1
|
Chuang YM, Dong Y, Stone H, Abouneameh S, Tang XD, Raduwan H, Dimopoulos G, Fikrig E. Anopheles gambiae lacking AgTRIO probe inefficiently on a mammalian host. Cell Rep 2024; 43:114600. [PMID: 39126653 PMCID: PMC11407849 DOI: 10.1016/j.celrep.2024.114600] [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: 05/03/2024] [Revised: 07/02/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Malaria is initiated as Plasmodium sporozoites are injected into the dermis when an infected mosquito probes on a vertebrate host for a blood meal. Factors in the mosquito saliva, such as AgTRIO, can alter the ability of Anopheles gambiae to transmit Plasmodium. We therefore used CRISPR-Cas9-mediated genome editing to generate AgTRIO knockout (KO) A. gambiae and examined the ability of these mosquitoes to probe on a vertebrate host. AgTRIO KO mosquitoes showed a diminished host probing capacity and required repetitive probing to locate a blood resource to complete a blood meal. This increased probing resulted in enhanced Plasmodium transmission to the vertebrate host. Our data demonstrate the importance of the A. gambiae saliva protein AgTRIO in probing and its influence on the ability of mosquitoes to transmit malaria.
Collapse
Affiliation(s)
- Yu-Min Chuang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Helen Stone
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Selma Abouneameh
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Xu-Dong Tang
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China
| | - Hamidah Raduwan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
2
|
Hagan CE, Snyder AG, Headley M, Oberst A. Apoptotic cells promote circulating tumor cell survival and metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595217. [PMID: 38826267 PMCID: PMC11142129 DOI: 10.1101/2024.05.21.595217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
During tumor progression and especially following cytotoxic therapy, cell death of both tumor and stromal cells is widespread. Despite clinical observations that high levels of apoptotic cells correlate with poorer patient outcomes, the physiological effects of dying cells on tumor progression remain incompletely understood. Here, we report that circulating apoptotic cells robustly enhance tumor cell metastasis to the lungs. Using intravenous metastasis models, we observed that the presence of apoptotic cells, but not cells dying by other mechanisms, supports circulating tumor cell (CTC) survival following arrest in the lung vasculature. Apoptotic cells promote CTC survival by recruiting platelets to the forming metastatic niche. Apoptotic cells externalize the phospholipid phosphatidylserine to the outer leaflet of the plasma membrane, which we found increased the activity of the coagulation initiator Tissue Factor, thereby triggering the formation of platelet clots that protect proximal CTCs. Inhibiting the ability of apoptotic cells to induce coagulation by knocking out Tissue Factor, blocking phosphatidylserine, or administering the anticoagulant heparin abrogated the pro-metastatic effect of apoptotic cells. This work demonstrates a previously unappreciated role for apoptotic cells in facilitating metastasis by establishing CTC-supportive emboli, and suggests points of intervention that may reduce the pro-metastatic effect of apoptotic cells. GRAPHICAL ABSTRACT
Collapse
|
3
|
Tsikis ST, Hirsch TI, Klouda T, Fligor SC, Pan A, Joiner MM, Wang SZ, Quigley M, Devietro A, Mitchell PD, Kishikawa H, Yuan K, Puder M. Direct thrombin inhibitors fail to reverse the negative effects of heparin on lung growth and function after murine left pneumonectomy. Am J Physiol Lung Cell Mol Physiol 2024; 326:L213-L225. [PMID: 38113296 PMCID: PMC11280676 DOI: 10.1152/ajplung.00096.2023] [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: 03/28/2023] [Revised: 10/20/2023] [Accepted: 12/18/2023] [Indexed: 12/21/2023] Open
Abstract
Neonates with congenital diaphragmatic hernia (CDH) frequently require cardiopulmonary bypass and systemic anticoagulation. We previously demonstrated that even subtherapeutic heparin impairs lung growth and function in a murine model of compensatory lung growth (CLG). The direct thrombin inhibitors (DTIs) bivalirudin and argatroban preserved growth in this model. Although DTIs are increasingly used for systemic anticoagulation clinically, patients with CDH may still receive heparin. In this experiment, lung endothelial cell proliferation was assessed following treatment with heparin-alone or mixed with increasing concentrations of bivalirudin or argatroban. The effects of subtherapeutic heparin with or without DTIs in the CLG model were also investigated. C57BL/6J mice underwent left pneumonectomy and subcutaneous implantation of osmotic pumps. Pumps were preloaded with normal saline, bivalirudin, or argatroban; treated animals received daily intraperitoneal low-dose heparin. In vitro, heparin-alone decreased endothelial cell proliferation and increased apoptosis. The effect of heparin on proliferation, but not apoptosis, was reversed by the addition of bivalirudin and argatroban. In vivo, low-dose heparin decreased lung volume compared with saline-treated controls. All three groups that received heparin demonstrated decreased lung function on pulmonary function testing and impaired exercise performance on treadmill tolerance testing. These findings correlated with decreases in alveolarization, vascularization, angiogenic signaling, and gene expression in the heparin-exposed groups. Together, these data suggest that bivalirudin and argatroban fail to reverse the inhibitory effects of subtherapeutic heparin on lung growth and function. Clinical studies on the impact of low-dose heparin with DTIs on CDH outcomes are warranted.NEW & NOTEWORTHY Infants with pulmonary hypoplasia frequently require cardiopulmonary bypass and systemic anticoagulation. We investigate the effects of simultaneous exposure to heparin and direct thrombin inhibitors (DTIs) on lung growth and pulmonary function in a murine model of compensatory lung growth (CGL). Our data suggest that DTIs fail to reverse the inhibitory effects of subtherapeutic heparin on lung growth and function. Clinical studies on the impact of heparin with DTIs on clinical outcomes are thus warranted.
