1
|
Sim DS, Shukla M, Mallari CR, Fernández JA, Xu X, Schneider D, Bauzon M, Hermiston TW, Mosnier LO. Divergent modulation of activated protein C pleiotropic functions by antibodies that differ by a single amino acid. Blood Adv 2025; 9:180-191. [PMID: 39471469 PMCID: PMC11788130 DOI: 10.1182/bloodadvances.2024013584] [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] [Received: 06/03/2024] [Revised: 10/07/2024] [Accepted: 10/22/2024] [Indexed: 11/01/2024] Open
Abstract
ABSTRACT Activated protein C (APC) is a pleiotropic plasma protease with diverse functions derived from its anticoagulant, anti-inflammatory, and cytoprotective activities. The selective uncoupling and/or modulation of these APC activities by antibodies may have therapeutic benefit in diseases such as traumatic bleeding, hemophilia, sepsis, and ischemia. TPP-26870 is an antibody that targets a nonactive site of APC for the selective modulation of APC activities. To optimize the potency of TPP-26870, variants with single amino acid mutation in the complementarity-determining regions (CDRs) were screened, and 21 variants with improved affinity constant were identified. Interestingly, the affinity maturation of TPP-26870 did not merely generate a panel of variants with higher potency in functional assays. Functional data demonstrated that the pleiotropic functions of APC were very sensitive to epitope-CDR interactions. Single amino acid mutations within the CDRs of TPP-26870 were sufficient to elicit divergent antagonistic and agonistic effects on the various APC functional activities. These include prolonged in vitro APC plasma half-life, increased inhibition of anticoagulant activity, and agonistic enhancement of histone H3 cleavage, while having less impact on protease-activated receptor 1 cleavage, compared with TPP-26870. This study illustrates that APC is highly sensitive to non-active site targeting that can lead to unpredictable changes in its activity profile of this pleiotropic enzyme. Furthermore, this study demonstrates the ability to modify APC functions to advance the potential development of APC-targeted antibodies as therapeutics for the treatment of diseases including trauma bleeding, hemophilia, ischemia, and sepsis.
Collapse
Affiliation(s)
- Derek S. Sim
- Coagulant Therapeutics Corporation, Berkeley, CA
| | - Meenal Shukla
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | | | - José A. Fernández
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Xiao Xu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | | | - Maxine Bauzon
- Consultants for Coagulant Therapeutics, Berkeley, CA
| | | | - Laurent O. Mosnier
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|
2
|
Liu G, Liao W, Lv X, Huang L, He M, Li L. A potential coagulation-related diagnostic model associated with immune infiltration for acute myocardial infarction. Genes Immun 2024; 25:471-482. [PMID: 39379556 DOI: 10.1038/s41435-024-00298-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/10/2024]
Abstract
The production of pro-coagulation factors can affect the development and prognosis of acute myocardial infarction (AMI). The clinical value of coagulation-related genes (CRGs) was investigated to discover new targets for diagnosing and treating AMI. We screened 335 differentially expressed genes (DEGs) between AMI and healthy individuals based on the GSE66360 dataset. We took the intersection of the obtained DEGs with 139 CRGs. Finally, 10 differentially expressed CEGs were screened out. The random forest algorithm was constructed to identify 6 signature CRGs (THBS1, SERPINA1, THBD, MMP9, MAFF, and PLAU). Subsequently, the established predictive model was found to have good diagnostic accuracy (AUC = 0.9694 in the training cohort [GSE66360 dataset] and 0.9076 in the external validation cohort [GSE48060 dataset]). Consensus clustering identified the CRG clusters, and the accuracy of the grouping was verified. We found that AMI patients can be divided into two distinct subgroups based on the differentially expressed CRGs. Immune cell infiltration level was consistent with the expression levels of CRGs based on single sample gene set enrichment analysis. These findings reveal the potential role of CRGs in AMI. Characterizing the coagulation features of AMI patients can help in the risk stratification of patients and provide personalized treatment strategies.
Collapse
Affiliation(s)
- Guoqing Liu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Wang Liao
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xiangwen Lv
- Department of Cardiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Lifeng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Min He
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Lang Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
3
|
Rajput P, Brookshier A, Kothari S, Eckstein L, Chang H, Liska S, Lamb J, Sances S, Lyden P. Differential Vulnerability and Response to Injury among Brain Cell Types Comprising the Neurovascular Unit. J Neurosci 2024; 44:e1093222024. [PMID: 38548341 PMCID: PMC11140689 DOI: 10.1523/jneurosci.1093-22.2024] [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: 05/21/2022] [Revised: 02/29/2024] [Accepted: 03/11/2024] [Indexed: 05/31/2024] Open
Abstract
The neurovascular unit (NVU) includes multiple different cell types, including neurons, astrocytes, endothelial cells, and pericytes, which respond to insults on very different time or dose scales. We defined differential vulnerability among these cell types, using response to two different insults: oxygen-glucose deprivation (OGD) and thrombin-mediated cytotoxicity. We found that neurons are most vulnerable, followed by endothelial cells and astrocytes. After temporary focal cerebral ischemia in male rats, we found significantly more injured neurons, compared with astrocytes in the ischemic area, consistent with differential vulnerability in vivo. We sought to illustrate different and shared mechanisms across all cell types during response to insult. We found that gene expression profiles in response to OGD differed among the cell types, with a paucity of gene responses shared by all types. All cell types activated genes relating to autophagy, apoptosis, and necroptosis, but the specific genes differed. Astrocytes and endothelial cells also activated pathways connected to DNA repair and antiapoptosis. Taken together, the data support the concept of differential vulnerability in the NVU and suggest that different elements of the unit will evolve from salvageable to irretrievable on different time scales while residing in the same brain region and receiving the same (ischemic) blood flow. Future work will focus on the mechanisms of these differences. These data suggest future stroke therapy development should target different elements of the NVU differently.
Collapse
Affiliation(s)
- Padmesh Rajput
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, California 90089-2821
| | - Allison Brookshier
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, California 90089-2821
| | - Shweta Kothari
- Chinook Therapeutics, Inc., Vancouver, British Columbia V5T 4T5, Canada
- Cedars-Sinai Medical Center, Los Angeles, California 90048
| | - Lillie Eckstein
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, California 90089-2821
| | - Heather Chang
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, California 90089-2821
| | - Sophie Liska
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, California 90089-2821
| | - Jessica Lamb
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, California 90089-2821
| | - Samuel Sances
- Cedars-Sinai Medical Center, Los Angeles, California 90048
| | - Patrick Lyden
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, California 90089-2821
| |
Collapse
|
4
|
Thielen O, Mitra S, Debot M, Schaid T, Hallas W, Gallagher LT, Erickson C, Cralley A, Stafford P, Silliman C, D'Alessandro A, Hansen K, Sauaia A, Moore E, Mosnier L, Griffin J, Cohen M. Mitigation of trauma-induced endotheliopathy by activated protein C: A potential therapeutic for postinjury thromboinflammation. J Trauma Acute Care Surg 2024; 96:116-122. [PMID: 37733304 PMCID: PMC10841096 DOI: 10.1097/ta.0000000000004142] [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] [Indexed: 09/22/2023]
Abstract
BACKGROUND Activated Protein C (aPC) plays dual roles after injury, driving both trauma-induced coagulopathy (TIC) by cleaving, and thus inactivating, factors Va and VIIIa and depressing fibrinolysis while also mediating an inflammomodulatory milieu via protease activated receptor-1 (PAR-1) cytoprotective signaling. Because of this dual role, it represents and ideal target for study and therapeutics after trauma. A known aPC variant, 3K3A-aPC, has been engineered to preserve cytoprotective activity while retaining minimal anticoagulant activity rendering it potentially ideal as a cytoprotective therapeutic after trauma. We hypothesized that 3K3A-aPC would mitigate the endotheliopathy of trauma by protecting against endothelial permeability. METHODS We used electric cell-substrate impedance sensing to measure permeability changes in real time in primary endothelial cells. These were cultured, grown to confluence, and treated with a 2 μg/mL solution of 3K3A-aPC at 180 minutes, 120 minutes, 60 minutes, 30 minutes prior to stimulation with ex vivo plasma taken from severely injured trauma patients (Injury Severity Score > 15 and BD < -6) (trauma plasma [TP]). Cells treated with thrombin and untreated cells were included in this study as control groups. Permeability changes were recorded in real time via electric cell-substrate impedance sensing for 30 minutes after treatment with TP. We quantified permeability changes in the control and treatment groups as area under the curve (AUC). Rac1/RhoA activity was also compared between these groups. Statistical significance was determined by one-way ANOVA followed by a post hoc analysis using Tukey's multiple comparison's test. RESULTS Treatment with aPC mitigated endothelial permeability induced by ex vivo trauma plasma at all pre-treatment time points. The AUC of the 30-minute 3K3A-aPC pretreatment group was higher than TP alone (mean diff. 22.12 95% CI [13.75, 30.49], p < 0.0001) (Figure). Moreover, the AUC of the 60-minute, 120-minute, and 180-minute pretreatment groups was also higher than TP alone (mean diff., 16.30; 95% confidence interval [CI], 7.93-24.67; 19.43; 95% CI, 11.06-27.80, and 18.65; 95% CI, 10.28-27.02;, all p < 0.0001, respectively). Rac1/RhoA activity was higher in the aPC pretreatment group when compared with all other groups ( p < 0.01). CONCLUSION Pretreatment with 3K3A-aPC, which retains its cytoprotective function but has only ~5% of its anticoagulant function, abrogates the effects of trauma-induced endotheliopathy. This represents a potential therapeutic treatment for dysregulated thromboinflammation for injured patients by minimizing aPC's role in trauma-induced coagulopathy while concurrently amplifying its essential cytoprotective function. LEVEL OF EVIDENCE Prognostic and Epidemiological; Level III.
