostase et thrombose, Lyon, France
| |
Collapse
|
7
|
Gangadaran N, Sekar MD, Rangarajan V, Manivannan P. Rare Coagulopathies in Hematologic Spotlight: Isolated Factor V Deficiency and Combined Factor V and VIII Deficiency. Int J Appl Basic Med Res 2024; 14:142-146. [PMID: 38912355 PMCID: PMC11189262 DOI: 10.4103/ijabmr.ijabmr_67_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 06/25/2024] Open
Abstract
Rare coagulation disorders pose significant diagnostic challenges emphasizing the importance of clinical vigilance and meticulous hemostatic workup for accurate diagnosis and timely management. We present two cases of exceptionally uncommon coagulopathies - isolated factor V deficiency (F5D) and combined factor V and VIII deficiency (F5F8D). Case 1 features a 24-year-old woman incidentally diagnosed with severe F5D during routine preoperative evaluation for an ovarian cyst. Despite the absence of any reported bleeding manifestations, a timely and accurate diagnosis was rendered. Perioperative management with fresh frozen plasma and postoperative monitoring ensured favorable surgical outcomes. Case 2 features a 10-year-old male presenting with prolonged gum bleeding. Following systematic hemostatic workup, a diagnosis of F5F8D was rendered, thereby guiding optimal therapeutic interventions. We herein aim to contribute valuable insights into the understanding of coagulation physiology and the diagnostic intricacies and management strategies of rare coagulation disorders.
Collapse
Affiliation(s)
- Nandhini Gangadaran
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Mithraa Devi Sekar
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Vidhyalakshmi Rangarajan
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Prabhu Manivannan
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| |
Collapse
|
8
|
Watanabe S, Kise Y, Yonezawa K, Inoue M, Shimizu N, Nureki O, Inaba K. Structure of full-length ERGIC-53 in complex with MCFD2 for cargo transport. Nat Commun 2024; 15:2404. [PMID: 38493152 PMCID: PMC10944485 DOI: 10.1038/s41467-024-46747-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 03/07/2024] [Indexed: 03/18/2024] Open
Abstract
ERGIC-53 transports certain subsets of newly synthesized secretory proteins and membrane proteins from the endoplasmic reticulum to the Golgi apparatus. Despite numerous structural and functional studies since its identification, the overall architecture and mechanism of action of ERGIC-53 remain unclear. Here we present cryo-EM structures of full-length ERGIC-53 in complex with its functional partner MCFD2. These structures reveal that ERGIC-53 exists as a homotetramer, not a homohexamer as previously suggested, and comprises a four-leaf clover-like head and a long stalk composed of three sets of four-helix coiled-coil followed by a transmembrane domain. 3D variability analysis visualizes the flexible motion of the long stalk and local plasticity of the head region. Notably, MCFD2 is shown to possess a Zn2+-binding site in its N-terminal lid, which appears to modulate cargo binding. Altogether, distinct mechanisms of cargo capture and release by ERGIC- 53 via the stalk bending and metal binding are proposed.
Collapse
Affiliation(s)
- Satoshi Watanabe
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Miyagi, 980-8577, Japan.
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, 980-8577, Japan.
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai, Miyagi, 980-8578, Japan.
| | - Yoshiaki Kise
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kento Yonezawa
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), Tsukuba, Ibaraki, 305-0801, Japan
- Center for Digital Green-innovation, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Mariko Inoue
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Miyagi, 980-8577, Japan
| | - Nobutaka Shimizu
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), Tsukuba, Ibaraki, 305-0801, Japan
| | - Osamu Nureki
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kenji Inaba
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Miyagi, 980-8577, Japan.
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, 980-8577, Japan.
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai, Miyagi, 980-8578, Japan.
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan.
- Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan.
| |
Collapse
|
9
|
Alasmari BG, Alpakra M, Rayees S, Hassanien SS, Almakki ZE. A Novel Variant of the LMAN1 Gene in Combined Factor V and Factor VIII Deficiency in a Saudi Female Child: A Case Report. Cureus 2024; 16:e56858. [PMID: 38659545 PMCID: PMC11040212 DOI: 10.7759/cureus.56858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2024] [Indexed: 04/26/2024] Open
Abstract
Combined factor V and factor VIII deficiency (F5F8D) is an exceedingly rare autosomal recessive disease that causes concomitantly low levels of factor V and factor VIII, leading to mild to moderate bleeding tendencies. Within this disorder, mutations manifest in the lectin mannose-binding protein (LMAN1) or multiple coagulation factor deficiency 2 (MCFD2) genes. This report presents a case of a five-year-old Saudi female child who was referred from an otolaryngology clinic, with an incidental finding of prolonged prothrombin time (PT), international normalized ratio (INR), and activated partial thromboplastin time (aPTT) detected during routine preoperative investigations for tonsillectomy, prompting further investigations. There was no prior history of bleeding symptoms in the patient. She was discovered to have low assays of factor V and factor VIII on subsequent investigations. Whole exome sequencing revealed the novel homozygous mutation c.604C>T in the LMAN1 gene, validating the diagnosis of F5F8D.
Collapse
Affiliation(s)
- Badriah G Alasmari
- Department of Pediatrics, Armed Forces Hospital Southern Region, Khamis Mushait, SAU
| | - Mohammed Alpakra
- Department of Hematology and Oncology, Armed Forces Hospital Southern Region, Khamis Mushait, SAU
| | - Syed Rayees
- Department of Pediatrics, Armed Forces Hospital Southern Region, Khamis Mushait, SAU
| | - Sara S Hassanien
- Department of Hematology and Oncology, Armed Forces Hospital Southern Region, Khamis Mushait, SAU
| | - Zainab E Almakki
- Department of Hematology, Maternity and Children Hospital (MCH), Dammam, SAU
| |
Collapse
|
10
|
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
|
11
|
Livanou ME, Matsas A, Valsami S, Papadimitriou DT, Kontogiannis A, Christopoulos P. Clotting Factor Deficiencies as an Underlying Cause of Abnormal Uterine Bleeding in Women of Reproductive Age: A Literature Review. Life (Basel) 2023; 13:1321. [PMID: 37374104 DOI: 10.3390/life13061321] [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: 04/25/2023] [Revised: 05/27/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Clotting Factor deficiencies are rare disorders with variations in clinical presentation and severity of symptoms ranging from asymptomatic to mild to life-threatening bleeding. Thus, they pose a diagnostic and therapeutic challenge, mainly for the primary health care providers, general practitioners, and gynecologists who are more likely to first encounter these patients. An additional diagnostic challenge arises from the variable laboratory presentations, as PT, PTT, and BT are not always affected. The morbidity is higher among women of reproductive age since Abnormal Uterine Bleeding-specifically Heavy Menstrual Bleeding-is one of the most prevalent manifestations of these disorders, and in some cases of severe deficiencies has led to life-threatening episodes of bleeding requiring blood transfusions or even immediate surgical intervention. Physician awareness is important as, in the case of some of these disorders-i.e., Factor XIII deficiency-prophylactic treatment is available and recommended. Although uncommon, the potential for rare bleeding disorders and for hemophilia carrier states should be considered in women with HMB, after more prevalent causes have been excluded. Currently, there is no consensus on the management of women in these instances and it is reliant on the physicians' knowledge.