Collapse
Affiliation(s)
- Savas T Tsikis
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Thomas I Hirsch
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Timothy Klouda
- Division of Pulmonary Medicine, Boston Children's Hospital, Boston, Massachusetts, United States
| | - Scott C Fligor
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Amy Pan
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Malachi M Joiner
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Sarah Z Wang
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Mikayla Quigley
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Angela Devietro
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Paul D Mitchell
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, Massachusetts, United States
| | - Hiroko Kishikawa
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Ke Yuan
- Division of Pulmonary Medicine, Boston Children's Hospital, Boston, Massachusetts, United States
| | - Mark Puder
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
4
|
Fabunmi OA, Dludla PV, Nkambule BB. High-fat diet promotes coagulation and endothelial activation in Sprague Dawley rats: Short-term effects of combined oral contraceptives. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36:60-70. [PMID: 37949735 DOI: 10.1016/j.arteri.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/21/2023] [Accepted: 10/09/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Combined oral contraceptives (COCs), use in individuals are associated with increased risk of thrombotic events. This highlights the significance of assessing the impact of COC on promoting coagulation and endothelial activation in high-fat diet (HFD)-fed Sprague Dawley rats. METHODS Twenty (20) five-weeks-old female Sprague Dawley rats weighing between 150 and 200g were subjected to both LFD and HFD-feeding for 8-weeks to determine its influence on basic metabolic status, hemostatic profile, hemodynamic parameters (blood pressure and heart rate), as well as selected biomarkers of coagulation (tissue factor and D-dimer) and endothelial activation (Von Willebrand factor and nitric oxide). Thereafter HFD-fed animals were treated with receive high dose combined oral contraceptive (HCOC) and low dose combine oral contraceptive (LCOC) for 6 weeks. RESULTS Our results showed that beyond weight gain, HFD-feeding was associated with hyperglycemia, increased mean arterial pressure, and reduced nitric oxide levels when compared with LFD group (p<0.05). Interestingly, treatment with high dose of COC for 6-weeks did not significantly alter atherothrombotic markers (p>0.05). However, this study is not without limitation as regulation of these markers remains to be confirmed within the cardiac tissues or endothelial cells of these animals. CONCLUSION HFD-feeding orchestrate the concomitant release of pro-coagulants and endothelial activation markers in rats leading to haemostatic imbalance and endothelial dysfunction. Short-term treatment with COC shows no detrimental effects in these HFD-fed rats. Although in terms of clinical relevance, our findings depict the notion that the risk of CVD in association with COC may depend on the dosage and duration of use among other factors especially in certain conditions. However, additional studies are required to confirm these findings, especially long-term effects of this treatment within the cardiac tissues or endothelial cells of these animals in certain conditions relating to postmenopausal state.
Collapse
Affiliation(s)
- Oyesanmi A Fabunmi
- School of Laboratory Medicine and Medical Sciences (SLMMS), College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; Department of Physiology, College of Medicine, Ekiti State University, Ado-Ekiti 5363, Nigeria
| | - Phiwayinkosi V Dludla
- Cochrane South Africa, South African Medical Research Council, Tygerberg 7505, South Africa; Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3880, South Africa
| | - Bongani B Nkambule
- School of Laboratory Medicine and Medical Sciences (SLMMS), College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa.
| |
Collapse
|
5
|
Wang Y, Lindstam M, Hwang D, Jedlina L, Liu M. Therapeutic Effects of a Novel Aptamer on Coronaviral Infection-Induced Lung Injury and Systemic Inflammatory Responses. Cells 2024; 13:422. [PMID: 38474386 PMCID: PMC10931054 DOI: 10.3390/cells13050422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/08/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Coronaviral infection-induced acute lung injury has become a major threat to public health, especially through the ongoing pandemic of COVID-19. Apta-1 is a newly discovered Aptamer that has anti-inflammatory effects on systemic septic responses. The therapeutic effects of Apta-1 on coronaviral infection-induced acute lung injury and systemic responses were evaluated in the present study. METHODS Female A/J mice (at 12-14 weeks of age) were challenged with murine hepatitis virus 1 (MHV-1), a coronavirus, at 5000 PFU intranasally, followed by Apta-1 intravenously administered (100 mg/kg, twice) 1.5 h or 2 days after viral delivery. Animals were sacrificed at Day 2 or Day 4. Lung tissues were examined with H&E, immunohistochemistry staining, and western blotting. RT-qPCR was used for cytokine gene expression. Serum and plasma were collected for laboratory assessments. RESULTS Apta-1 treatment reduced viral titers, prevented MHV-1-induced reduction of circulating blood volume and hemolysis, reduced alveolar space hemorrhage, and protease-activated receptor 1 (PAR-1) cleavage. Apta-1 treatment also significantly reduced chemokine (MKC, MCP-1, and RANTES) levels, as well as AST, ALT, total bilirubin, and reduced unconjugated bilirubin levels in the serum. CONCLUSION Apta-1 showed therapeutic benefits in coronaviral infection-induced hemorrhage and PAR-1 cleavage in the lung. It also has anti-inflammatory effects systemically.
Collapse
Affiliation(s)
- Yingchun Wang
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada;
| | | | - David Hwang
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | | | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada;
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada;
- Departments of Surgery, Medicine, and Physiology, Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
6
|
Qiao Y, Xiao J, He T, Wang R, Xu Y, Wei Y, Wang J, Hu R, Li Z. Predictive value of coagulation function, alpha-fetoprotein and placental growth factor in patients with perilous placenta previa. Am J Transl Res 2024; 16:567-576. [PMID: 38463595 PMCID: PMC10918133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/15/2023] [Indexed: 03/12/2024]
Abstract
OBJECTIVE To analyze the predictive value of coagulation function, alpha-fetoprotein (AFP) and placental growth factor (PIGF) for postpartum hemorrhage in patients with perilous placenta previa (PPP). METHODS The clinical data of 104 PPP patients were retrospectively analyzed. The patients were divided into a hemorrhage group (n=68) and a non-hemorrhage group (n=36). A total of 55 healthy pregnant women were recruited as controls. The coagulation function, AFP and PIGF were compared between the three groups. Multivariate logistic regression was performed to determine independent risk factors for hemorrhage. RESULTS PT, TT, APTT, FIB and AFP were significantly higher while PIGF was lower in the PPP group than the control group (all P<0.05). Placental adhesion (OR 3.924, 95% CI 1.389-11.083, P=0.01), anterior placenta (OR 4.583, 95% CI 1.589-13.22, P=0.005), AFP (OR 0.208, 95% CI 0.068-0.635, P=0.006) and PIGF (OR 3.963, 95% CI 1.385-11.34, P=0.01) were independent risk factors for hemorrhage. CONCLUSION Coagulation function, AFP and PIGF could predict postpartum hemorrhage in PPP patients.