Collapse
Affiliation(s)
- Otto Thielen
- From the Department of Gastrointestinal, Trauma, and Endocrine Surgery (O.T., S.M., M.D., T.S., W.H., L.T.G., C.E., A.C., P.S., C.S., A.D'A., K.H., A.S., E.M., M.C.), University of Colorado, Denver, Colorado; Department of Surgery (A.S., E.M.), Denver Health Medical Center, Denver, Colorado; and Department of Molecular Medicine (L.M., J.G.), Scripps Research, La Jolla, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Huckriede JB, Beurskens DMH, Wildhagen KCCA, Reutelingsperger CPM, Wichapong K, Nicolaes GAF. Design and characterization of novel activated protein C variants for the proteolysis of cytotoxic extracellular histone H3. J Thromb Haemost 2023; 21:3557-3567. [PMID: 37657561 DOI: 10.1016/j.jtha.2023.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/24/2023] [Accepted: 08/23/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND Extracellular histone H3 is implicated in several pathologies including inflammation, cell death, and organ failure. Neutralization of histone H3 is a strategy that was shown beneficial in various diseases, such as rheumatoid arthritis, myocardial infarction, and sepsis. It was shown that activated protein C (APC) can cleave histone H3, which reduces histone cytotoxicity. However, due to the anticoagulant properties of APC, the use of APC is not optimal for the treatment of histone-mediated cytotoxicity, in view of its associated bleeding side effects. OBJECTIVES This study aimed to investigate the detailed molecular interactions between human APC and human histone H3, and subsequently use molecular docking and molecular dynamics simulation methods to identify key interacting residues that mediate the interaction between APC and histone H3 and to generate novel optimized APC variants. METHODS After molecular simulations, the designed APC variants 3D2D-APC (Lys37-39Asp and Lys62-63Asp) and 3D2D2A-APC (Lys37-39Asp, Lys62-63Asp, and Arg74-75Ala) were recombinantly expressed and their abilities to function as anticoagulant, to bind histones, and to cleave histones were tested and correlated with their cytoprotective properties. RESULTS Compared with wild type-APC, both the 3D2D-APC and 3D2D2A-APC variants showed a significantly decreased anticoagulant activity, increased binding to histone H3, and similar ability to proteolyze histone H3. CONCLUSIONS Our data show that it is possible to rationally design APC variants that may be further developed into therapeutic biologicals to treat histone-mediated disease, by proteolytic reduction of histone-associated cytotoxic properties that do not induce an increased bleeding risk.
Collapse
Affiliation(s)
- Joram B Huckriede
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Daniëlle M H Beurskens
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Karin C C A Wildhagen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Chris P M Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Kanin Wichapong
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Gerry A F Nicolaes
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
6
|
Sim DS, Shukla M, Mallari CR, Fernández JA, Xu X, Schneider D, Bauzon M, Hermiston TW, Mosnier LO. Selective modulation of activated protein C activities by a nonactive site-targeting nanobody library. Blood Adv 2023; 7:3036-3048. [PMID: 36735416 PMCID: PMC10331410 DOI: 10.1182/bloodadvances.2022008740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 01/13/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023] Open
Abstract
Activated protein C (APC) is a pleiotropic coagulation protease with anticoagulant, anti-inflammatory, and cytoprotective activities. Selective modulation of these APC activities contributes to our understanding of the regulation of these physiological mechanisms and permits the development of therapeutics for the pathologies associated with these pathways. An antibody library targeting the nonactive site of APC was generated using llama antibodies (nanobodies). Twenty-one nanobodies were identified that selectively recognize APC compared with the protein C zymogen. Overall, 3 clusters of nanobodies were identified based on the competition for APC in biolayer interferometry studies. APC functional assays for anticoagulant activity, histone H3 cleavage, and protease-activated receptor 1 (PAR1) cleavage were used to understand their diversity. These functional assays revealed 13 novel nanobody-induced APC activity profiles via the selective modulation of APC pleiotropic activities, with the potential to regulate specific mechanisms for therapeutic purposes. Within these, 3 nanobodies (LP2, LP8, and LP17) inhibited all 3 APC functions. Four nanobodies (LP1, LP5, LP16, and LP20) inhibited only 2 of the 3 functions. Monofunction inhibition specific to APC anticoagulation activity was observed only by 2 nanobodies (LP9 and LP11). LP11 was also found to shift the ratio of APC cleavage of PAR1 at R46 relative to R41, which results in APC-mediated biased PAR1 signaling and APC cytoprotective effects. Thus, LP11 has an activity profile that could potentially promote hemostasis and cytoprotection in bleedings associated with hemophilia or coagulopathy by selectively modulating APC anticoagulation and PAR1 cleavage profile.
Collapse
Affiliation(s)
- Derek S. Sim
- Coagulant Therapeutics Corporation, Berkeley, CA
| | - Meenal Shukla
- Department of Molecular Medicine, Scripps Research, La Jolla, CA
| | | | | | - Xiao Xu
- Department of Molecular Medicine, Scripps Research, La Jolla, CA
| | | | - Maxine Bauzon
- Consultants for Coagulant Therapeutics, Berkeley, CA
| | | | | |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW To provide an overview of the state-of-the-art in protein C (PC) pathway research. RECENT FINDINGS The PC pathway is crucial for maintaining hemostasis to prevent venous thromboembolism. This is evident from genetic mutations that result in impaired PC pathway activity and contribute to increased venous thromboembolism risk in affected individuals. In addition to its anticoagulant role, activated PC (APC) also mediates a complex, pleiotropic role in the maintenance of vascular cell health, which it achieves via anti-inflammatory and antiapoptotic cell signaling on endothelial cells. Emerging data have demonstrated that cell signaling by APC, mediated by multiple receptor interactions on different cell types, also confers cytoprotective and anti-inflammatory benefits. Defects in both arms of the PC pathway are associated with increased susceptibility to thrombo-inflammatory disease in various preclinical thrombotic, proinflammatory and neurological disease models. Moreover, recent studies have identified attenuation of anticoagulant PC pathway activity as an exciting therapeutic opportunity to promote hemostasis in patients with inherited or acquired bleeding disorders. SUMMARY In this review, we provide an overview of some recent developments in our understanding of the PC pathways.
Collapse
Affiliation(s)
- Gemma Leon
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Crumlin, Dublin 12, Ireland
| | - Aisling M Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Crumlin, Dublin 12, Ireland
| |
Collapse
|
8
|
Wang Y, Kisler K, Nikolakopoulou AM, Fernandez JA, Griffin JH, Zlokovic BV. 3K3A-Activated Protein C Protects the Blood-Brain Barrier and Neurons From Accelerated Ischemic Injury Caused by Pericyte Deficiency in Mice. Front Neurosci 2022; 16:841916. [PMID: 35431776 PMCID: PMC9005806 DOI: 10.3389/fnins.2022.841916] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/16/2022] [Indexed: 11/16/2022] Open
Abstract
Pericytes, mural cells of brain capillaries, maintain the blood-brain barrier (BBB), regulate cerebral blood flow (CBF), and protect neurons against ischemic damage. To further investigate the role of pericytes in ischemia, we induced stroke by 45-min transient middle cerebral artery occlusion (tMCAo) in 6-month-old pericyte-deficient Pdgfrb + /- mice and control Pdgfrb+/+ littermates. Compared to controls, Pdgfrb + /- mice showed a 26% greater loss of CBF during early reperfusion, and 40-50% increase in the infarct and edema volumes and motor neurological score 24 h after tMCAo. These changes were accompanied by 50% increase in both immunoglobulin G and fibrinogen pericapillary deposits in the ischemic cortex 8 h after tMCAo indicating an accelerated BBB breakdown, and 35 and 55% greater losses of pericyte coverage and number of degenerating neurons 24 h after tMCAo, respectively. Treatment of Pdgfrb + /- mice with 3K3A-activated protein C (APC), a cell-signaling analog of plasma protease APC, administered intravenously 10 min and 4 h after tMCAo normalized CBF during the early reperfusion phase and reduced infarct and edema volume and motor neurological score by 55-60%, with similar reductions in BBB breakdown and number of degenerating neurons. Our data suggest that pericyte deficiency results in greater brain injury, BBB breakdown, and neuronal degeneration in stroked mice and that 3K3A-APC protects the brain from accelerated injury caused by pericyte deficiency. These findings may have implications for treatment of ischemic brain injury in neurological conditions associated with pericyte loss such as those seen during normal aging and in neurodegenerative disorders such as Alzheimer's disease.