Collapse
Affiliation(s)
- Maria Effrosyni Livanou
- Second Department of Obstetrics and Gynecology, "Aretaieion" Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, 115 28 Athens, Greece
| | - Alkis Matsas
- Second Department of Obstetrics and Gynecology, "Aretaieion" Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, 115 28 Athens, Greece
| | - Serena Valsami
- Hematology Laboratory-Blood Bank, Aretaieion Hospital, National and Kapodistrian University of Athens, 115 28 Athens, Greece
| | - Dimitrios T Papadimitriou
- Second Department of Obstetrics and Gynecology, "Aretaieion" Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, 115 28 Athens, Greece
| | - Athanasios Kontogiannis
- Second Department of Obstetrics and Gynecology, "Aretaieion" Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, 115 28 Athens, Greece
| | - Panagiotis Christopoulos
- Second Department of Obstetrics and Gynecology, "Aretaieion" Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, 115 28 Athens, Greece
| |
Collapse
|
12
|
Benítez Hidalgo O, Martinez Garcia MF, Corrrales Insa I, Fernández‐Caballero M, Ramírez Orihuela L, Cortina Giner V, Comes Fernández N, Juarez Gimenez JC. VHrare study: Prevalence, clinical features and management of severe rare bleeding disorders in a large cohort. EJHAEM 2023; 4:476-482. [PMID: 37206292 PMCID: PMC10188473 DOI: 10.1002/jha2.664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 02/10/2023] [Indexed: 05/21/2023]
Abstract
INTRODUCTION Rare bleeding disorders (RBD) constitute 5% of total hereditary bleeding disorders, although the number could be higher, due to the presence of undiagnosed asymptomatic patients. The objective of this study was to analyze the prevalence and characteristics of patients with severe RBDs in our area. MATERIAL AND METHODS We analyzed the patients with RBD followed at a tertiary-level hospital between January 2014 and December 2021. RESULTS A total of 101 patients were analyzed, with a median age at diagnosis of 27.67 years (range 0-89), of which 52.47% were male. The most frequent RBD in our population was FVII deficiency. Regarding the diagnostic reason, the most frequent cause was a preoperative test and only 14.8% reported bleeding symptoms at the time of diagnosis. A genetic study was carried out in 63.36% of patients and the most frequent mutation type found was finding a missense mutation. CONCLUSIONS The distribution of RBDs in our centre is similar to the one reported in the literature. The majority of RBDs were diagnosed from a preoperative test and this allowed preventive treatment prior to invasive procedures to avoid bleeding complications. 83% of patients did not have a pathological bleeding phenotype according to ISTH-BAT.
Collapse
Affiliation(s)
- Olga Benítez Hidalgo
- Hematology DepartmentHospital Universitari Vall d'Hebron, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital CampusBarcelonaSpain
- Medicine DepartmentUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Maria Fernanda Martinez Garcia
- Hematology DepartmentHospital Universitari Vall d'Hebron, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital CampusBarcelonaSpain
- Medicine DepartmentUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Irene Corrrales Insa
- Congenital CoagulopatiesBanc de Sang i TeixitsBarcelonaSpain
- Transfusional Medicine, Vall d'Hebron Institut de RecercaUniversitat Autònoma de Barcelona (VHIR‐UAB)BarcelonaSpain
| | - Mariana Fernández‐Caballero
- Laboratory of Haematology, ICO‐Badalona, Germans Trias i Pujol University Hospital, Institut Josep Carreras Contra la LeucemiaUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Lorena Ramírez Orihuela
- Congenital CoagulopatiesBanc de Sang i TeixitsBarcelonaSpain
- Transfusional Medicine, Vall d'Hebron Institut de RecercaUniversitat Autònoma de Barcelona (VHIR‐UAB)BarcelonaSpain
| | | | - Natàlia Comes Fernández
- Congenital CoagulopatiesBanc de Sang i TeixitsBarcelonaSpain
- Transfusional Medicine, Vall d'Hebron Institut de RecercaUniversitat Autònoma de Barcelona (VHIR‐UAB)BarcelonaSpain
| | | |
Collapse
|
13
|
Dorgalaleh A, Bahraini M, Shams M, Parhizkari F, Dabbagh A, Naderi T, Fallah A, Fazeli A, Ahmadi SE, Samii A, Daneshi M, Heydari F, Tabibian S, Tavasoli B, Noroozi-Aghideh A, Tabatabaei T, Gholami MS. Molecular basis of rare congenital bleeding disorders. Blood Rev 2022; 59:101029. [PMID: 36369145 DOI: 10.1016/j.blre.2022.101029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/26/2022] [Accepted: 10/23/2022] [Indexed: 11/11/2022]
Abstract
Rare bleeding disorders (RBDs), including factor (F) I, FII, FV, FVII, combined FV and FVIII (CF5F8), FXI, FXIII and vitamin-K dependent coagulation factors (VKCF) deficiencies, are a heterogeneous group of hemorrhagic disorder with a variable bleeding tendency. RBDs are due to mutation in underlying coagulation factors genes, except for CF5F8 and VKCF deficiencies. FVII deficiency is the most common RBD with >330 variants in the F7 gene, while only 63 variants have been identified in the F2 gene. Most detected variants in the affected genes are missense (>50% of all RBDs), while large deletions are the rarest, having been reported in FVII, FX, FXI and FXIII deficiencies. Most were located in the catalytic and activated domains of FXI, FX, FXIII and prothrombin deficiencies. Understanding the proper molecular basis of RBDs not only can help achieve a timely and cost-effective diagnosis, but also can help to phenotype properties of the disorders.