Collapse
Affiliation(s)
- Yuan Qiao
- Obstetrics and Gynecology Intensive Care Unit, Northwest Women’s and Children’s HospitalNo. 1616 Yanxiang Road, Xi’an 710061, Shaanxi, China
| | - Jin Xiao
- Obstetrics and Gynecology Intensive Care Unit, Northwest Women’s and Children’s HospitalNo. 1616 Yanxiang Road, Xi’an 710061, Shaanxi, China
| | - Tongqiang He
- Obstetrics and Gynecology Intensive Care Unit, Northwest Women’s and Children’s HospitalNo. 1616 Yanxiang Road, Xi’an 710061, Shaanxi, China
| | - Rui Wang
- Department of Obstetrics, Northwest Women’s and Children’s HospitalNo. 1616 Yanxiang Road, Xi’an 710061, Shaanxi, China
| | - Yehong Xu
- Department of Obstetrics, Northwest Women’s and Children’s HospitalNo. 1616 Yanxiang Road, Xi’an 710061, Shaanxi, China
| | - Yang Wei
- Obstetrics and Gynecology Intensive Care Unit, Northwest Women’s and Children’s HospitalNo. 1616 Yanxiang Road, Xi’an 710061, Shaanxi, China
| | - Jun Wang
- Obstetrics and Gynecology Intensive Care Unit, Northwest Women’s and Children’s HospitalNo. 1616 Yanxiang Road, Xi’an 710061, Shaanxi, China
| | - Rui Hu
- Obstetrics and Gynecology Intensive Care Unit, Northwest Women’s and Children’s HospitalNo. 1616 Yanxiang Road, Xi’an 710061, Shaanxi, China
| | - Zhibin Li
- Department of Obstetrics, Northwest Women’s and Children’s HospitalNo. 1616 Yanxiang Road, Xi’an 710061, Shaanxi, China
| |
Collapse
|
7
|
Wang H, Mazzocca A, Gao P. Cadherin dysregulation in gastric cancer: insights into gene expression, pathways, and prognosis. J Gastrointest Oncol 2023; 14:2064-2082. [PMID: 37969819 PMCID: PMC10643585 DOI: 10.21037/jgo-23-700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/12/2023] [Indexed: 11/17/2023] Open
Abstract
Background The Cadherin gene family holds immense significance in maintaining the integrity and functionality of stomach tissues, playing crucial roles in cell-cell adhesion, cell migration and differentiation. Dysregulation of cadherin expression and function has been closely associated with various gastric diseases, particularly gastric cancer (GC). Understanding the regulation and clinical implications of cadherin genes in GC is essential to improve our knowledge and to identify new potential prognostic markers and therapeutic targets. Methods In this study, we provide an overview on the role of cadherin family genes in GC using bioinformatics analysis. We analyzed the expression, mutational status, and prognostic value of these genes based on available public datasets. Our methodology involved data mining, differential expression analysis, functional enrichment analysis, and survival analysis to explore the association between cadherin gene expression and clinical outcomes in GC patients. Additionally, we investigated the relationship between cadherin expression and immune cell infiltration to gain insights into the tumor microenvironment's role in GC progression. Results Our bioinformatics analysis revealed significant differential expression of 16 cadherin genes in GC samples compared to normal tissues. Approximately up to 52% of the analyzed cancer samples exhibited genomic alterations in these cadherins, indicating their potential relevance in GC development. Functional enrichment analysis demonstrated that these differentially expressed cadherins were closely associated with critical cellular processes, including cell adhesion and immune-modulation. Remarkably, lower expression levels of most cadherin genes were linked to improved prognosis in GC patients, suggesting their potential importance as valuable prognostic biomarkers. Conclusions The findings deriving from our comprehensive study provide important insights into the dysregulation of cadherin genes in GC and their impact on gene expression, molecular pathways, and prognosis. The associations with clinical outcomes and immune cell infiltration highlight the potential role of cadherin genes as prognostic biomarkers and therapeutic targets in GC.
Collapse
Affiliation(s)
- Huan Wang
- Department of Medical Oncology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | | | - Puyue Gao
- Department of Digestive Medicine, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| |
Collapse
|
8
|
Siebert AE, Brake MA, Verbeek SC, Johnston AJ, Morgan AP, Cleuren AC, Jurek AM, Schneider CD, Germain DM, Battistuzzi FU, Zhu G, Miller DR, Johnsen JM, Pardo-Manuel de Villena F, Rondina MT, Westrick RJ. Identification of genomic loci regulating platelet plasminogen activator inhibitor-1 in mice. J Thromb Haemost 2023; 21:2917-2928. [PMID: 37364776 PMCID: PMC10826891 DOI: 10.1016/j.jtha.2023.06.018] [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: 08/29/2022] [Revised: 05/09/2023] [Accepted: 06/09/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Plasminogen activator inhibitor-1 (PAI-1, Serpine1) is an important circulating fibrinolysis inhibitor. PAI-1 exists in 2 pools, packaged within platelet α-granules and freely circulating in plasma. Elevated plasma PAI-1 levels are associated with cardiovascular disease. However, little is known about the regulation of platelet PAI-1 (pPAI-1). OBJECTIVES We investigated the genetic control of pPAI-1 levels in mice and humans. METHODS We measured pPAI-1 antigen levels via enzyme-linked immunosorbent assay in platelets isolated from 10 inbred mouse strains, including LEWES/EiJ (LEWES) and C57BL/6J (B6). LEWES and B6 were crossed to produce the F1 generation, B6LEWESF1. B6LEWESF1 mice were intercrossed to produce B6LEWESF2 mice. These mice were subjected to genome-wide genetic marker genotyping followed by quantitative trait locus analysis to identify pPAI-1 regulatory loci. RESULTS We identified differences in pPAI-1 between several laboratory strains, with LEWES having pPAI-1 levels more than 10-fold higher than those in B6. Quantitative trait locus analysis of B6LEWESF2 offspring identified a major pPAI-1 regulatory locus on chromosome 5 from 136.1 to 137.6 Mb (logarithm of the odds score, 16.2). Significant pPAI-1 modifier loci on chromosomes 6 and 13 were also identified. CONCLUSION Identification of pPAI-1 genomic regulatory elements provides insights into platelet/megakaryocyte-specific and cell type-specific gene expression. This information can be used to design more precise therapeutic targets for diseases where PAI-1 plays a role.