Collapse
Affiliation(s)
- Yaoming Wang
- Department of Physiology and Neuroscience, Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| | - Kassandra Kisler
- Department of Physiology and Neuroscience, Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| | - Angeliki Maria Nikolakopoulou
- Department of Physiology and Neuroscience, Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| | - Jose A. Fernandez
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
- Division of Hematology/Oncology, Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience, Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
9
|
Liu J, Dean DA. Gene Therapy for Acute Respiratory Distress Syndrome. Front Physiol 2022; 12:786255. [PMID: 35111077 PMCID: PMC8801611 DOI: 10.3389/fphys.2021.786255] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a devastating clinical syndrome that leads to acute respiratory failure and accounts for over 70,000 deaths per year in the United States alone, even prior to the COVID-19 pandemic. While its molecular details have been teased apart and its pathophysiology largely established over the past 30 years, relatively few pharmacological advances in treatment have been made based on this knowledge. Indeed, mortality remains very close to what it was 30 years ago. As an alternative to traditional pharmacological approaches, gene therapy offers a highly controlled and targeted strategy to treat the disease at the molecular level. Although there is no single gene or combination of genes responsible for ARDS, there are a number of genes that can be targeted for upregulation or downregulation that could alleviate many of the symptoms and address the underlying mechanisms of this syndrome. This review will focus on the pathophysiology of ARDS and how gene therapy has been used for prevention and treatment. Strategies for gene delivery to the lung, such as barriers encountered during gene transfer, specific classes of genes that have been targeted, and the outcomes of these approaches on ARDS pathogenesis and resolution will be discussed.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, United States
| | - David A. Dean
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, United States
| |
Collapse
|
10
|
Huuskonen MT, Wang Y, Nikolakopoulou AM, Montagne A, Dai Z, Lazic D, Sagare AP, Zhao Z, Fernandez JA, Griffin JH, Zlokovic BV. Protection of ischemic white matter and oligodendrocytes in mice by 3K3A-activated protein C. J Exp Med 2022; 219:e20211372. [PMID: 34846535 PMCID: PMC8635278 DOI: 10.1084/jem.20211372] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/19/2021] [Accepted: 10/29/2021] [Indexed: 12/13/2022] Open
Abstract
Subcortical white matter (WM) stroke accounts for 25% of all strokes and is the second leading cause of dementia. Despite such clinical importance, we still do not have an effective treatment for ischemic WM stroke, and the mechanisms of WM postischemic neuroprotection remain elusive. 3K3A-activated protein C (APC) is a signaling-selective analogue of endogenous blood protease APC that is currently in development as a neuroprotectant for ischemic stroke patients. Here, we show that 3K3A-APC protects WM tracts and oligodendrocytes from ischemic injury in the corpus callosum in middle-aged mice by activating protease-activated receptor 1 (PAR1) and PAR3. We show that PAR1 and PAR3 were also required for 3K3A-APC's suppression of post-WM stroke microglia and astrocyte responses and overall improvement in neuropathologic and functional outcomes. Our data provide new insights into the neuroprotective APC pathway in the WM and illustrate 3K3A-APC's potential for treating WM stroke in humans, possibly including multiple WM strokes that result in vascular dementia.
Collapse
Affiliation(s)
- Mikko T. Huuskonen
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Yaoming Wang
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Angeliki Maria Nikolakopoulou
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Axel Montagne
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Zhonghua Dai
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Divna Lazic
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Abhay P. Sagare
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Zhen Zhao
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Jose A. Fernandez
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
- Division of Hematology/Oncology, Department of Medicine, University of California, San Diego, San Diego, CA
| | - Berislav V. Zlokovic
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| |
Collapse
|
11
|
Yan A, Pan X, Wen X, Nie X, Li Y. Activated protein C overexpression suppresses the pyroptosis of subarachnoid hemorrhage model cells by regulating the NLRP3 inflammasome pathway. Exp Ther Med 2021; 22:1391. [PMID: 34650639 PMCID: PMC8506940 DOI: 10.3892/etm.2021.10827] [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: 07/20/2021] [Accepted: 09/13/2021] [Indexed: 11/12/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is a condition with a high associated mortality rate that is caused by hemorrhagic stroke. Activated protein C (APC) serves a neuroprotective role in central nervous system diseases. However, its role in SAH remains unclear. The present study aimed to investigate the role of APC and its regulatory mechanism in SAH. The SAH rat model was constructed through internal carotid artery puncture, while the SAH cell model was established via the application of oxygenated hemoglobin. ELISA was performed to detect the level of cytokines, and flow cytometry was used to determine the population of pyroptotic cells. Reverse transcription-quantitative PCR and western blotting were used to examine the relative mRNA and protein levels of APC. APC was silenced using specific APC short hairpin RNA. Neurological functions of rats were estimated using modified Garcia scoring and the balance beam test, while SAH was estimated using modified Sugawara's scoring. The results demonstrated that the expression of APC was significantly decreased, whereas the expression of NLR family pyrin domain-containing 3 (NLRP3) was increased in the SAH rat model in a time-dependent manner. The application of APC recombinant protein 3K3A-APC could significantly ameliorate SAH and improve neurological functions. In addition, 3K3A-APC could inhibit pyroptosis in a dose-dependent manner in the SAH cell model. Moreover, the NLRP3 inhibitor BAY11-7082 could reverse the upregulation of pyroptosis induced by APC-knockdown. Overall, the present study revealed that APC could ameliorate SAH-induced early brain injury by suppressing pyroptosis via inhibition of the NLRP3 inflammasome, which could provide a novel strategy for the treatment of SAH.
Collapse
Affiliation(s)
- Ai Yan
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Hospital of Huzhou Normal University, Huzhou, Zhejiang 313000, P.R. China
| | - Xuyan Pan
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Hospital of Huzhou Normal University, Huzhou, Zhejiang 313000, P.R. China
| | - Xianqiang Wen
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Hospital of Huzhou Normal University, Huzhou, Zhejiang 313000, P.R. China
| | - Xiaohu Nie
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Hospital of Huzhou Normal University, Huzhou, Zhejiang 313000, P.R. China
| | - Yuntao Li
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Hospital of Huzhou Normal University, Huzhou, Zhejiang 313000, P.R. China
| |
Collapse
|
12
|
El Jurdi N, Elhusseini H, Beckman J, DeFor TE, Okoev G, Rogosheske J, Lazaryan A, Weiler K, Bachanova V, Betts BC, Blazar BR, Brunstein CG, He F, Holtan SG, Janakiram M, Gangaraju R, Maakaron J, MacMillan ML, Rashidi A, Warlick ED, Bhatia S, Vercellotti G, Weisdorf DJ, Arora M. High incidence of thromboembolism in patients with chronic GVHD: association with severity of GVHD and donor-recipient ABO blood group. Blood Cancer J 2021; 11:96. [PMID: 34006823 PMCID: PMC8131386 DOI: 10.1038/s41408-021-00488-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/20/2021] [Accepted: 04/29/2021] [Indexed: 02/08/2023] Open
Abstract
Chronic graft-versus-host disease (cGVHD) after allogeneic hematopoietic cell transplantation (HCT) is associated with systemic inflammation and endothelial dysfunction, increasing risk for thromboembolic events (TEE). In 145 adult recipients who developed cGVHD after a matched sibling or umbilical cord blood donor HCT from 2010 to 2018, 32(22%) developed at least 1 TEE event, and 14(10%) developed 2 TEE events. The 5-year cumulative incidence of TEE was 22% (95% CI, 15–29%) with a median time from cGVHD to TEE of 234 days (range, 12–2050). Median time to the development of LE DVT or PE was 107 (range, 12–1925) compared to 450 days (range, 158–1300) for UE DVT. Cumulative incidence of TEE was 9% (95% CI, 0–20%), 17% (95% CI, 9–25%), and 38% (95% CI, 22–55%) in those with mild, moderate, and severe GVHD, respectively. Higher risk for TEE was associated with cGVHD severity (hazard ratio [HR] 4.9, [95% CI, 1.1–22.0]; p = 0.03), non-O-donor to recipient ABO match compared to O-donor to O-recipient match (HR 2.7, [95% CI, 1.0–7.5]; p = 0.053), and personal history of coronary artery disease (HR 2.4, [95% CI, 1.1–5.3]; p = 0.03). TEE was not associated with 2-year non-relapse mortality or 5-year overall survival. Patients with chronic GVHD after allogeneic hematopoietic cell transplantation are at high risk for thromboembolic events occurring years after diagnosis. More severe chronic GVHD, non-O donor-recipient ABO compared to O-O match and personal history of coronary artery disease are associated with higher risk of thromboembolic events.