Collapse
|
14
|
Johnsen JM, Fletcher SN, Dove A, McCracken H, Martin BK, Kircher M, Josephson NC, Shendure J, Ruuska SE, Valentino LA, Pierce GF, Watson C, Cheng D, Recht M, Konkle BA. Results of genetic analysis of 11 341 participants enrolled in the My Life, Our Future hemophilia genotyping initiative in the United States. J Thromb Haemost 2022; 20:2022-2034. [PMID: 35770352 DOI: 10.1111/jth.15805] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 06/21/2022] [Accepted: 06/25/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Hemophilia A (HA) and hemophilia B (HB) are rare inherited bleeding disorders. Although causative genetic variants are clinically relevant, in 2012 only 20% of US patients had been genotyped. OBJECTIVES My Life, Our Future (MLOF) was a multisector cross-sectional US initiative to improve our understanding of hemophilia through widespread genotyping. METHODS Subjects and potential genetic carriers were enrolled at US hemophilia treatment centers (HTCs). Bloodworks performed genotyping and returned results to providers. Clinical data were abstracted from the American Thrombosis and Hemostasis Network dataset. Community education was provided by the National Hemophilia Foundation. RESULTS From 2013 to 2017, 107 HTCs enrolled 11 341 subjects (68.8% male, 31.2% female) for testing for HA (n = 8976), HB (n = 2358), HA/HB (n = 3), and hemophilia not otherwise specified (n = 4). Variants were detected in most male patients (98.2%% HA, 98.1% HB). 1914 unique variants were found (1482 F8, 431 F9); 744 were novel (610 F8, 134 F9). Inhibitor data were available for 6986 subjects (5583 HA; 1403 HB). In severe HA, genotypes with the highest inhibitor rates were large deletions (77/80), complex intron 22 inversions (9/17), and no variant found (7/14). In severe HB, the highest rates were large deletions (24/42). Inhibitors were reported in 27.3% of Black versus 16.2% of White patients. CONCLUSIONS The findings of MLOF are reported, the largest hemophilia genotyping project performed to date. The results support the need for comprehensive genetic approaches in hemophilia. This effort has contributed significantly towards better understanding variation in the F8 and F9 genes in hemophilia and risks of inhibitor formation.
Collapse
Affiliation(s)
- Jill M Johnsen
- Research Institute, Bloodworks, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Angela Dove
- Research Institute, Bloodworks, Seattle, Washington, USA
| | | | - Beth K Martin
- Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Martin Kircher
- Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Neil C Josephson
- Research Institute, Bloodworks, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Jay Shendure
- Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Sarah E Ruuska
- Research Institute, Bloodworks, Seattle, Washington, USA
| | | | | | - Crystal Watson
- American Thrombosis and Hemostasis Network, Rochester, New York, USA
| | - Dunlei Cheng
- American Thrombosis and Hemostasis Network, Rochester, New York, USA
| | - Michael Recht
- American Thrombosis and Hemostasis Network, Rochester, New York, USA
| | - Barbara A Konkle
- Research Institute, Bloodworks, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
15
|
Pablo-Moreno JAD, Serrano LJ, Revuelta L, Sánchez MJ, Liras A. The Vascular Endothelium and Coagulation: Homeostasis, Disease, and Treatment, with a Focus on the Von Willebrand Factor and Factors VIII and V. Int J Mol Sci 2022; 23:ijms23158283. [PMID: 35955419 PMCID: PMC9425441 DOI: 10.3390/ijms23158283] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 11/27/2022] Open
Abstract
The vascular endothelium has several important functions, including hemostasis. The homeostasis of hemostasis is based on a fine balance between procoagulant and anticoagulant proteins and between fibrinolytic and antifibrinolytic ones. Coagulopathies are characterized by a mutation-induced alteration of the function of certain coagulation factors or by a disturbed balance between the mechanisms responsible for regulating coagulation. Homeostatic therapies consist in replacement and nonreplacement treatments or in the administration of antifibrinolytic agents. Rebalancing products reestablish hemostasis by inhibiting natural anticoagulant pathways. These agents include monoclonal antibodies, such as concizumab and marstacimab, which target the tissue factor pathway inhibitor; interfering RNA therapies, such as fitusiran, which targets antithrombin III; and protease inhibitors, such as serpinPC, which targets active protein C. In cases of thrombophilia (deficiency of protein C, protein S, or factor V Leiden), treatment may consist in direct oral anticoagulants, replacement therapy (plasma or recombinant ADAMTS13) in cases of a congenital deficiency of ADAMTS13, or immunomodulators (prednisone) if the thrombophilia is autoimmune. Monoclonal-antibody-based anti-vWF immunotherapy (caplacizumab) is used in the context of severe thrombophilia, regardless of the cause of the disorder. In cases of disseminated intravascular coagulation, the treatment of choice consists in administration of antifibrinolytics, all-trans-retinoic acid, and recombinant soluble human thrombomodulin.
Collapse
Affiliation(s)
- Juan A. De Pablo-Moreno
- Department of Genetics, Physiology and Microbiology, School of Biology, Complutense University, 28040 Madrid, Spain; (J.A.D.P.-M.); (L.J.S.)
| | - Luis Javier Serrano
- Department of Genetics, Physiology and Microbiology, School of Biology, Complutense University, 28040 Madrid, Spain; (J.A.D.P.-M.); (L.J.S.)
| | - Luis Revuelta
- Department of Physiology, School of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain;
| | - María José Sánchez
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía, Pablo de Olavide University, 41013 Sevilla, Spain;
| | - Antonio Liras
- Department of Genetics, Physiology and Microbiology, School of Biology, Complutense University, 28040 Madrid, Spain; (J.A.D.P.-M.); (L.J.S.)
- Correspondence:
| |
Collapse
|
16
|
Barg AA, Brutman-Barazani T, Avishai E, Budnik I, Cohen O, Dardik R, Levy-Mendelovich S, Livnat T, Kenet G. Anti-TFPI for hemostasis induction in patients with rare bleeding disorders, an ex vivo thrombin generation (TG) guided pilot study. Blood Cells Mol Dis 2022; 95:102663. [PMID: 35525014 DOI: 10.1016/j.bcmd.2022.102663] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/07/2022] [Accepted: 04/08/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Rare bleeding disorders (RBD) are inherited coagulopathies, whose hemostatic control is based upon replacement therapy. Marstacimab (PF-06741086) is a human monoclonal IgG that targets the Kunitz2 domain of tissue factor pathway inhibitor [TFPI]. Marstacimab is currently in development for bleeding prophylaxis in patients with hemophilia. OBJECTIVES To assess the potential impact of Marstacimab upon thrombin generation (TG) in RBD patients' plasma samples. RESULTS Our cohort included 18 RBD patients, with severe deficiencies: 5 Von Willebrand Disease (VWD) type 3, 4 FVII, 3 FXI, 2 FXIII deficiency and 1 patient with: FX, FV + FVIII, Fibrinogen, combined vitamin K dependent factors' deficiency. Citrated samples from RBD patients were collected and spiked with Marstacimab, TG was measured by calibrated automated thrombogram. Among all patients a reduced baseline TG was observed as compared to controls. Improvement of median (lag time, peak and ETP was observed in Marstacimab spiked samples from 8 min, 99 nM, 1116 nMx min to 5.5 min, 194 nM,1614 nMx min, respectively. None of the values measured among RBD patients exceeded normal controls. CONCLUSION These in vitro data suggest that Marstacimab may serve as a promising approach for restoring the hemostatic balance in various RBD, though potential clinical implications should be further investigated.