Collapse
Affiliation(s)
- Amy E Siebert
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | - Marisa A Brake
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | - Stephanie C Verbeek
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | | | - Andrew P Morgan
- Department of Medicine, Duke University School of Medicine, Duke University, Durham, North Carolina, USA
| | - Audrey C Cleuren
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Adrianna M Jurek
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | - Caitlin D Schneider
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | - Derrik M Germain
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | - Fabia U Battistuzzi
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA; Department of Bioengineering, Oakland University, Rochester, Michigan, USA; Centers for Data Science and Big Data Analytics and Biomedical Research, Oakland University, Rochester, Michigan, USA
| | - Guojing Zhu
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Darla R Miller
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jill M Johnsen
- Department of Medicine, Institute for Stem Cell & Regenerative Medicine, and Center for Cardiovascular Biology, University of Washington, Seattle, Washington, USA
| | - Fernando Pardo-Manuel de Villena
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Matthew T Rondina
- Molecular Medicine Program, Departments of Internal Medicine and Pathology, the University of Utah, Salt Lake City, Utah, USA; The George E. Wahlen Department of Medical Affairs Medical Center, Salt Lake City, Utah, USA
| | - Randal J Westrick
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA; Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA; Department of Bioengineering, Oakland University, Rochester, Michigan, USA; Centers for Data Science and Big Data Analytics and Biomedical Research, Oakland University, Rochester, Michigan, USA; Eye Research Center and Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, Michigan, USA.
| |
Collapse
|
9
|
Alissa M, Hjazi A, Abusalim GS, Aloraini GS, Alghamdi SA, Rizg WY, Hosny KM, Alblowi JA, Alkharobi H. Development and Optimization of a Novel Lozenge Containing a Metronidazole-Peppermint Oil-Tranexamic Acid Self-Nanoemulsified Delivery System to Be Used after Dental Extraction: In Vitro Evaluation and In Vivo Appraisal. Pharmaceutics 2023; 15:2342. [PMID: 37765310 PMCID: PMC10535350 DOI: 10.3390/pharmaceutics15092342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/07/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
In-depth studies on essential oil-based nanoemulsions (NEs) have centered on a variety of oral health issues. NEs improve the delivery of nonpolar active agents to sites and thereby boost the dissolution and distribution of the agents. Metronidazole-peppermint oil-tranexamic acid self-nanoemulsifying drug delivery systems (MZ-PO-TX-SNEDDS) were created and loaded into novel lozenges to act as antifungal, hemostatic, antimicrobial, and analgesic dosage forms after dental extractions. The design-of-experiments approach was used in creating them. To generate the NEs, different concentrations of MZ-PO (240, 180, and 120 mg), 2% TX (600, 450, and 300 mg), and Smix1:1 (600, 400, and 200 mg) were used. The ideal formulation had serum levels of 1530 U/mL of interleukin-6, a minimal inhibitory concentration against bacteria of 1.5 µg/mL, a droplet size of 96 nm, and a blood coagulation time of 16.5 min. Moreover, the produced NE offered better MZ release. The adopted design was used to produce the ideal formulation; it contained 240 mg of MZ-PO, 600 mg of 2% TX, and 600 mg of Smix1:1. It was incorporated into lozenges with acceptable characteristics and an improved capability for drug release. These lozenges had reasonable coagulation times, IL-6 serum levels, and MIC values. All of these characteristics are desirable for managing symptoms following tooth extractions. Therefore, these lozenges loaded with MZ-PO-TX-SNEDDs might be considered a beneficial paradigm for relieving complications encountered after tooth extractions.
Collapse
Affiliation(s)
- Mohammed Alissa
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (A.H.); (G.S.A.); (G.S.A.); (S.A.A.)
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (A.H.); (G.S.A.); (G.S.A.); (S.A.A.)
| | - Ghadah S. Abusalim
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (A.H.); (G.S.A.); (G.S.A.); (S.A.A.)
| | - Ghfren S. Aloraini
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (A.H.); (G.S.A.); (G.S.A.); (S.A.A.)
| | - Suad A. Alghamdi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (A.H.); (G.S.A.); (G.S.A.); (S.A.A.)
| | - Waleed Y. Rizg
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (W.Y.R.); (K.M.H.)
- Center of Innovation in Personalized Medicine (CIPM), 3D Bioprinting Unit, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Khaled M. Hosny
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (W.Y.R.); (K.M.H.)
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Jazia A. Alblowi
- Department of Periodontology, Faculty of Dentistry, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Hanaa Alkharobi
- Department of Oral Biology, Faculty of Dentistry, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| |
Collapse
|
10
|
Jeon HM, Kim JY, Cho HJ, Lee WJ, Nguyen D, Kim SS, Oh YT, Kim HJ, Jung CW, Pinero G, Joshi T, Hambardzumyan D, Sakaguchi T, Hubert CG, McIntyre TM, Fine HA, Gladson CL, Wang B, Purow BW, Park JB, Park MJ, Nam DH, Lee J. Tissue factor is a critical regulator of radiation therapy-induced glioblastoma remodeling. Cancer Cell 2023; 41:1480-1497.e9. [PMID: 37451272 PMCID: PMC10530238 DOI: 10.1016/j.ccell.2023.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 02/28/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023]
Abstract
Radiation therapy (RT) provides therapeutic benefits for patients with glioblastoma (GBM), but inevitably induces poorly understood global changes in GBM and its microenvironment (TME) that promote radio-resistance and recurrence. Through a cell surface marker screen, we identified that CD142 (tissue factor or F3) is robustly induced in the senescence-associated β-galactosidase (SA-βGal)-positive GBM cells after irradiation. F3 promotes clonal expansion of irradiated SA-βGal+ GBM cells and orchestrates oncogenic TME remodeling by activating both tumor-autonomous signaling and extrinsic coagulation pathways. Intratumoral F3 signaling induces a mesenchymal-like cell state transition and elevated chemokine secretion. Simultaneously, F3-mediated focal hypercoagulation states lead to activation of tumor-associated macrophages (TAMs) and extracellular matrix (ECM) remodeling. A newly developed F3-targeting agent potently inhibits the aforementioned oncogenic events and impedes tumor relapse in vivo. These findings support F3 as a critical regulator for therapeutic resistance and oncogenic senescence in GBM, opening potential therapeutic avenues.