Collapse
Affiliation(s)
- Najla El Jurdi
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
| | - Heba Elhusseini
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Joan Beckman
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Todd E DeFor
- Biostatistics and Informatics, Clinical and Translational Science Institute, University of Minnesota, Minneapolis, MN, USA
| | - Grigori Okoev
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - John Rogosheske
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Aleksandr Lazaryan
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Kristen Weiler
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Veronika Bachanova
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Brian C Betts
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Bruce R Blazar
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Claudio G Brunstein
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Fiona He
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Shernan G Holtan
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Murali Janakiram
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Radhika Gangaraju
- Department of Pediatrics, University of Alabama, Tuscaloosa, AL, USA
| | - Joseph Maakaron
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Margaret L MacMillan
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Armin Rashidi
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Erica D Warlick
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Smita Bhatia
- Department of Pediatrics, University of Alabama, Tuscaloosa, AL, USA
| | - Gregory Vercellotti
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Daniel J Weisdorf
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Mukta Arora
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
13
|
Lyden PD, Pryor KE, Minigh J, Davis TP, Griffin JH, Levy H, Zlokovic BV. Stroke Treatment With PAR-1 Agents to Decrease Hemorrhagic Transformation. Front Neurol 2021; 12:593582. [PMID: 33790846 PMCID: PMC8005555 DOI: 10.3389/fneur.2021.593582] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 02/08/2021] [Indexed: 12/20/2022] Open
Abstract
Ischemic stroke is the most widespread cause of disability and a leading cause of death in developed countries. To date, the most potent approved treatment for acute stroke is recanalization therapy with thrombolytic drugs such as tissue plasminogen activator (rt-PA or tPA) or endovascular mechanical thrombectomy. Although tPA and thrombectomy are widely available in the United States, it is currently estimated that only 10-20% of stroke patients get tPA treatment, in part due to restrictive selection criteria. Recently, however, tPA and thrombectomy selection criteria have loosened, potentially allowing more patients to qualify. The relatively low rate of treatment may also reflect the perceived risk of brain hemorrhage following treatment with tPA. In translational research and a single patient study, protease activated receptor 1 (PAR-1) targeted therapies given along with thrombolysis and thrombectomy appear to reduce hemorrhagic transformation after recanalization. Such adjuncts may likely enhance the availability of recanalization and encourage more physicians to use the recently expanded selection criteria for applying recanalization therapies. This narrative review discusses stroke therapies, the role of hemorrhagic transformation in producing poor outcomes, and presents the data suggesting that PAR-1 acting agents show promise for decreasing hemorrhagic transformation and improving outcomes.
Collapse
Affiliation(s)
- Patrick D. Lyden
- Department of Physiology and Neuroscience, Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
- *Correspondence: Patrick D. Lyden
| | | | | | - Thomas P. Davis
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Howard Levy
- Howard Levy Consulting LLC, Hopewell, NJ, United States
| | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience, Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
14
|
Vassiliou AG, Kotanidou A, Dimopoulou I, Orfanos SE. Endothelial Damage in Acute Respiratory Distress Syndrome. Int J Mol Sci 2020; 21:ijms21228793. [PMID: 33233715 PMCID: PMC7699909 DOI: 10.3390/ijms21228793] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/14/2020] [Accepted: 11/18/2020] [Indexed: 01/01/2023] Open
Abstract
The pulmonary endothelium is a metabolically active continuous monolayer of squamous endothelial cells that internally lines blood vessels and mediates key processes involved in lung homoeostasis. Many of these processes are disrupted in acute respiratory distress syndrome (ARDS), which is marked among others by diffuse endothelial injury, intense activation of the coagulation system and increased capillary permeability. Most commonly occurring in the setting of sepsis, ARDS is a devastating illness, associated with increased morbidity and mortality and no effective pharmacological treatment. Endothelial cell damage has an important role in the pathogenesis of ARDS and several biomarkers of endothelial damage have been tested in determining prognosis. By further understanding the endothelial pathobiology, development of endothelial-specific therapeutics might arise. In this review, we will discuss the underlying pathology of endothelial dysfunction leading to ARDS and emerging therapies. Furthermore, we will present a brief overview demonstrating that endotheliopathy is an important feature of hospitalised patients with coronavirus disease-19 (COVID-19).
Collapse
Affiliation(s)
- Alice G. Vassiliou
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
| | - Anastasia Kotanidou
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
| | - Ioanna Dimopoulou
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
| | - Stylianos E. Orfanos
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
- 2nd Department of Critical Care, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, 124 62 Athens, Greece
- Correspondence: or ; Tel.: +30-2107-235-521
| |
Collapse
|
15
|
Mukherjee P, Lyden P, Fernández JA, Davis TP, Pryor KE, Zlokovic BV, Griffin JH. 3K3A-Activated Protein C Variant Does Not Interfere With the Plasma Clot Lysis Activity of Tenecteplase. Stroke 2020; 51:2236-2239. [PMID: 32568648 DOI: 10.1161/strokeaha.120.028793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE A recombinant engineered variant of APC (activated protein C), 3K3A-APC, lacks anticoagulant properties (<10%) while preserving APCs anti-inflammatory, anti-apoptotic, and neuroprotective functions and is very promising in clinical trials for ischemic stroke. Therapeutic intervention with single bolus administration of the third-generation tPA (tissue-type plasminogen activator), tenecteplase, is anticipated to be widely adopted for treatment of acute ischemic stroke. 3K3A-APC is well-tolerated in stroke patients dosed with alteplase, and in vitro studies show 3K3A-APC does not interfere with alteplase-induced clot lysis. The purpose of this in vitro study was to assess the influence of 3K3A-APC on tenecteplase-induced clot lysis. METHODS Tenecteplase-mediated lysis of thrombin generated plasma clots of human normal pooled plasma was monitored in the presence of varying doses of 3K3A-APC. The effects on fibrinolysis by tenecteplase and alteplase were compared. RESULTS The presence of 3K3A-APC shortened the time for clot lysis induced by tenecteplase at very low levels but not at higher therapeutic concentrations of tenecteplase. Comparisons of alteplase-mediated clot lysis to tenecteplase clot lysis showed that both thrombolytic agents behaved similarly in the presence of 3K3A-APC. CONCLUSIONS These results indicate that 3K3A-APC does not interfere with tenecteplase's clot lysis function.
Collapse
Affiliation(s)
- Purba Mukherjee
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA (P.M., J.A.F., J.H.G.)
| | - Patrick Lyden
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.)
| | - José A Fernández
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA (P.M., J.A.F., J.H.G.)
| | - Thomas P Davis
- Department of Medical Pharmacology, University of Arizona, Tucson (T.P.D.)
| | | | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA (B.V.Z.)
| | - John H Griffin
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA (P.M., J.A.F., J.H.G.)
| |
Collapse
|
16
|
Zhao XY, Wilmen A, Wang D, Wang X, Bauzon M, Kim JY, Linden L, Li L, Egner U, Marquardt T, Moosmayer D, Tebbe J, Glück JM, Ellinger P, McLean K, Yuan S, Yegneswaran S, Jiang X, Evans V, Gu JM, Schneider D, Zhu Y, Xu Y, Mallari C, Hesslein A, Wang Y, Schmidt N, Gutberlet K, Ruehl-Fehlert C, Freyberger A, Hermiston T, Patel C, Sim D, Mosnier LO, Laux V. Targeted inhibition of activated protein C by a non-active-site inhibitory antibody to treat hemophilia. Nat Commun 2020; 11:2992. [PMID: 32532974 PMCID: PMC7293249 DOI: 10.1038/s41467-020-16720-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 05/15/2020] [Indexed: 02/06/2023] Open
Abstract
Activated protein C (APC) is a plasma serine protease with antithrombotic and cytoprotective functions. Based on the hypothesis that specific inhibition of APC’s anticoagulant but not its cytoprotective activity can be beneficial for hemophilia therapy, 2 types of inhibitory monoclonal antibodies (mAbs) are tested: A type I active-site binding mAb and a type II mAb binding to an exosite on APC (required for anticoagulant activity) as shown by X-ray crystallography. Both mAbs increase thrombin generation and promote plasma clotting. Type I blocks all APC activities, whereas type II preserves APC’s cytoprotective function. In normal monkeys, type I causes many adverse effects including animal death. In contrast, type II is well-tolerated in normal monkeys and shows both acute and prophylactic dose-dependent efficacy in hemophilic monkeys. Our data show that the type II mAb can specifically inhibit APC’s anticoagulant function without compromising its cytoprotective function and offers superior therapeutic opportunities for hemophilia. Activated protein C (APC) is a plasma serine protease with antithrombotic and cytoprotective functions. Here, the authors develop a monoclonal antibody that specifically inhibits APC’s anticoagulant function without compromising its cytoprotective function, and shows efficacy in animal models.