Collapse
Affiliation(s)
- Assaf A Barg
- National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel; Amalia Biron Research Institute of Thrombosis and Hemostasis, Sackler School of Medicine, Tel Aviv University, Israel
| | - Tami Brutman-Barazani
- National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel; Amalia Biron Research Institute of Thrombosis and Hemostasis, Sackler School of Medicine, Tel Aviv University, Israel
| | - Einat Avishai
- National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel; Amalia Biron Research Institute of Thrombosis and Hemostasis, Sackler School of Medicine, Tel Aviv University, Israel
| | - Ivan Budnik
- Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), Russia
| | - Omri Cohen
- National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel; Amalia Biron Research Institute of Thrombosis and Hemostasis, Sackler School of Medicine, Tel Aviv University, Israel
| | - Rima Dardik
- National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel; Amalia Biron Research Institute of Thrombosis and Hemostasis, Sackler School of Medicine, Tel Aviv University, Israel
| | - Sarina Levy-Mendelovich
- National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel; Amalia Biron Research Institute of Thrombosis and Hemostasis, Sackler School of Medicine, Tel Aviv University, Israel
| | - Tami Livnat
- National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel; Amalia Biron Research Institute of Thrombosis and Hemostasis, Sackler School of Medicine, Tel Aviv University, Israel
| | - Gili Kenet
- National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel; Amalia Biron Research Institute of Thrombosis and Hemostasis, Sackler School of Medicine, Tel Aviv University, Israel.
| |
Collapse
|
17
|
Development and Characterization of a Factor V-Deficient CRISPR Cell Model for the Correction of Mutations. Int J Mol Sci 2022; 23:ijms23105802. [PMID: 35628611 PMCID: PMC9148015 DOI: 10.3390/ijms23105802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 12/04/2022] Open
Abstract
Factor V deficiency, an ultra-rare congenital coagulopathy, is characterized by bleeding episodes that may be more or less intense as a function of the levels of coagulation factor activity present in plasma. Fresh-frozen plasma, often used to treat patients with factor V deficiency, is a scarcely effective palliative therapy with no specificity to the disease. CRISPR/Cas9-mediated gene editing, following precise deletion by non-homologous end-joining, has proven to be highly effective for modeling on a HepG2 cell line a mutation similar to the one detected in the factor V-deficient patient analyzed in this study, thus simulating the pathological phenotype. Additional CRISPR/Cas9-driven non-homologous end-joining precision deletion steps allowed correction of 41% of the factor V gene mutated cells, giving rise to a newly developed functional protein. Taking into account the plasma concentrations corresponding to the different levels of severity of factor V deficiency, it may be argued that the correction achieved in this study could, in ideal conditions, be sufficient to turn a severe phenotype into a mild or asymptomatic one.
Collapse
|
18
|
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: 11] [Impact Index Per Article: 3.7] [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
|
19
|
Bernal S, Pelaez I, Alias L, Baena M, De Pablo-Moreno JA, Serrano LJ, Camero MD, Tizzano EF, Berrueco R, Liras A. High Mutational Heterogeneity, and New Mutations in the Human Coagulation Factor V Gene. Future Perspectives for Factor V Deficiency Using Recombinant and Advanced Therapies. Int J Mol Sci 2021; 22:9705. [PMID: 34575869 PMCID: PMC8465496 DOI: 10.3390/ijms22189705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/02/2021] [Accepted: 09/04/2021] [Indexed: 02/07/2023] Open
Abstract
Factor V is an essential clotting factor that plays a key role in the blood coagulation cascade on account of its procoagulant and anticoagulant activity. Eighty percent of circulating factor V is produced in the liver and the remaining 20% originates in the α-granules of platelets. In humans, the factor V gene is about 80 kb in size; it is located on chromosome 1q24.2, and its cDNA is 6914 bp in length. Furthermore, nearly 190 mutations have been reported in the gene. Factor V deficiency is an autosomal recessive coagulation disorder associated with mutations in the factor V gene. This hereditary coagulation disorder is clinically characterized by a heterogeneous spectrum of hemorrhagic manifestations ranging from mucosal or soft-tissue bleeds to potentially fatal hemorrhages. Current treatment of this condition consists in the administration of fresh frozen plasma and platelet concentrates. This article describes the cases of two patients with severe factor V deficiency, and of their parents. A high level of mutational heterogeneity of factor V gene was identified, nonsense mutations, frameshift mutations, missense changes, synonymous sequence variants and intronic changes. These findings prompted the identification of a new mutation in the human factor V gene, designated as Jaén-1, which is capable of altering the procoagulant function of factor V. In addition, an update is provided on the prospects for the treatment of factor V deficiency on the basis of yet-to-be-developed recombinant products or advanced gene and cell therapies that could potentially correct this hereditary disorder.
Collapse
Affiliation(s)
- Sara Bernal
- Department of Genetics, Santa Creu i Sant Pau Hospital and IIB Sant Pau, 08041 Barcelona, Spain; (S.B.); (L.A.); (M.B.)
- CIBERER. U-705, 18014 Barcelona, Spain
| | - Irene Pelaez
- Department of Pediatric and Oncohematology, University Hospital Virgen de las Nieves, 18014 Granada, Spain;
| | - Laura Alias
- Department of Genetics, Santa Creu i Sant Pau Hospital and IIB Sant Pau, 08041 Barcelona, Spain; (S.B.); (L.A.); (M.B.)