Collapse
Affiliation(s)
- Hye-Min Jeon
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jeong-Yub Kim
- Divisions of Radiation Cancer Research, Research Center for Radio-Senescence, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Hee Jin Cho
- Department of Biomedical Convergence Science and Technology, Kyungpook National University, Daegu, Korea
| | - Won Jun Lee
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Dayna Nguyen
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sung Soo Kim
- Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Young Taek Oh
- Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Hee-Jin Kim
- Divisions of Radiation Cancer Research, Research Center for Radio-Senescence, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Chan-Woong Jung
- Divisions of Radiation Cancer Research, Research Center for Radio-Senescence, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Gonzalo Pinero
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tanvi Joshi
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Takuya Sakaguchi
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Christopher G Hubert
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Thomas M McIntyre
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Howard A Fine
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Candece L Gladson
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Bingcheng Wang
- Department of Medicine, MetroHealth Campus, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Benjamin W Purow
- Department of Neurology, UVA Cancer Center, University of Virginia Health System, Charlottesville, VA, USA
| | - Jong Bae Park
- Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Myung Jin Park
- Divisions of Radiation Cancer Research, Research Center for Radio-Senescence, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Do-Hyun Nam
- Institute for Refractory Cancer Research, Samsung Medical Center, Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Department of Neurosurgery Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeongwu Lee
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
11
|
Nguyen GN, Lindgren JR, Seleme MC, Kafle S, Zander CB, Zheng XL, Sabatino DE. Altered cleavage of human factor VIII at the B-domain and acidic region 3 interface enhances expression after gene therapy in hemophilia A mice. J Thromb Haemost 2023; 21:2101-2113. [PMID: 37080538 PMCID: PMC11157168 DOI: 10.1016/j.jtha.2023.04.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/17/2023] [Accepted: 04/04/2023] [Indexed: 04/22/2023]
Abstract
BACKGROUND Variants of human factor VIII (hFVIII) have been developed to further understand the structure and function of hFVIII and improve gene-based therapeutics. We have previously characterized several hFVIII variants of the furin cleavage site (1645-1648) with improved secretion. We have also identified a second cleavage site in the acidic region 3 (a3) (1657-1658) that becomes the primary hFVIII intracellular cleavage position in the absence of the furin site. We tested a hypothesis that modification of this site may confer additional functional advantages to hFVIII. OBJECTIVES The aim of this study was to conduct the biochemical and functional characterization of hFVIII variants of the furin cleavage site, the a3 cleavage site, or in combination, both in vitro and in vivo after AAV mediated gene therapy. METHODS Recombinant hFVIII variants of the furin cleavage site (hFVIII-Δ3), the a3 cleavage site (hFVIII-S1657P/D1658E [SP/DE]), or in combination (hFVIII-Δ3-SP/DE) were purified and characterized in vitro and in vivo. RESULTS Recombinant hFVIII-Δ3, hFVIII-SP/DE, and hFVIII-Δ3-SP/DE variants all had comparable specific activity to B-domain deleted (BDD) hFVIII. Hemophilia A mice tolerant to hFVIII did not develop immune responses to hFVIII after protein challenge with these variants or after adeno-associated virus (AAV) delivery. Following AAV delivery, hFVIII-Δ3-SP/DE resulted in expression levels that were 2- to 5-fold higher than those with hFVIII-BDD in hemophilia A mice. CONCLUSION The novel hFVIII-Δ3-SP/DE variant of the furin and a3 cleavage sites significantly improved secretion compared with hFVIII-BDD. This key feature of the Δ3-SP/DE variant provides a unique strategy that can be combined with other approaches to further improve factor VIII expression to achieve superior efficacy in AAV-based gene therapy for hemophilia A.
Collapse
Affiliation(s)
- Giang N Nguyen
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jonathan R Lindgren
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Maria C Seleme
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Samita Kafle
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Catherine B Zander
- Department of Pathology, University of Alabama at Birmingham School of Medicine, University of Alabama, Birmingham, Alabama, USA
| | - X Long Zheng
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, Kansas, USA; Institute of Reproductive Medicine and Developmental Science, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Denise E Sabatino
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Pediatrics, Division of Hematology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
12
|
Bhoopalan V, Gardiner EE, Kaur A. An Optimized Method of Collecting Murine Peripheral Blood and Dilution Correction for Accurate Blood Cell Enumeration. Curr Protoc 2023; 3:e765. [PMID: 37232369 DOI: 10.1002/cpz1.765] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Accurate measurement of whole blood counts from mice is an essential quantitative tool across the fields of vascular cell biology. In particular, the measurement of platelet counts can be challenging as the process relies upon good phlebotomy technique, the inclusion of a sufficient amount of the appropriate anticoagulant, and very often dilution of the sample to meet the sample volume requirements of an automated analyzer. To minimize sample dilution, blood collection tubes pre-coated with the anticoagulant can be used; however, these are expensive and prone to blood clotting issues. Here, we describe a simple dilution correction method that accurately calculates blood-to-anticoagulant dilutions to generate appropriate volumes for automated blood cell analysis while minimizing blood clotting. We also discuss some simple steps that can be incorporated into blood collection methods to avoid artefacts during blood collection. Blood count data analysis involving volume correction and clot exclusion can significantly reduce variable blood cell count values among healthy untreated littermates. It also detects subtle changes in blood cell counts, mainly of platelets and RBCs in experimental settings, which can be masked in the absence of careful and precise volume correction. Blood count analysis with volume correction precisely determines mouse whole blood cell counts for investigators. The decreased variability in cell count values reduces the number of experimental animals required for meaningful analysis. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol: An optimized method of collecting murine peripheral blood and dilution correction for accurate blood cell enumeration.
Collapse
Affiliation(s)
- Vijay Bhoopalan
- Division of Genome Sciences and Cancer, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Elizabeth E Gardiner
- Division of Genome Sciences and Cancer, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Amandeep Kaur
- Division of Genome Sciences and Cancer, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| |
Collapse
|
13
|
Laroche L, Ayhan N, Charrel R, Bañuls AL, Prudhomme J. Persistence of Toscana virus in sugar and blood meals of phlebotomine sand flies: epidemiological and experimental consequences. Sci Rep 2023; 13:5608. [PMID: 37019992 PMCID: PMC10076283 DOI: 10.1038/s41598-023-32431-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 03/27/2023] [Indexed: 04/07/2023] Open
Abstract
Many virological studies have tested the persistence of enveloped RNA viruses in various environmental and laboratory conditions and shown their short-term persistence. In this article, we analyzed Toscana virus (TOSV) infectivity, a pathogenic sandfly-borne phlebovirus, in two different conditions: in the sugar meal and blood meal of sand flies. Our results showed that TOSV RNA was detectable up to 15 days in sugar solution at 26 °C and up to 6 h in blood at 37 °C. Moreover, TOSV remains infective for 7 days in sugar solution and for minimum 6 h in rabbit blood. TOSV has shown persistent infectivity/viability under different conditions, which may have important epidemiological consequences. These results strengthen new hypotheses about the TOSV natural cycle, such as the possibility of horizontal transmission between sand flies through infected sugar meal.