Collapse
Affiliation(s)
- Xiao-Yan Zhao
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA.
| | - Andreas Wilmen
- Biological Research, Bayer AG, 42113, Wuppertal, Germany
| | - Dongli Wang
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xinquan Wang
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Maxine Bauzon
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | - Ji-Yun Kim
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | - Lars Linden
- Biological Research, Bayer AG, 42113, Wuppertal, Germany
| | - Liang Li
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Ursula Egner
- Structural Biology, Bayer AG, 13342, Berlin, Germany
| | | | | | - Jan Tebbe
- Biological Research, Bayer AG, 42113, Wuppertal, Germany
| | | | | | - Kirk McLean
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | - Shujun Yuan
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | | | - Xiaoqiao Jiang
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | - Vince Evans
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | - Jian-Ming Gu
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | - Doug Schneider
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | - Ying Zhu
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | - Yifan Xu
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | - Cornell Mallari
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | | | - Yan Wang
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | - Nicole Schmidt
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | | | | | | | - Terry Hermiston
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | - Chandra Patel
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | - Derek Sim
- US Innovation Center, Bayer, 455 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | - Laurent O Mosnier
- The Scripps Research Institute, 10550 North Torrey Pines Rd., La Jolla, CA, 92037, USA.
| | - Volker Laux
- TRG-Cardiology/Hematology, Bayer AG, Aprather Weg 18a, 42113, Wuppertal, Germany.
| |
Collapse
|
17
|
Hamedani NS, Müller J, Tolle F, Rühl H, Pezeshkpoor B, Liphardt K, Oldenburg J, Mayer G, Pötzsch B. Selective Modulation of the Protease Activated Protein C Using Exosite Inhibiting Aptamers. Nucleic Acid Ther 2020; 30:276-288. [PMID: 32486960 DOI: 10.1089/nat.2020.0844] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Activated protein C (APC) is a serine protease with anticoagulant and cytoprotective activities. Nonanticoagulant APC mutants show beneficial effects as cytoprotective agents. To study, if such biased APC signaling can be achieved by APC-binding ligands, the aptamer technology has been used. A G-quadruplex-containing aptamer, G-NB3, has been selected that binds to the basic exosite of APC with a KD of 0.2 nM and shows no binding to APC-related serine proteases or the zymogen protein C. G-NB3 inhibits the inactivation of activated cofactors V and VIII with IC50 values of 11.6 and 13.1 nM, respectively, without inhibiting the cytoprotective and anti-inflammatory functions of APC as tested using a staurosporine-induced apoptosis assay and a vascular barrier protection assay. In addition, G-NB3 prolongs the plasma half-life of APC through inhibition of APC-serine protease inhibitor complex formation. These physicochemical and functional characteristics qualify G-NB3 as a promising therapeutic agent usable to enhance the cytoprotective functions of APC without increasing the risk of APC-related hemorrhage.
Collapse
Affiliation(s)
- Nasim Shahidi Hamedani
- Institute of Experimental Hematology and Transfusion Medicine, University of Bonn Medical Center, Bonn, Germany
| | - Jens Müller
- Institute of Experimental Hematology and Transfusion Medicine, University of Bonn Medical Center, Bonn, Germany
| | - Fabian Tolle
- Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Heiko Rühl
- Institute of Experimental Hematology and Transfusion Medicine, University of Bonn Medical Center, Bonn, Germany
| | - Behnaz Pezeshkpoor
- Institute of Experimental Hematology and Transfusion Medicine, University of Bonn Medical Center, Bonn, Germany
| | - Kerstin Liphardt
- Institute of Experimental Hematology and Transfusion Medicine, University of Bonn Medical Center, Bonn, Germany
| | - Johannes Oldenburg
- Institute of Experimental Hematology and Transfusion Medicine, University of Bonn Medical Center, Bonn, Germany
| | - Günter Mayer
- Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Bernd Pötzsch
- Institute of Experimental Hematology and Transfusion Medicine, University of Bonn Medical Center, Bonn, Germany
| |
Collapse
|
18
|
Yamashita A, Zhang Y, Sanner MF, Griffin JH, Mosnier LO. C-terminal residues of activated protein C light chain contribute to its anticoagulant and cytoprotective activities. J Thromb Haemost 2020; 18:1027-1038. [PMID: 32017367 PMCID: PMC7380734 DOI: 10.1111/jth.14756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 01/23/2020] [Accepted: 01/28/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Activated protein C (APC) is an important homeostatic blood coagulation protease that conveys anticoagulant and cytoprotective activities. Proteolytic inactivation of factors Va and VIIIa facilitated by cofactor protein S is responsible for APC's anticoagulant effects, whereas cytoprotective effects of APC involve primarily the endothelial protein C receptor (EPCR), protease activated receptor (PAR)1 and PAR3. OBJECTIVE To date, several binding exosites in the protease domain of APC have been identified that contribute to APC's interaction with its substrates but potential contributions of the C-terminus of the light chain have not been studied in detail. METHODS Site-directed Ala-scanning mutagenesis of six positively charged residues within G142-L155 was used to characterize their contributions to APC's anticoagulant and cytoprotective activities. RESULTS AND CONCLUSIONS K151 was involved in protein S dependent-anticoagulant activity of APC with some contribution of K150. 3D structural analysis supported that these two residues were exposed in an extended protein S binding site on one face of APC. Both K150 and K151 were important for PAR1 and PAR3 cleavage by APC, suggesting that this region may also mediate interactions with PARs. Accordingly, APC's cytoprotective activity as determined by endothelial barrier protection was impaired by Ala substitutions of these residues. Thus, both K150 and K151 are involved in APC's anticoagulant and cytoprotective activities. The differential contribution of K150 relative to K151 for protein S-dependent anticoagulant activity and PAR cleavage highlights that binding exosites for protein S binding and for PAR cleavage in the C-terminal region of APC's light chain overlap.
Collapse
Affiliation(s)
- Atsuki Yamashita
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Yuqi Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla
| | - Michel F. Sanner
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Laurent O. Mosnier
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|
19
|
Kant R, Halder SK, Fernández JA, Griffin JH, Milner R. Activated Protein C Attenuates Experimental Autoimmune Encephalomyelitis Progression by Enhancing Vascular Integrity and Suppressing Microglial Activation. Front Neurosci 2020; 14:333. [PMID: 32351356 PMCID: PMC7174764 DOI: 10.3389/fnins.2020.00333] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 03/20/2020] [Indexed: 12/16/2022] Open
Abstract
Background Activated protein C (APC), a serine protease with antithrombotic effects, protects in animal models of ischemic stroke by suppressing inflammation and enhancing vascular integrity, angiogenesis, neurogenesis and neuroprotection. A small number of animal studies suggest it might also have therapeutic potential in multiple sclerosis (MS), though results have been mixed. Based on these conflicting data, the goals of this study were to clarify the therapeutic potential of APC in the experimental autoimmune encephalomyelitis (EAE) model of MS and to determine mechanistically how APC mediates this protective effect. Methods The protective potential of APC was examined in a chronic progressive model of EAE. Vascular breakdown, tight junction protein expression and vascular expression of fibronectin and α5β1 integrin as well as vascularity and glial activation were analyzed using immunofluorescence (IF) of spinal cord sections taken from mice with established EAE. The direct influence of APC on microglial activation was evaluated in vitro by a combination of morphology and MMP-9 expression. Results APC attenuated the progression of EAE, and this was strongly associated at the histopathological level with reduced levels of leukocyte infiltration and concomitant demyelination. Further analysis revealed that APC reduced vascular breakdown which was associated with maintained endothelial expression of the tight junction protein zonula occludens-1 (ZO-1). In addition, APC suppressed microglial activation in this EAE model and in vitro studies revealed that APC strongly inhibited microglial activation at both the morphological level and by the expression of the pro-inflammatory protease MMP-9. Conclusion These findings build on the work of others in demonstrating strong therapeutic potential for APC in the treatment of inflammatory demyelinating disease and suggest that enhancement of vascular integrity and suppression of microglial activation may be important mediators of this protection.