- CIBERER. U-705, 18014 Barcelona, Spain
| | - Manel Baena
- Department of Genetics, Santa Creu i Sant Pau Hospital and IIB Sant Pau, 08041 Barcelona, Spain; (S.B.); (L.A.); (M.B.)
| | - Juan A. De Pablo-Moreno
- Department of Genetic, Physiology and Microbiology, School of Biology, Complutense University, 28040 Madrid, Spain; (J.A.D.P.-M.); (L.J.S.)
| | - Luis J. Serrano
- Department of Genetic, Physiology and Microbiology, School of Biology, Complutense University, 28040 Madrid, Spain; (J.A.D.P.-M.); (L.J.S.)
| | - M. Dolores Camero
- Association for the Investigation and Cure of Factor V Deficiency, 23002 Jaén, Spain;
| | - Eduardo F. Tizzano
- Department of Clinical and Molecular Genetics, University Hospital Vall d’Hebron and Medicine Genetics Group, Vall d’Hebron Research Institute, 08035 Barcelona, Spain;
| | - Ruben Berrueco
- Pediatric Hematology Department, Hospital Sant Joan de Déu, University of Barcelona and Research Institute Hospital Sant Joan de Déu, 08950 Barcelona, Spain;
| | - Antonio Liras
- Department of Genetic, Physiology and Microbiology, School of Biology, Complutense University, 28040 Madrid, Spain; (J.A.D.P.-M.); (L.J.S.)
| |
Collapse
|
20
|
ATP7A-Regulated Enzyme Metalation and Trafficking in the Menkes Disease Puzzle. Biomedicines 2021; 9:biomedicines9040391. [PMID: 33917579 PMCID: PMC8067471 DOI: 10.3390/biomedicines9040391] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 12/12/2022] Open
Abstract
Copper is vital for numerous cellular functions affecting all tissues and organ systems in the body. The copper pump, ATP7A is critical for whole-body, cellular, and subcellular copper homeostasis, and dysfunction due to genetic defects results in Menkes disease. ATP7A dysfunction leads to copper deficiency in nervous tissue, liver, and blood but accumulation in other tissues. Site-specific cellular deficiencies of copper lead to loss of function of copper-dependent enzymes in all tissues, and the range of Menkes disease pathologies observed can now be explained in full by lack of specific copper enzymes. New pathways involving copper activated lysosomal and steroid sulfatases link patient symptoms usually related to other inborn errors of metabolism to Menkes disease. Additionally, new roles for lysyl oxidase in activation of molecules necessary for the innate immune system, and novel adapter molecules that play roles in ERGIC trafficking of brain receptors and other proteins, are emerging. We here summarize the current knowledge of the roles of copper enzyme function in Menkes disease, with a focus on ATP7A-mediated enzyme metalation in the secretory pathway. By establishing mechanistic relationships between copper-dependent cellular processes and Menkes disease symptoms in patients will not only increase understanding of copper biology but will also allow for the identification of an expanding range of copper-dependent enzymes and pathways. This will raise awareness of rare patient symptoms, and thus aid in early diagnosis of Menkes disease patients.
Collapse
|
21
|
Preisler B, Pezeshkpoor B, Banchev A, Fischer R, Zieger B, Scholz U, Rühl H, Kemkes-Matthes B, Schmitt U, Redlich A, Unal S, Laws HJ, Olivieri M, Oldenburg J, Pavlova A. Familial Multiple Coagulation Factor Deficiencies (FMCFDs) in a Large Cohort of Patients-A Single-Center Experience in Genetic Diagnosis. J Clin Med 2021; 10:jcm10020347. [PMID: 33477601 PMCID: PMC7831305 DOI: 10.3390/jcm10020347] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/05/2021] [Accepted: 01/14/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Familial multiple coagulation factor deficiencies (FMCFDs) are a group of inherited hemostatic disorders with the simultaneous reduction of plasma activity of at least two coagulation factors. As consequence, the type and severity of symptoms and the management of bleeding/thrombotic episodes vary among patients. The aim of this study was to identify the underlying genetic defect in patients with FMCFDs. Methods: Activity levels were collected from the largest cohort of laboratory-diagnosed FMCFD patients described so far. Genetic analysis was performed using next-generation sequencing. Results: In total, 52 FMCFDs resulted from coincidental co-inheritance of single-factor deficiencies. All coagulation factors (except factor XII (FXII)) were involved in different combinations. Factor VII (FVII) deficiency showed the highest prevalence. The second group summarized 21 patients with FMCFDs due to a single-gene defect resulting in combined FV/FVIII deficiency or vitamin K–dependent coagulation factor deficiency. In the third group, nine patients with a combined deficiency of FVII and FX caused by the partial deletion of chromosome 13 were identified. The majority of patients exhibited bleeding symptoms while thrombotic events were uncommon. Conclusions: FMCFDs are heritable abnormalities of hemostasis with a very low population frequency rendering them orphan diseases. A combination of comprehensive screening of residual activities and molecular genetic analysis could avoid under- and misdiagnosis.
Collapse
Affiliation(s)
- Barbara Preisler
- Institute of Experimental Hematology and Transfusion Medicine, University Clinic Bonn, 53127 Bonn, Germany; (B.P.); (B.P.); (H.R.); (J.O.)
| | - Behnaz Pezeshkpoor
- Institute of Experimental Hematology and Transfusion Medicine, University Clinic Bonn, 53127 Bonn, Germany; (B.P.); (B.P.); (H.R.); (J.O.)
| | - Atanas Banchev
- Department of Paediatric Haematology and Oncology, University Hospital “Tzaritza Giovanna—ISUL”, 1527 Sofia, Bulgaria;
| | - Ronald Fischer
- Hemophilia Care Center, SRH Kurpfalzkrankenhaus Heidelberg, 69123 Heidelberg, Germany;
| | - Barbara Zieger
- Department of Pediatrics and Adolescent Medicine, University Medical Center–University of Freiburg, 79106 Freiburg, Germany;
| | - Ute Scholz
- Center of Hemostasis, MVZ Labor Leipzig, 04289 Leipzig, Germany;
| | - Heiko Rühl
- Institute of Experimental Hematology and Transfusion Medicine, University Clinic Bonn, 53127 Bonn, Germany; (B.P.); (B.P.); (H.R.); (J.O.)
| | | | - Ursula Schmitt
- Center of Hemostasis Berlin, 10789 Berlin-Schöneberg, Germany;
| | - Antje Redlich
- Pediatric Oncology Department, Otto von Guericke University Children’s Hospital Magdeburg, 39120 Magdeburg, Germany;
| | - Sule Unal
- Division of Pediatric Hematology Ankara, Hacettepe University, 06100 Ankara, Turkey;
| | - Hans-Jürgen Laws
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University of Duesseldorf, 40225 Duesseldorf, Germany;
| | - Martin Olivieri
- Pediatric Hemostasis and Thrombosis Unit, Department of Pediatrics, Pediatric Hemophilia Centre, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany;
| | - Johannes Oldenburg
- Institute of Experimental Hematology and Transfusion Medicine, University Clinic Bonn, 53127 Bonn, Germany; (B.P.); (B.P.); (H.R.); (J.O.)
| | - Anna Pavlova
- Institute of Experimental Hematology and Transfusion Medicine, University Clinic Bonn, 53127 Bonn, Germany; (B.P.); (B.P.); (H.R.); (J.O.)