Collapse
Affiliation(s)
- Lison Laroche
- UMR MIVEGEC, Université de Montpellier - IRD 224 - CNRS 5290, 911 Avenue Agropolis, 34394, Montpellier, France.
| | - Nazli Ayhan
- UVE, Aix Marseille Université - IRD 190 - Inserm 1207 - AP-HM Hôpitaux Universitaires de Marseille, Marseille, France
| | - Rémi Charrel
- UVE, Aix Marseille Université - IRD 190 - Inserm 1207 - AP-HM Hôpitaux Universitaires de Marseille, Marseille, France
| | - Anne-Laure Bañuls
- UMR MIVEGEC, Université de Montpellier - IRD 224 - CNRS 5290, 911 Avenue Agropolis, 34394, Montpellier, France
| | - Jorian Prudhomme
- UMR MIVEGEC, Université de Montpellier - IRD 224 - CNRS 5290, 911 Avenue Agropolis, 34394, Montpellier, France
- INTHERES, Université de Toulouse, INRAE, ENVT, Toulouse, France
| |
Collapse
|
14
|
Yousif Al-Fatlawi AC. Evaluation of the effects of mobile phone electromagnetic radiation on some physiological parameters and histological structure in some laboratory male mice organs. BIONATURA 2022. [DOI: 10.21931/rb/2022.07.04.56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Recently, the researcher has shown great interest in Electromagnetic radiation released from different devices such as TV, microwaves, medical apparatus, and satellites because of its effect on animals' growth and health. Exposure to "EMR" from mobiles phone can cause adverse effects on different cell functions. This study aimed to evaluate the effects of these radiations on histological and some blood parameters. The present study used 20 mice divided into two groups, the first one contains five animals as control, and the second experiment group contains 15 animals. EMR exposed from mobile for 12 h\day for one month. Histological examination of lungs, hearts and spleen showed a dramatic effect in these organs, such as necrosis, congestion, infiltrations, edema, splitting of muscle bundles and degenerations. This study shows that radiation from mobile phones contributes to histological changes in various visceral organs. Blood parameters showed a significant increase in platelets, bleeding and clotting time compared to the control group. The effect of EMR (Electromagnetic Radiation) on histology related to free radicals, increased lipid peroxidation in the cell membrane, and change in electrolyte concentration. An increase in platelets, bleeding and clotting time can also affect the rise in body temperature, ions and stimulations of stem cell divisions.
Keywords: electromagnetic radiations, mice, physiology, histology, mobile phone.
Collapse
|
15
|
Direct thrombin inhibitors as alternatives to heparin to preserve lung growth and function in a murine model of compensatory lung growth. Sci Rep 2022; 12:21117. [PMID: 36477689 PMCID: PMC9729628 DOI: 10.1038/s41598-022-25773-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Infants with congenital diaphragmatic hernia (CDH) may require cardiopulmonary bypass and systemic anticoagulation. Expeditious lung growth while on bypass is essential for survival. Previously, we demonstrated that heparin impairs lung growth and function in a murine model of compensatory lung growth (CLG). We investigated the effects of the direct thrombin inhibitors (DTIs) bivalirudin and argatroban. In vitro assays of lung endothelial cell proliferation and apoptosis were performed. C57BL/6 J mice underwent left pneumonectomy and subcutaneous implantation of osmotic pumps. Pumps were pre-loaded with normal saline (control), bivalirudin, argatroban, or heparin and outcomes were assessed on postoperative day 8. Heparin administration inhibited endothelial cell proliferation in vitro and significantly decreased lung volume in vivo, while bivalirudin and argatroban preserved lung growth. These findings correlated with changes in alveolarization on morphometric analysis. Treadmill exercise tolerance testing demonstrated impaired exercise performance in heparinized mice; bivalirudin/argatroban did not affect exercise tolerance. On lung protein analysis, heparin decreased angiogenic signaling which was not impacted by bivalirudin or argatroban. Together, this data supports the use of DTIs as alternatives to heparin for systemic anticoagulation in CDH patients on bypass. Based on this work, clinical studies on the impact of heparin and DTIs on CDH outcomes are warranted.
Collapse
|
16
|
Meng Q, Li B, Huang N, Wei S, Ren Q, Wu S, Li X, Chen R. Folic acid targets splenic extramedullary hemopoiesis to attenuate carbon black-induced coagulation-thrombosis potential. JOURNAL OF HAZARDOUS MATERIALS 2022; 424:127354. [PMID: 34634699 DOI: 10.1016/j.jhazmat.2021.127354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/14/2021] [Accepted: 09/23/2021] [Indexed: 06/13/2023]
Abstract
Due to its wide applications in tire and rubber products, carbon black (CB) implicates concerns on its safety during production, collection, and handling. Here we report that exposure CB, increases coagulation-thrombosis potential in a splenic extramedullary hemopoiesis (EMH)-dependent manner. Adult C57BL/6 mice are kept in whole-body inhalation chambers, and exposed to filtered room air (FRA) or CB for 28 consecutive days. CB exposure resulted in splenic EMH characterized with platelet precursor cells, megakaryocytes (MKs), hyperplasia and enhanced in vivo blood coagulation ability. Metabolomics analysis suggests significant enhance in PGE2 production but reduction in folic acid (FA) levels in murine serum following CB exposure. Mechanistically, activation of COX-dependent PGE2 production promotes IL-6 expression in splenic macrophages, which subsequently results in splenic EMH and increased platelet counts in circulation. Administration of FA protects the mice against CB-induced splenic EMH through inhibiting prostaglandin-endoperoxide synthase 2 (Ptgs2 or Cox2) and prostaglandin E synthase (Ptges) expression in splenic macrophages, eventually recover the coagulation capacity to normal level. The results strongly suggest the involvement of splenic EMH in response to CB exposure and subsequently increased coagulation-thrombosis potential. Supplementation with FA may be a candidate to prevent thrombosis potential attributable to CB exposure.
Collapse
Affiliation(s)
- Qingtao Meng
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China; School of Public Health, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, PR China
| | - Bin Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China
| | - Nannan Huang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China
| | - Shengnan Wei
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China
| | - Quanzhong Ren
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China; School of Public Health, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, PR China
| | - Shenshen Wu
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China; School of Public Health, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, PR China
| | - Xiaobo Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China.
| | - Rui Chen
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China; School of Public Health, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, PR China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
17
|
Mohammed BM, Cheng Q, Ivanov IS, Gailani D. Murine Models in the Evaluation of Heparan Sulfate-Based Anticoagulants. Methods Mol Biol 2022; 2303:789-805. [PMID: 34626423 PMCID: PMC8552346 DOI: 10.1007/978-1-0716-1398-6_59] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Evaluating prospective anticoagulant therapies in animal thrombosis and bleeding models are standard pre-clinical approaches. Mice are frequently used for initial evaluations because a variety of models have been developed in this well-characterized species, and mice are relatively inexpensive to maintain. Because mice seem to be resistant to forming "spontaneous" thrombosis, vessel injury is used to induce intravascular clot formation. For the purpose of testing heparin-based drugs, we adapted a well-established model in which thrombus formation in the carotid artery is induced by exposing the vessel to ferric chloride. For studying anticoagulant effects on venous thrombosis, we use a model in which the inferior vena cava is ligated and the size of the resulting clots are measured. The most common adverse effect of anticoagulation therapy is bleeding. We describe a simple tail bleeding time that has been used for many years to study the effects of anticoagulants on hemostasis. We also describe a more reproducible, but more technically challenging, saphenous vein bleeding model that is also used for this purpose.