Collapse
Affiliation(s)
- Ravi Kant
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Sebok K Halder
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Jose A Fernández
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - John H Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Richard Milner
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
20
|
Activated protein C ameliorates chronic graft-versus-host disease by PAR1-dependent biased cell signaling on T cells. Blood 2019; 134:776-781. [PMID: 31243040 DOI: 10.1182/blood.2019001259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 06/10/2019] [Indexed: 12/14/2022] Open
Abstract
Soluble thrombomodulin plasma concentrations are elevated in steroid-resistant graft-versus-host disease (GVHD), implying endothelial hypofunctioning for thrombomodulin-dependent generation of activated protein C's (APC) anticoagulant, anti-inflammatory, and antiapoptotic functions. Recombinant thrombomodulin or APC administration decreases acute GVHD, manifested by intense inflammation and tissue destruction. Here, we administered recombinant murine wild-type (WT) APC to mice with established chronic GVHD (cGVHD), a less-inflammatory autoimmune-like disease. WT APC normalized bronchiolitis obliterans-induced pulmonary dysfunction. Signaling-selective APC variants (3A-APC [APC with lysine 191-193 replaced with 3 alanines] or 5A-APC [APC with lysine 191-193 replaced with 3 alanines and arginine 229/230 replaced with 2 alanines]) with normal cytoprotective properties, but greatly reduced anticoagulant activity, provided similar results. Mechanistically, WT APC and signaling-selective variants reduced T follicular helper cells, germinal center formation, immunoglobulin, and collagen deposition. WT APC can potentially cleave protease-activated receptor 1 (PAR1) at Arg41 or Arg46, the latter causing anti-inflammatory signaling. cGVHD was reduced in recipients of T cells from WT PAR1 or mutated Gln41-PAR1 donors but not from mutated Gln46-PAR1 donors. These data implicate donor T-cell APC-induced noncanonical cleavage at Arg46-PAR1, which is known to confer cytoprotective and anti-inflammatory activities. Together, these data indicate that APC anticoagulant activity is dispensable, whereas anti-inflammatory signaling and cytoprotective cell signaling by APC are essential. Because a phase 2 ischemic stroke clinical trial did not raise any safety issues for 3A-APC treatment, our studies provide a foundational platform for testing in clinical cGVHD therapy.
Collapse
|
21
|
Rajput PS, Lamb JA, Fernández JÁ, Bai J, Pereira BR, Lei IF, Leung J, Griffin JH, Lyden PD. Neuroprotection and vasculoprotection using genetically targeted protease-ligands. Brain Res 2019; 1715:13-20. [PMID: 30880117 DOI: 10.1016/j.brainres.2019.03.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 03/05/2019] [Accepted: 03/13/2019] [Indexed: 12/26/2022]
Abstract
Thrombin and activated protein C (APC) are known coagulation factors that exhibit profound effects in brain by acting on the protease activated receptor (PAR). The wild type (WT) proteases appear to impact cell survival powerfully, and therapeutic forms of APC are under development. Engineered recombinant thrombin or APC were designed to separate their procoagulant or anticoagulant effects from their cytoprotective properties. We measured vascular disruption and neuronal degeneration after a standard rodent filament stroke model. For comparison to a robust anticoagulant, we used a GpIIb/IIIa inhibitor, GR144053. During 2 h MCAo both WT murine APC and its mutant, 5A-APC, significantly decreased neuronal death 30 min after reperfusion. During 4 h MCAo, only 5A-APC significantly protected neurons but both WT-APC and 5A-APC exacerbated vascular disruption during 4 h MCAo. Human APC mutants appeared to reduce 24 h neuronal injury significantly when given after 2 h delay after MCAo. In contrast, 24 h vascular damage was worsened by high doses of WT and mutant APCs, although only statistically significantly for high dose 3K3A-APC. Mutated thrombin worsened vascular damage significantly without affecting neuron damage. GR144053 failed to ameliorate vascular disruption or neuronal injury despite significant anticoagulation. Differential effects on neurons and the vasculature were demonstrated using wild-type and mutated proteases. The mutants murine 3K3A-APC and 5A-APC protected neurons in this rodent model but in high doses worsened vascular leakage. Cytoactive effects of plasma proteases may be separated from their coagulation effects. Further studies should explore impact of dose and timing on cytoactive and vasculoactive properties of these drugs.
Collapse
Affiliation(s)
- Padmesh S Rajput
- Department of Neurology, Cedars Sinai Medical Center, 127 S San Vicente Blvd, Los Angeles, CA 90048, United States
| | - Jessica A Lamb
- Department of Neurology, Cedars Sinai Medical Center, 127 S San Vicente Blvd, Los Angeles, CA 90048, United States
| | - Jose Á Fernández
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| | - Jilin Bai
- Department of Neurology, Cedars Sinai Medical Center, 127 S San Vicente Blvd, Los Angeles, CA 90048, United States
| | - Benedict R Pereira
- Department of Neurology, Cedars Sinai Medical Center, 127 S San Vicente Blvd, Los Angeles, CA 90048, United States
| | - I-Farn Lei
- Department of Neurology, Cedars Sinai Medical Center, 127 S San Vicente Blvd, Los Angeles, CA 90048, United States
| | - Jennifer Leung
- Department of Neurology, Cedars Sinai Medical Center, 127 S San Vicente Blvd, Los Angeles, CA 90048, United States
| | - John H Griffin
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| | - Patrick D Lyden
- Department of Neurology, Cedars Sinai Medical Center, 127 S San Vicente Blvd, Los Angeles, CA 90048, United States.
| |
Collapse
|
22
|
Activated Protein C in Cutaneous Wound Healing: From Bench to Bedside. Int J Mol Sci 2019; 20:ijms20040903. [PMID: 30791425 PMCID: PMC6412604 DOI: 10.3390/ijms20040903] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/14/2019] [Accepted: 02/16/2019] [Indexed: 12/12/2022] Open
Abstract
Independent of its well-known anticoagulation effects, activated protein C (APC) exhibits pleiotropic cytoprotective properties. These include anti-inflammatory actions, anti-apoptosis, and endothelial and epithelial barrier stabilisation. Such beneficial effects have made APC an attractive target of research in a plethora of physiological and pathophysiological processes. Of note, the past decade or so has seen the emergence of its roles in cutaneous wound healing-a complex process involving inflammation, proliferation and remodelling. This review will highlight APC's functions and mechanisms, and detail its pre-clinical and clinical studies on cutaneous wound healing.
Collapse
|
23
|
Lazic D, Sagare AP, Nikolakopoulou AM, Griffin JH, Vassar R, Zlokovic BV. 3K3A-activated protein C blocks amyloidogenic BACE1 pathway and improves functional outcome in mice. J Exp Med 2019; 216:279-293. [PMID: 30647119 PMCID: PMC6363429 DOI: 10.1084/jem.20181035] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 10/05/2018] [Accepted: 10/30/2018] [Indexed: 12/29/2022] Open
Abstract
3K3A-activated protein C (APC), a cell-signaling analogue of endogenous blood serine protease APC, exerts vasculoprotective, neuroprotective, and anti-inflammatory activities in rodent models of stroke, brain injury, and neurodegenerative disorders. 3K3A-APC is currently in development as a neuroprotectant in patients with ischemic stroke. Here, we report that 3K3A-APC inhibits BACE1 amyloidogenic pathway in a mouse model of Alzheimer's disease (AD). We show that a 4-mo daily treatment of 3-mo-old 5XFAD mice with murine recombinant 3K3A-APC (100 µg/kg/d i.p.) prevents development of parenchymal and cerebrovascular amyloid-β (Aβ) deposits by 40-50%, which is mediated through NFκB-dependent transcriptional inhibition of BACE1, resulting in blockade of Aβ generation in neurons overexpressing human Aβ-precursor protein. Consistent with reduced Aβ deposition, 3K3A-APC normalized hippocampus-dependent behavioral deficits and cerebral blood flow responses, improved cerebrovascular integrity, and diminished neuroinflammatory responses. Our data suggest that 3K3A-APC holds potential as an effective anti-Aβ prevention therapy for early-stage AD.
Collapse
Affiliation(s)
- Divna Lazic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA.,Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA.,Department of Neurobiology, Institute for Biological Research, University of Belgrade, Belgrade, Republic of Serbia
| | - Abhay P Sagare
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA.,Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Angeliki M Nikolakopoulou
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA.,Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - John H Griffin
- The Scripps Research Institute, La Jolla, CA.,Department of Medicine, University of California, San Diego, San Diego, CA
| | - Robert Vassar
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA .,Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA
| |
Collapse
|
24
|
Eliwan HO, Watson WRG, Regan I, Philbin B, O'Hare FM, Strickland T, O'Neill A, O'Rourke M, Blanco A, Healy M, Nolan B, Smith O, Molloy EJ. Pediatric Intensive Care: Immunomodulation With Activated Protein C ex vivo. Front Pediatr 2019; 7:386. [PMID: 31612119 PMCID: PMC6776988 DOI: 10.3389/fped.2019.00386] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 09/06/2019] [Indexed: 11/21/2022] Open
Abstract
Objective: Sepsis is major cause of morbidity and mortality in the Pediatric Intensive Care Unit (PICU). PICU patients may develop transient immune deficiency during sepsis. Activated Protein C (APC) has significant anti-inflammatory and cytoprotective effects. Clinical trials of APC in adult sepsis initially showed improved outcome but recent trials showed no benefit in adults or children. We aimed to assess the effects of APC treatment on innate immune responses in children. Design and Subjects: We compared neutrophil and monocyte responses to lipopolysaccharide (LPS) with and without APC treatment in PICU patients at the time of evaluation for sepsis compared with healthy adults and age-matched pediatric controls. We used flow cytometry to examine cell activation (CD11b expression), function [intracellular reactive oxygen intermediate (ROI) release] and LPS recognition [Toll like Receptor 4 (TLR4) expression]. Results: PICU patients had significantly decreased protein c levels and LPS responses compared with adult and pediatric controls for all parameters. APC reduced LPS-induced neutrophil PICU TLR4 and adult ROI (p < 0.05). PICU non-survivors had increased LPS induced neutrophil and monocyte ROI production vs. survivors which was significantly reduced by APC. Conclusion: PICU patients demonstrate significantly reduced endotoxin reactivity which may predispose them to sepsis and alter effective antibacterial responses. APC reduces LPS-induced ROI production in adults and may have a role in treating severely compromised PICU patients especially given that newer APC forms are associated with decreased bleeding risk and enhanced anti-inflammatory effects.