- Correspondence: ; Tel.: +49-228-287-19711
| |
Collapse
|
22
|
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: 0.4] [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
|
23
|
Favaloro EJ, Lippi G. Understanding the extent of the diagnostic potential of coagulation factors. Expert Rev Mol Diagn 2020; 20:273-276. [PMID: 31903796 DOI: 10.1080/14737159.2020.1711735] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Emmanuel J Favaloro
- Haematology, Institute of Clinical Pathology and Medical Research (ICPMR), NSW Health Pathology, Westmead Hospital, Westmead, Australia.,Sydney Centres for Thrombosis and Haemostasis, Westmead Hospital, Westmead, Australia
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, University of Verona, Verona, Italy
| |
Collapse
|
24
|
Gierula M, Salles‐Crawley II, Santamaria S, Teraz‐Orosz A, Crawley JTB, Lane DA, Ahnström J. The roles of factor Va and protein S in formation of the activated protein C/protein S/factor Va inactivation complex. J Thromb Haemost 2019; 17:2056-2068. [PMID: 31364267 PMCID: PMC6916587 DOI: 10.1111/jth.14594] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/26/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Activated protein C (APC)-mediated inactivation of factor (F)Va is greatly enhanced by protein S. For inactivation to occur, a trimolecular complex among FVa, APC, and protein S must form on the phospholipid membrane. However, direct demonstration of complex formation has proven elusive. OBJECTIVES To elucidate the nature of the phospholipid-dependent interactions among APC, protein S, and FVa. METHODS We evaluated binding of active site blocked APC to phospholipid-coated magnetic beads in the presence and absence of protein S and/or FVa. The importance of protein S and FV residues were evaluated functionally. RESULTS Activated protein C alone bound weakly to phospholipids. Protein S mildly enhanced APC binding to phospholipid surfaces, whereas FVa did not. However, FVa together with protein S enhanced APC binding (>14-fold), demonstrating formation of an APC/protein S/FVa complex. C4b binding protein-bound protein S failed to enhance APC binding, agreeing with its reduced APC cofactor function. Protein S variants (E36A and D95A) with reduced APC cofactor function exhibited essentially normal augmentation of APC binding to phospholipids, but diminished APC/protein S/FVa complex formation, suggesting involvement in interactions dependent upon FVa. Similarly, FVaNara (W1920R), an APC-resistant FV variant, also did not efficiently incorporate into the trimolecular complex as efficiently as wild-type FVa. FVa inactivation assays suggested that the mutation impairs its affinity for phospholipid membranes and with protein S within the complex. CONCLUSIONS FVa plays a central role in the formation of its inactivation complex. Furthermore, membrane proximal interactions among FVa, APC, and protein S are essential for its cofactor function.
Collapse
Affiliation(s)
| | | | | | | | | | - David A. Lane
- Centre for HaematologyImperial College LondonLondonUK
| | | |
Collapse
|
25
|
Clinical and laboratory diagnosis of rare coagulation disorders (RCDs). Thromb Res 2019; 196:603-608. [PMID: 31515069 DOI: 10.1016/j.thromres.2019.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/17/2022]
Abstract
Rare coagulation disorders (RCDs) are a group of diseases due to coagulation factors deficiency leading to life-long bleeding diathesis. The diagnosis of RCDs is challenging due to the limited knowledge of these disorders and the large heterogeneity of their bleeding patterns. The clinical symptoms of RCDs are extremely diverse in terms of bleeding type, site, severity, age at onset, and duration. The strength of the association between clotting factor activity level in plasma and clinical symptoms is also variable within each RCD. The clinical evaluation of RCDs starts with a detailed collection of clinical history and has been facilitated by bleeding assessment tools, however their effectiveness in diagnosing RCDs requires further investigation. The following laboratory diagnosis of RCDs involves coagulation screening tests, including activated partial thromboplastin time, prothrombin time, and thrombin time. After ruling out the presence of an inhibitor by mixing studies, in case of abnormal results, the specific deficiency is identified by performing one-stage clotting assays using the specific factor-depleted plasmas as substrate. In fibrinogen and FXIII deficiencies coagulation screening tests are not informative, therefore additional tests are needed. Global assays have been developed and are thought to aid in patient management, however, they are not well standardized yet. In addition to outlining the principles of clinical and laboratory diagnosis, this review explores molecular basis of RCDs and laboratory techniques for genetic analysis, and discusses the importance and effectiveness of quality control programs to ensure standardized laboratory results.
Collapse
|
26
|
Deng J, Li D, Mei H, Tang L, Wang HF, Hu Y. Combined deficiency of factors V and VIII in a Chinese family due to a novel nonsense mutation in lectin mannose binding protein 1. Int J Lab Hematol 2019; 42:e7-e9. [PMID: 31162802 DOI: 10.1111/ijlh.13054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/26/2019] [Accepted: 05/03/2019] [Indexed: 11/29/2022]
Affiliation(s)
- Jun Deng
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical and Research Centre of Thrombosis and Haemostasis, Wuhan, China
| | - Dan Li
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Mei
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical and Research Centre of Thrombosis and Haemostasis, Wuhan, China
| | - Liang Tang
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical and Research Centre of Thrombosis and Haemostasis, Wuhan, China
| | - Hua-Fang Wang
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical and Research Centre of Thrombosis and Haemostasis, Wuhan, China
| | - Yu Hu
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Clinical and Research Centre of Thrombosis and Haemostasis, Wuhan, China
| |
Collapse
|
27
|
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: 18] [Impact Index Per Article: 3.0] [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
|
28
|
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: 30] [Impact Index Per Article: 5.0] [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
|
29
|
Abstract
Rare bleeding disorders (RBDs) comprise inherited deficiencies of factors I (fibrinogen), II (prothrombin), V, VII, X, XI, and XIII as well as combined factor V + VIII and vitamin K-dependent factors. They represent 3-5% of all congenital bleeding disorders and are usually transmitted as autosomal recessive traits. These disorders often manifest during childhood and have varied clinical presentations from mucocutaneous bleeding to life-threatening symptoms such as central nervous system and gastrointestinal bleeding. Bleeding manifestations generally vary within the same RBD and may also vary from 1 RBD to the other. Laboratory diagnosis is based on coagulation screening tests and specific factor assays, with molecular techniques providing diagnostic accuracy and enabling prenatal counseling. The approach to treatment of bleeding episodes and invasive procedures needs to be individualized and depends on the severity, frequency and procedure-related risk of bleeding. The first line of treatment of RBDs is replacement of the deficient factor, using specific plasma-derived or recombinant products and using fresh frozen plasma or cryoprecipitate when specific products are not available or in resource-limited countries. Prophylaxis may be considered in individuals with recurrent serious bleeding and especially after life-threatening bleeding episodes. Novel no-replacement strategies promoting hemostasis by through different mechanisms need to be studied in RBDs as alternative therapeutic options.