Collapse
Affiliation(s)
- Bassem M Mohammed
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Doisy Research Center, St. Louis, MO, USA
| | - Qiufang Cheng
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Ivan S Ivanov
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
18
|
Miguel ZD, Khoury N, Betley MJ, Lehallier B, Willoughby D, Olsson N, Yang AC, Hahn O, Lu N, Vest RT, Bonanno LN, Yerra L, Zhang L, Saw NL, Fairchild JK, Lee D, Zhang H, McAlpine PL, Contrepois K, Shamloo M, Elias JE, Rando TA, Wyss-Coray T. Exercise plasma boosts memory and dampens brain inflammation via clusterin. Nature 2021; 600:494-499. [PMID: 34880498 PMCID: PMC9721468 DOI: 10.1038/s41586-021-04183-x] [Citation(s) in RCA: 201] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/26/2021] [Indexed: 12/20/2022]
Abstract
Physical exercise is generally beneficial to all aspects of human and animal health, slowing cognitive ageing and neurodegeneration1. The cognitive benefits of physical exercise are tied to an increased plasticity and reduced inflammation within the hippocampus2-4, yet little is known about the factors and mechanisms that mediate these effects. Here we show that 'runner plasma', collected from voluntarily running mice and infused into sedentary mice, reduces baseline neuroinflammatory gene expression and experimentally induced brain inflammation. Plasma proteomic analysis revealed a concerted increase in complement cascade inhibitors including clusterin (CLU). Intravenously injected CLU binds to brain endothelial cells and reduces neuroinflammatory gene expression in a mouse model of acute brain inflammation and a mouse model of Alzheimer's disease. Patients with cognitive impairment who participated in structured exercise for 6 months had higher plasma levels of CLU. These findings demonstrate the existence of anti-inflammatory exercise factors that are transferrable, target the cerebrovasculature and benefit the brain, and are present in humans who engage in exercise.
Collapse
Affiliation(s)
- Zurine De Miguel
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.,Present address: Psychology Department, California State University, Monterey Bay, CA, USA
| | - Nathalie Khoury
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.,These authors contributed equally: Nathalie Khoury, Michael J. Betley
| | - Michael J. Betley
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Neurosciences Graduate Training Program, Stanford University School of Medicine, Stanford, CA, USA.,These authors contributed equally: Nathalie Khoury, Michael J. Betley
| | - Benoit Lehallier
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.,Present address: Alkahest Inc, San Carlos, CA, USA
| | - Drew Willoughby
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Niclas Olsson
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA.,Present address: Calico Life Sciences, South San Francisco, CA, USA
| | - Andrew C. Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Oliver Hahn
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Nannan Lu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Ryan T. Vest
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Liana N. Bonanno
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Lakshmi Yerra
- The Veterans Affairs Palo Alto HealthCare System, Palo Alto, CA, USA
| | | | - Nay Lui Saw
- Behavioral and Functional Neuroscience Laboratory, Stanford University School of Medicine, Stanford, CA, USA
| | - J. Kaci Fairchild
- The Veterans Affairs Palo Alto HealthCare System, Palo Alto, CA, USA
| | - Davis Lee
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Hui Zhang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Patrick L. McAlpine
- Otolaryngology Head and Neck Surgery Research Division, Stanford University, Stanford, CA, USA
| | | | - Mehrdad Shamloo
- Behavioral and Functional Neuroscience Laboratory, Stanford University School of Medicine, Stanford, CA, USA
| | - Joshua E. Elias
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA.,Chan Zuckerberg Biohub, Stanford, CA, USA
| | - Thomas A. Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.,The Veterans Affairs Palo Alto HealthCare System, Palo Alto, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA. .,Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA. .,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
19
|
Schroeder JA, Kuether EA, Fang J, Jing W, Weiler H, Wilcox DA, Montgomery RR, Shi Q. Thromboelastometry assessment of hemostatic properties in various murine models with coagulopathy and the effect of factor VIII therapeutics. J Thromb Haemost 2021; 19:2417-2427. [PMID: 34245090 PMCID: PMC8865566 DOI: 10.1111/jth.15456] [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: 05/09/2021] [Accepted: 07/07/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Rotational thromboelastometry (ROTEM) has been commonly used to assess the viscoelastic properties of the blood clotting process in the clinic for patients with a hemostatic or prothrombotic disorder. OBJECTIVE To evaluate the capability of ROTEM in assessing hemostatic properties in whole blood from various mouse models with genetic bleeding or clotting disease and the effect of factor VIII (FVIII) therapeutics in FVIIInull mice. METHODS Mice with a genetic deficiency in either a coagulation factor or a platelet glycoprotein were used in this study. The properties of platelet- or plasma-FVIII were also assessed. Citrated blood from mice was recalcified and used for ROTEM analysis. RESULTS We found that blood collected from the vena cava could generate reliable results from ROTEM analysis, but not blood collected from the tail vein, retro-orbital plexus, or submandibular vein. Age and sex did not significantly affect the hemostatic properties determined by ROTEM analysis. Clotting time (CT) and clot formation time (CFT) were significantly prolonged in FVIIInull (5- and 9-fold, respectively) and FIXnull (4- and 5.7-fold, respectively) mice compared to wild-type (WT)-C57BL/6J mice. Platelet glycoprotein (GP)IIIanull mice had significantly prolonged CFT (8.4-fold) compared to WT-C57BL/6J mice. CT and CFT in factor V (FV) Leiden mice were significantly shortened with an increased α-angle compared to WT-C57BL/6J mice. Using ROTEM analysis, we showed that FVIII expressed in platelets or infused into whole blood restored hemostasis of FVIIInull mice in a dose-dependent manner. CONCLUSION ROTEM is a reliable and sensitive assay for assessing therapeutics on hemostatic properties in mouse models with a bleeding or clotting disorder.