Collapse
Affiliation(s)
- Hassan O Eliwan
- Neonatology, National Maternity Hospital, Dublin, Ireland.,National Children's Research Centre, Dublin, Ireland.,Paediatrics, Royal College of Surgeons in Ireland, Dublin, Ireland.,University College Dublin School of Medicine and Medical Sciences, Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - William R G Watson
- University College Dublin School of Medicine and Medical Sciences, Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Irene Regan
- Haematology, Our Lady's Children's Hospital, Dublin, Ireland
| | - Brian Philbin
- Haematology, Our Lady's Children's Hospital, Dublin, Ireland
| | - Fiona M O'Hare
- Neonatology, National Maternity Hospital, Dublin, Ireland.,National Children's Research Centre, Dublin, Ireland.,University College Dublin School of Medicine and Medical Sciences, Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Tammy Strickland
- Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland
| | - Amanda O'Neill
- University College Dublin School of Medicine and Medical Sciences, Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | | | - Alfonso Blanco
- University College Dublin School of Medicine and Medical Sciences, Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Martina Healy
- Critical Care, Our Lady's Children's Hospital, Dublin, Ireland
| | - Beatrice Nolan
- Haematology, Our Lady's Children's Hospital, Dublin, Ireland
| | - Owen Smith
- Haematology, Our Lady's Children's Hospital, Dublin, Ireland
| | - Eleanor J Molloy
- Neonatology, National Maternity Hospital, Dublin, Ireland.,National Children's Research Centre, Dublin, Ireland.,Paediatrics, Royal College of Surgeons in Ireland, Dublin, Ireland.,University College Dublin School of Medicine and Medical Sciences, Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.,Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Neonatology, Our Lady's Children's Hospital, Dublin, Ireland.,Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland
| |
Collapse
|
25
|
Griffin JH, Zlokovic BV, Mosnier LO. Activated protein C, protease activated receptor 1, and neuroprotection. Blood 2018; 132:159-169. [PMID: 29866816 PMCID: PMC6043978 DOI: 10.1182/blood-2018-02-769026] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/01/2018] [Indexed: 02/08/2023] Open
Abstract
Protein C is a plasma serine protease zymogen whose active form, activated protein C (APC), exerts potent anticoagulant activity. In addition to its antithrombotic role as a plasma protease, pharmacologic APC is a pleiotropic protease that activates diverse homeostatic cell signaling pathways via multiple receptors on many cells. Engineering of APC by site-directed mutagenesis provided a signaling selective APC mutant with 3 Lys residues replaced by 3 Ala residues, 3K3A-APC, that lacks >90% anticoagulant activity but retains normal cell signaling activities. This 3K3A-APC mutant exerts multiple potent neuroprotective activities, which require the G-protein-coupled receptor, protease activated receptor 1. Potent neuroprotection in murine ischemic stroke models is linked to 3K3A-APC-induced signaling that arises due to APC's cleavage in protease activated receptor 1 at a noncanonical Arg46 site. This cleavage causes biased signaling that provides a major explanation for APC's in vivo mechanism of action for neuroprotective activities. 3K3A-APC appeared to be safe in ischemic stroke patients and reduced bleeding in the brain after tissue plasminogen activator therapy in a recent phase 2 clinical trial. Hence, it merits further clinical testing for its efficacy in ischemic stroke patients. Recent studies using human fetal neural stem and progenitor cells show that 3K3A-APC promotes neurogenesis in vitro as well as in vivo in the murine middle cerebral artery occlusion stroke model. These recent advances should encourage translational research centered on signaling selective APC's for both single-agent therapies and multiagent combination therapies for ischemic stroke and other neuropathologies.
Collapse
Affiliation(s)
- John H Griffin
- The Scripps Research Institute, La Jolla, CA
- Department of Medicine, University of California, San Diego, CA; and
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, Los Angeles, CA
| | | |
Collapse
|
26
|
PAR1 agonists stimulate APC-like endothelial cytoprotection and confer resistance to thromboinflammatory injury. Proc Natl Acad Sci U S A 2018; 115:E982-E991. [PMID: 29343648 DOI: 10.1073/pnas.1718600115] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Stimulation of protease-activated receptor 1 (PAR1) on endothelium by activated protein C (APC) is protective in several animal models of disease, and APC has been used clinically in severe sepsis and wound healing. Clinical use of APC, however, is limited by its immunogenicity and its anticoagulant activity. We show that a class of small molecules termed "parmodulins" that act at the cytosolic face of PAR1 stimulates APC-like cytoprotective signaling in endothelium. Parmodulins block thrombin generation in response to inflammatory mediators and inhibit platelet accumulation on endothelium cultured under flow. Evaluation of the antithrombotic mechanism showed that parmodulins induce cytoprotective signaling through Gβγ, activating a PI3K/Akt pathway and eliciting a genetic program that includes suppression of NF-κB-mediated transcriptional activation and up-regulation of select cytoprotective transcripts. STC1 is among the up-regulated transcripts, and knockdown of stanniocalin-1 blocks the protective effects of both parmodulins and APC. Induction of this signaling pathway in vivo protects against thromboinflammatory injury in blood vessels. Small-molecule activation of endothelial cytoprotection through PAR1 represents an approach for treatment of thromboinflammatory disease and provides proof-of-principle for the strategy of targeting the cytoplasmic surface of GPCRs to achieve pathway selective signaling.
Collapse
|
27
|
Wildhagen K, Lutgens E, Loubele S, Cate HT, Nicolaes G. The structure-function relationship of activated protein C. Thromb Haemost 2017; 106:1034-45. [DOI: 10.1160/th11-08-0522] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 09/22/2011] [Indexed: 11/05/2022]
Abstract
SummaryProtein C is the central enzyme of the natural anticoagulant pathway and its activated form APC (activated protein C) is able to proteolyse non-active as well as active coagulation factors V and VIII. Proteolysis renders these cofactors inactive, resulting in an attenuation of thrombin formation and overall down-regulation of coagulation. Presences of the APC cofactor, protein S, thrombomodulin, endothelial protein C receptor and a phospholipid surface are important for the expression of anticoagulant APC activity. Notably, APC also has direct cytoprotective effects on cells: APC is able to protect the endothelial barrier function and expresses anti-inflammatory and anti-apoptotic activities. Exact molecular mechanisms have thus far not been completely described but it has been shown that both the protease activated receptor 1 and EPCR are essential for the cytoprotective activity of APC. Recently it was shown that also other receptors like sphingosine 1 phosphate receptor 1, Cd11b/CD18 and tyrosine kinase with immunoglobulin-like and EGFlike domains 2 are likewise important for APC signalling. Mutagenesis studies are being performed to map the various APC functions and interactions onto its 3D structure and to dissect anticoagulant and cytoprotective properties. The results of these studies have provided a wealth of structure-function information. With this review we describe the state-of-the-art of the intricate structure-function relationships of APC, a protein that harbours several important functions for the maintenance of both humoral and tissue homeostasis.Lessons from natural and engineered mutations
Collapse
|
28
|
Colling ME, Bendapudi PK. Purpura Fulminans: Mechanism and Management of Dysregulated Hemostasis. Transfus Med Rev 2017; 32:69-76. [PMID: 29157918 DOI: 10.1016/j.tmrv.2017.10.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/21/2017] [Accepted: 10/13/2017] [Indexed: 01/30/2023]
Abstract
Purpura fulminans (PF) is a highly thrombotic subtype of disseminated intravascular coagulation that can accompany severe bacterial, and more rarely, viral infections. PF is associated with an extremely high mortality rate, and patients often die of overwhelming multisystemic thrombosis rather than septic shock. Survivors typically experience amputation of involved extremities and significant scarring in affected areas. Despite the devastating clinical course associated with this hemostatic complication of infection, the mechanism of PF remains poorly understood. Severe acquired deficiency of protein C and dysfunction of the protein C-thrombomodulin pathway as well as other systems that exert a negative regulatory effect on coagulation have been implicated. Management of PF involves treatment of the underlying infection, aggressive anticoagulation, and robust transfusion support aimed at correcting acquired deficiencies in natural anticoagulant proteins. In this review, we address the diagnosis and management of PF with a focus on a rational approach to this condition informed by the available data. Proposed mechanisms underlying the dysregulation of coagulation seen in PF are also covered, and implications for therapy are discussed.