Collapse
Affiliation(s)
- Shilpa Jain
- Division of Pediatric Hematology-Oncology, Women and Children's Hospital of Buffalo and Hemophilia Center of Western New York, Buffalo, NY, 14209, United States.
| | - Suchitra S Acharya
- Bleeding Disorders and Thrombosis Program, Cohen Children's Medical Center of New York and Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| |
Collapse
|
30
|
Favaloro EJ, Mohammed S, Lippi G. Postanalytical considerations that may improve the diagnosis or exclusion of haemophilia and von Willebrand disease. Haemophilia 2018; 24:849-861. [PMID: 30024638 DOI: 10.1111/hae.13587] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/17/2018] [Indexed: 01/07/2023]
Abstract
von Willebrand disease (VWD) and haemophilia represent the most common inherited or acquired bleeding disorders. However, many laboratories and clinicians may be challenged by their accurate diagnosis or exclusion. Difficulties in diagnosis/exclusion may include analytical issues, where assays occasionally generate an incorrect result (ie representing an analytical error) or have limitations in their measurement range of and/or low analytical sensitivity. Also increasingly recognized is the influence of preanalytical issues on the diagnosis of VWD or haemophilia. Unfortunately, postanalytical considerations are often not well considered in the diagnostic process. Therefore, this narrative review aims to provide an overview of some important postanalytical considerations that may help improve the diagnosis of VWD and haemophilia. This review primarily discusses aspects around reporting of test results. However, we also discuss other less well-recognized postanalytical considerations, including the use of assay ratios to help identify differential diagnoses and then guide further investigation.
Collapse
Affiliation(s)
- Emmanuel J Favaloro
- Diagnostic Haemostasis Laboratory, Department of Haematology, Institute of Clinical Pathology and Medical Research (ICPMR), NSW Health Pathology, Westmead Hospital, Westmead, NSW, Australia.,Sydney Centres for Thrombosis and Haemostasis, Westmead, NSW, Australia
| | - Soma Mohammed
- Diagnostic Haemostasis Laboratory, Department of Haematology, Institute of Clinical Pathology and Medical Research (ICPMR), NSW Health Pathology, Westmead Hospital, Westmead, NSW, Australia
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, University of Verona, Verona, Italy
| |
Collapse
|
31
|
Kesireddy S. Recurrent Ruptured Hemorrhagic Corpus Luteal Cyst in a Known Case of Combined Deficiency of Factor V and VIII. J Obstet Gynaecol India 2018; 68:229-231. [DOI: 10.1007/s13224-017-1028-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 06/12/2017] [Indexed: 11/27/2022] Open
|
32
|
|
33
|
Favaloro EJ, Lippi G. Preanalytical issues that may cause misdiagnosis in haemophilia and von Willebrand disease. Haemophilia 2017; 24:198-210. [DOI: 10.1111/hae.13396] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2017] [Indexed: 12/16/2022]
Affiliation(s)
- E. J. Favaloro
- Diagnostic Haemostasis Laboratory; Department of Haematology; Institute of Clinical Pathology and Medical Research (ICPMR); NSW Health Pathology; Westmead Hospital; Westmead NSW Australia
- Sydney Centres for Thrombosis and Haemostasis; Westmead NSW Australia
| | - G. Lippi
- Section of Clinical Biochemistry; University of Verona; Verona Italy
| |
Collapse
|
34
|
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
|
35
|
Diagnosis and Treatment of von Willebrand Disease and Rare Bleeding Disorders. J Clin Med 2017; 6:jcm6040045. [PMID: 28394285 PMCID: PMC5406777 DOI: 10.3390/jcm6040045] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/04/2017] [Accepted: 04/05/2017] [Indexed: 12/19/2022] Open
Abstract
Along with haemophilia A and B, von Willebrand disease (VWD) and rare bleeding disorders (RBDs) cover all inherited bleeding disorders of coagulation. Bleeding tendency, which can range from extremely severe to mild, is the common symptom. VWD, due to a deficiency and/or abnormality of von Willebrand factor (VWF), represents the most frequent bleeding disorder, mostly inherited as an autosomal dominant trait. The diagnosis may be difficult, based on a bleeding history and different diagnostic assays, which evaluate the pleiotropic functions of VWF. Different treatment options are available for optimal management of bleeding and their prevention, and long-term outcomes are generally good. RBDs are autosomal recessive disorders caused by a deficiency of any other clotting factor, apart from factor XII, and cover roughly 5% of all bleeding disorders. The prevalence of the severe forms can range from 1 case in 500,000 up to 1 in 2–3 million, according to the defect. Diagnosis is based on bleeding history, coagulation screening tests and specific factor assays. A crucial problem in RBDs diagnosis is represented by the non-linear relationship between clinical bleeding severity and residual clotting levels; genetic diagnosis may help in understanding the phenotype. Replacement therapies are differently available for patients with RBDs, allowing the successful treatment of the vast majority of bleeding symptoms.
Collapse
|
36
|
|
37
|
Abstract
Hemophilia A is an inherited bleeding disorder caused by a reduced level of factor VIII coagulant activity (FVIII:C) in blood. Bleeding episodes may occur spontaneously in the severe form of hemophilia or after trauma in the milder forms. It is important that patients are diagnosed correctly, which includes placing them into the correct severity category of the disorder so that appropriate treatment can be given. Diagnosis is made by determination of the amount of FVIII:C in the blood, usually using a one-stage factor VIII:C assay. However, approximately one third of patients with mild or moderate hemophilia will have much lower results by the chromogenic assay, with some of them having normal results by the one-stage assay. The chromogenic factor VIII assay is used in some specialized hemophilia reference centers and is recommended for the diagnosis of mild hemophilia A, as this assay is considered to better reflect the severity status of hemophilia patients than the one-stage assay.