Collapse
Affiliation(s)
- Jocelyn A. Schroeder
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, USA
- MACC Fund Research Center, Milwaukee, Wisconsin, USA
| | - Erin A. Kuether
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, USA
- MACC Fund Research Center, Milwaukee, Wisconsin, USA
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- MACC Fund Research Center, Milwaukee, Wisconsin, USA
| | - Weiqing Jing
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA
| | - Hartmut Weiler
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA
| | - David A. Wilcox
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, USA
- MACC Fund Research Center, Milwaukee, Wisconsin, USA
| | - Robert R. Montgomery
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, USA
- MACC Fund Research Center, Milwaukee, Wisconsin, USA
| | - Qizhen Shi
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, USA
- MACC Fund Research Center, Milwaukee, Wisconsin, USA
| |
Collapse
|
20
|
Cui W, Wu X, Feng D, Luo J, Shi Y, Guo W, Liu H, Wang Q, Wang L, Ge S, Qu Y. Acrolein Induces Systemic Coagulopathy via Autophagy-dependent Secretion of von Willebrand Factor in Mice after Traumatic Brain Injury. Neurosci Bull 2021; 37:1160-1175. [PMID: 33939120 PMCID: PMC8353051 DOI: 10.1007/s12264-021-00681-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/05/2020] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI)-induced coagulopathy has increasingly been recognized as a significant risk factor for poor outcomes, but the pathogenesis remains poorly understood. In this study, we aimed to investigate the causal role of acrolein, a typical lipid peroxidation product, in TBI-induced coagulopathy, and further explore the underlying molecular mechanisms. We found that the level of plasma acrolein in TBI patients suffering from coagulopathy was higher than that in those without coagulopathy. Using a controlled cortical impact mouse model, we demonstrated that the acrolein scavenger phenelzine prevented TBI-induced coagulopathy and recombinant ADAMTS-13 prevented acrolein-induced coagulopathy by cleaving von Willebrand factor (VWF). Our results showed that acrolein may contribute to an early hypercoagulable state after TBI by regulating VWF secretion. mRNA sequencing (mRNA-seq) and transcriptome analysis indicated that acrolein over-activated autophagy, and subsequent experiments revealed that acrolein activated autophagy partly by regulating the Akt/mTOR pathway. In addition, we demonstrated that acrolein was produced in the perilesional cortex, affected endothelial cell integrity, and disrupted the blood-brain barrier. In conclusion, in this study we uncovered a novel pro-coagulant effect of acrolein that may contribute to TBI-induced coagulopathy and vascular leakage, providing an alternative therapeutic target.
Collapse
Affiliation(s)
- Wenxing Cui
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Xun Wu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Dayun Feng
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Jianing Luo
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Yingwu Shi
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Wei Guo
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Haixiao Liu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Qiang Wang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Liang Wang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Shunnan Ge
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.
| |
Collapse
|
21
|
Schulman S, El-Darzi E, Florido MH, Friesen M, Merrill-Skoloff G, Brake MA, Schuster CR, Lin L, Westrick RJ, Cowan CA, Flaumenhaft R, Ouwehand WH, Peerlinck K, Freson K, Turro E, Furie B. A coagulation defect arising from heterozygous premature termination of tissue factor. J Clin Invest 2020; 130:5302-5312. [PMID: 32663190 PMCID: PMC7524505 DOI: 10.1172/jci133780] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 07/01/2020] [Indexed: 11/17/2022] Open
Abstract
Tissue factor (TF) is the primary initiator of blood coagulation in vivo and the only blood coagulation factor for which a human genetic defect has not been described. As there are no routine clinical assays that capture the contribution of endogenous TF to coagulation initiation, the extent to which reduced TF activity contributes to unexplained bleeding is unknown. Using whole genome sequencing, we identified a heterozygous frameshift variant (p.Ser117HisfsTer10) in F3, the gene encoding TF, causing premature termination of TF (TFshort) in a woman with unexplained bleeding. Routine hematological laboratory evaluation of the proposita was normal. CRISPR-edited human induced pluripotent stem cells recapitulating the variant were differentiated into vascular smooth muscle and endothelial cells that demonstrated haploinsufficiency of TF. The variant F3 transcript is eliminated by nonsense-mediated decay. Neither overexpression nor addition of exogenous recombinant TFshort inhibited factor Xa or thrombin generation, excluding a dominant-negative mechanism. F3+/- mice provide an animal model of TF haploinsufficiency and exhibited prolonged bleeding times, impaired thrombus formation, and reduced survival following major injury. Heterozygous TF deficiency is present in at least 1 in 25,000 individuals and could limit coagulation initiation in undiagnosed individuals with abnormal bleeding but a normal routine laboratory evaluation.
Collapse
Affiliation(s)
- Sol Schulman
- Division of Hemostasis and Thrombosis
- Division of Hematology and Oncology, and
| | | | - Mary H.C. Florido
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | - Max Friesen
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | | | - Marisa A. Brake
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | | | - Lin Lin
- Division of Hemostasis and Thrombosis
| | - Randal J. Westrick
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | - Chad A. Cowan
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | | | - NIHR BioResource
- NIHR BioResource, Cambridge University Hospitals (detailed in the Supplemental Acknowledgments)
| | - Willem H. Ouwehand
- Department of Haematology, University of Cambridge, and
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Kathelijne Peerlinck
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Kathleen Freson
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Ernest Turro
- NIHR BioResource, Cambridge University Hospitals (detailed in the Supplemental Acknowledgments)
- Department of Haematology, University of Cambridge, and
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Medical Research Council Biostatistics Unit, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | | |
Collapse
|
22
|
Mohammed BM, Monroe DM, Gailani D. Mouse models of hemostasis. Platelets 2020; 31:417-422. [PMID: 31992118 PMCID: PMC7244364 DOI: 10.1080/09537104.2020.1719056] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 02/02/2023]
Abstract
Hemostasis is the normal process that produces a blood clot at a site of vascular injury. Mice are widely used to study hemostasis and abnormalities of blood coagulation because their hemostatic system is similar in most respects to that of humans, and their genomes can be easily manipulated to create models of inherited human coagulation disorders. Two of the most widely used techniques for assessing hemostasis in mice are the tail bleeding time (TBT) and saphenous vein bleeding (SVB) models. Here we discuss the use of these methods in the evaluation of hemostasis, and the advantages and limits of using mice as surrogates for studying hemostasis in humans.
Collapse
Affiliation(s)
- Bassem M. Mohammed
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN
- Department of Pathology and Immunology, Washington University, St. Louis, MO
| | - Dougald M. Monroe
- UNC Blood Research Center and Hematology/Oncology, University of North Carolina, Chapel Hill, NC
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN
| |
Collapse
|