Collapse
Affiliation(s)
- Susan Rodgers
- Haematology Division, IMVS, SA Pathology, Frome Rd., Adelaide, SA, Australia.
| | - Elizabeth Duncan
- Haematology Division, IMVS, SA Pathology, Frome Rd., Adelaide, SA, Australia
| |
Collapse
|
38
|
Olson NJ, Shajani-Yi Z, Cervinski MA, Ornstein DL. Prolonged Bleeding in a 34-Year-Old Man following Oral Surgery. Clin Chem 2016; 62:1676-1677. [PMID: 27899461 DOI: 10.1373/clinchem.2016.264069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 07/26/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Nicholas J Olson
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH; and Geisel School of Medicine, Hanover, NH.
| | - Zahra Shajani-Yi
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH; and Geisel School of Medicine, Hanover, NH
| | - Mark A Cervinski
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH; and Geisel School of Medicine, Hanover, NH
| | - Deborah L Ornstein
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH; and Geisel School of Medicine, Hanover, NH
| |
Collapse
|
39
|
Boender J, Kruip MJHA, Leebeek FWG. A diagnostic approach to mild bleeding disorders. J Thromb Haemost 2016; 14:1507-16. [PMID: 27208505 DOI: 10.1111/jth.13368] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Indexed: 01/02/2023]
Abstract
Mild inherited bleeding disorders are relatively common in the general population. Despite recent advances in diagnostic approaches, mild inherited bleeding disorders still pose a significant diagnostic challenge. Hemorrhagic diathesis can be caused by disorders in primary hemostasis (von Willebrand disease, inherited platelet function disorders), secondary hemostasis (hemophilia A and B, other (rare) coagulant factor deficiencies) and fibrinolysis, and in connective tissue or vascular formation. This review summarizes the currently available diagnostic methods for mild bleeding disorders and their pitfalls, from structured patient history to highly specialized laboratory diagnosis. A comprehensive framework for a diagnostic approach to mild inherited bleeding disorders is proposed.
Collapse
Affiliation(s)
- J Boender
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - M J H A Kruip
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - F W G Leebeek
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
40
|
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: 0.7] [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
|
41
|
Dahlbäck B. Pro- and anticoagulant properties of factor V in pathogenesis of thrombosis and bleeding disorders. Int J Lab Hematol 2016; 38 Suppl 1:4-11. [PMID: 27161771 DOI: 10.1111/ijlh.12508] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2016] [Indexed: 11/29/2022]
Abstract
Factor V (FV) serves an important role in the regulation of blood coagulation, having both pro- and anticoagulant properties. The circulating high molecular weight single-chain FV molecule undergoes a series of proteolytic cleavages during both activation of coagulation and during anticoagulant regulation of coagulation by activated protein C (APC). It is noteworthy that mutations in the factor V gene (F5) either cause thrombosis or bleeding. New insights into the importance and complexity of FV functions have been generated from elucidation of the pathogenic mechanisms of two familial mutations in the F5 gene. The first mutation was identified as a result of the discovery of APC resistance as the most common risk factor for venous thrombosis. The mutation (FV Leiden) predicts the Arg(506) Gln replacement, which impairs the normal regulation of FVa by APC, as the Arg506 site is an important APC cleavage site. In addition, elucidation of APC resistance resulted in the discovery of the anticoagulant APC cofactor activity of FV. The second FV mutation (FV(A2440G) ), identified in a family with an autosomal dominant bleeding disorder, has led to the discovery of an alternative splicing generating a previously unidentified FV isoform (FV-Short), which inhibits coagulation via an unexpected and intriguing mechanism involving the coagulation inhibitor TFPI-α. These are naturally occurring mutations in the F5 gene that have generated new knowledge on the role of FV in regulation of coagulation and the importance of genetic risk factors for thrombosis and bleeding.
Collapse
Affiliation(s)
- Björn Dahlbäck
- Department of Translational Medicine, Lund University, Malmö, Sweden.,Wallenberg Laboratory, University Hospital, Malmö, Sweden
| |
Collapse
|
42
|
Saraste J, Marie M. Intermediate Compartment: A Sorting Station between the Endoplasmic Reticulum and the Golgi Apparatus. ENCYCLOPEDIA OF CELL BIOLOGY 2016. [PMCID: PMC7150006 DOI: 10.1016/b978-0-12-394447-4.20013-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
43
|
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
|
44
|
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: 2.2] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
45
|
Roedersheimer M. Solving the Measurement Problem and then Steppin' Out over the Line Riding the Rarest Italian: Crossing the Streams to Retrieve Stable Bioactivity in Majorana Bound States of Dialy zed Human Platelet Lysates. Open Neurol J 2015; 9:32-44. [PMID: 26191092 PMCID: PMC4503829 DOI: 10.2174/1874205x01509010032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 03/24/2014] [Accepted: 03/25/2014] [Indexed: 11/22/2022] Open
Abstract
Exhaustive dialysis (ED) of lysed human platelets against dilute HCl yields stable angiogenic activity. Dialysis against a constrained external volume, with subsequent relaxation of the separation upon opening the dialysis bag, produces material able to maintain phenotypes and viability of human cells in culture better than ED material. Significant graded changes in MTT viability measurement tracked with external volume. The presence of elements smaller than the MW cutoff, capable of setting up cycling currents initiated by oriented flow of HCl across the membrane, suggests that maturation of bioactivity occurred through establishment of a novel type of geometric phase. These information-rich bound states fit recent descriptions of topological order and Majorana fermions, suggesting relevance in testing Penrose and Hameroff's theory of Orchestrated Objective Reduction, under conditions more general, and on finer scales, than those dependent on tubulin protein. The Berry curvature appears to be a good tool for building a general field theory of physiologic stress dependent on the quantum Hall effect. A new form of geometric phase, and an associated "geometric" quantum Hall effect underlying memory retrieval, dependent on the rate of path traversal and reduction from more than two initial field influences is described.
Collapse
|
46
|
Lippi G, Pasalic L, Favaloro EJ. Detection of mild inherited disorders of blood coagulation: current options and personal recommendations. Expert Rev Hematol 2015; 8:527-42. [DOI: 10.1586/17474086.2015.1039978] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
47
|
Abstract
Despite the worldwide prevalence of rare bleeding disorders (RBDs), knowledge of these conditions and their management is suboptimal; health care professionals often have little diagnostic and treatment experience with variable access to diagnostic modalities required for accurate identification. Therefore, patients often experience morbidity and mortality due to delayed diagnosis. As RBDs represent a small potential commercial market, few, if any, specific therapies exist for these conditions. As a result, affected individuals commonly face delayed diagnosis, incomplete laboratory evaluation, and limited treatment options. Standardization and customization of coagulation assays, full genome sequencing, and global clotting assays will significantly improve diagnosis of patients with RBDs. In addition, new therapeutic modalities, both recombinant and plasma derived, are emerging, at least in developed countries. Registries and clinical trials have demonstrated decreased bleeding and improved outcomes when patients are appropriately diagnosed and properly treated. Expansion and harmonization of international registries has been initiated to correlate genotype, laboratory, and clinical phenotypes including bleeding severity to improve the diagnosis and therapeutic approach. This review focuses on the latest advances in our understanding, diagnosis, and treatment of RBDs.
Collapse
|