Collapse
Affiliation(s)
- Meaghan E Colling
- Department of Medicine, Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Pavan K Bendapudi
- Harvard Medical School, Boston, MA; Division of Hematology, Massachusetts General Hospital, Boston, MA; Blood Transfusion Service, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
29
|
Gorbacheva LR, Kiseleva EV, Savinkova IG, Strukova SM. A new concept of action of hemostatic proteases on inflammation, neurotoxicity, and tissue regeneration. BIOCHEMISTRY (MOSCOW) 2017; 82:778-790. [DOI: 10.1134/s0006297917070033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
30
|
Healy LD, Puy C, Fernández JA, Mitrugno A, Keshari RS, Taku NA, Chu TT, Xu X, Gruber A, Lupu F, Griffin JH, McCarty OJT. Activated protein C inhibits neutrophil extracellular trap formation in vitro and activation in vivo. J Biol Chem 2017; 292:8616-8629. [PMID: 28408624 DOI: 10.1074/jbc.m116.768309] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 04/11/2017] [Indexed: 12/11/2022] Open
Abstract
Activated protein C (APC) is a multifunctional serine protease with anticoagulant, cytoprotective, and anti-inflammatory activities. In addition to the cytoprotective effects of APC on endothelial cells, podocytes, and neurons, APC cleaves and detoxifies extracellular histones, a major component of neutrophil extracellular traps (NETs). NETs promote pathogen clearance but also can lead to thrombosis; the pathways that negatively regulate NETosis are largely unknown. Thus, we studied whether APC is capable of directly inhibiting NETosis via receptor-mediated cell signaling mechanisms. Here, by quantifying extracellular DNA or myeloperoxidase, we demonstrate that APC binds human leukocytes and prevents activated platelet supernatant or phorbol 12-myristate 13-acetate (PMA) from inducing NETosis. Of note, APC proteolytic activity was required for inhibiting NETosis. Moreover, antibodies against the neutrophil receptors endothelial protein C receptor (EPCR), protease-activated receptor 3 (PAR3), and macrophage-1 antigen (Mac-1) blocked APC inhibition of NETosis. Select mutations in the Gla and protease domains of recombinant APC caused a loss of NETosis. Interestingly, pretreatment of neutrophils with APC prior to induction of NETosis inhibited platelet adhesion to NETs. Lastly, in a nonhuman primate model of Escherichia coli-induced sepsis, pretreatment of animals with APC abrogated release of myeloperoxidase from neutrophils, a marker of neutrophil activation. These findings suggest that the anti-inflammatory function of APC at therapeutic concentrations may include the inhibition of NETosis in an EPCR-, PAR3-, and Mac-1-dependent manner, providing additional mechanistic insight into the diverse functions of neutrophils and APC in disease states including sepsis.
Collapse
Affiliation(s)
- Laura D Healy
- From the Departments of Cell, Developmental & Cancer Biology and
| | - Cristina Puy
- Biomedical Engineering, Oregon Health & Science University, Portland, Oregon 97230
| | - José A Fernández
- the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, and
| | - Annachiara Mitrugno
- Biomedical Engineering, Oregon Health & Science University, Portland, Oregon 97230
| | - Ravi S Keshari
- the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | - Nyiawung A Taku
- Biomedical Engineering, Oregon Health & Science University, Portland, Oregon 97230
| | - Tiffany T Chu
- Biomedical Engineering, Oregon Health & Science University, Portland, Oregon 97230
| | - Xiao Xu
- the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, and
| | - András Gruber
- Biomedical Engineering, Oregon Health & Science University, Portland, Oregon 97230
| | - Florea Lupu
- the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | - John H Griffin
- the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, and
| | - Owen J T McCarty
- From the Departments of Cell, Developmental & Cancer Biology and.,Biomedical Engineering, Oregon Health & Science University, Portland, Oregon 97230
| |
Collapse
|
31
|
Griffin JH, Fernández JA, Lyden PD, Zlokovic BV. Activated protein C promotes neuroprotection: mechanisms and translation to the clinic. Thromb Res 2017; 141 Suppl 2:S62-4. [PMID: 27207428 DOI: 10.1016/s0049-3848(16)30368-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Activated protein C (APC) is a plasma serine protease that is capable of antithrombotic, anti-inflammatory, anti-apoptotic, and cell-signaling activities. Animal injury studies show that recombinant APC and some of its mutants are remarkably therapeutic for a wide range of injuries. In particular, for neurologic injuries, APC reduces damage caused by ischemia/reperfusion in the brain, by acute brain trauma, and by chronic neurodegenerative conditions. For these neuroprotective effects, APC requires endothelial cell protein C receptor. APC activates cell signaling networks with alterations in gene expression profiles by activating protease activated receptors 1 and 3. To minimize APC-induced bleeding risk, APC variants were engineered to lack > 90% anticoagulant activity but retain normal cell signaling. The neuroprotective APC mutant, 3K3A-APC which has Lys191-193 mutated to Ala191-193, is very neuroprotective and it is currently in clinical trials for ischemic stroke.
Collapse
Affiliation(s)
- John H Griffin
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA; Department of Medicine, Division of Hematology/Oncology, University of California San Diego, San Diego, CA, USA.
| | - José A Fernández
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Patrick D Lyden
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Berislav V Zlokovic
- Department of Neurosurgery, University of Southern California, Keck School of Medicine, Los Angeles, CA; Department of Neurosurgery, Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
32
|
Wong VM, Côté O, Bienzle D, Hayes MA, Wood RD. Endothelial protein C receptor-dependent antichemotactic effects of canine protein C. Am J Vet Res 2017; 78:186-194. [PMID: 28140640 DOI: 10.2460/ajvr.78.2.186] [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: 11/20/2022]
Abstract
OBJECTIVE To determine whether canine protein C (CnPC) had antichemotactic effects on canine neutrophils, whether endothelial protein C receptor (EPCR) was expressed on canine neutrophils, and the role of EPCR in neutrophil chemotaxis. SAMPLE Neutrophils isolated from blood samples from healthy dogs (n = 6) and sick dogs with (2) or without (3) an inflammatory leukogram. PROCEDURES Neutrophils were analyzed by reverse transcriptase PCR assay and flow cytometry for detection of EPCR mRNA and protein expression, respectively. Neutrophils were incubated with CnPC zymogen or canine activated protein C (CnAPC), with or without RCR-379 (an anti-human EPCR antibody). Neutrophils were then allowed to migrate through a filter membrane toward a chemokine. Untreated neutrophils served as positive control samples. Migration was quantified by fluorescence measurement, and chemotaxis index (Chx) values (fluorescence of test sample/fluorescence of positive control sample) were computed. RESULTS The cDNA for EPCR was amplified, and EPCR expression was detected on neutrophil surfaces. Obtained Chx values were significantly higher in cells treated with RCR-379 than in cells treated with CnPC or CnAPC alone. The Chx values for neutrophils treated with RCR-379 were not significantly different from 1, whereas those for neutrophils treated without RCR-379 were significantly less than 1. The effects of RCR-379 on neutrophil migration were independent of concentration or activation status of protein C. CONCLUSIONS AND CLINICAL RELEVANCE Canine neutrophils expressed EPCR, and inhibition of neutrophil chemotaxis by CnPC and CnAPC depended on EPCR. Interventions with EPCR signaling may have therapeutic application in dogs.
Collapse
|
33
|
Lyden P, Weymer S, Coffey C, Cudkowicz M, Berg S, O'Brien S, Fisher M, Haley EC, Khatri P, Saver J, Levine S, Levy H, Rymer M, Wechsler L, Jadhav A, McNeil E, Waddy S, Pryor K. Selecting Patients for Intra-Arterial Therapy in the Context of a Clinical Trial for Neuroprotection. Stroke 2016; 47:2979-2985. [PMID: 27803392 DOI: 10.1161/strokeaha.116.013881] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 08/12/2016] [Accepted: 09/12/2016] [Indexed: 02/02/2023]
Abstract
BACKGROUND AND PURPOSE The advent of intra-arterial neurothrombectomy (IAT) for acute ischemic stroke opens a potentially transformative opportunity to improve neuroprotection studies. Combining a putative neuroprotectant with recanalization could produce more powerful trials but could introduce heterogeneity and adverse event possibilities. We sought to demonstrate feasibility of IAT in neuroprotectant trials by defining IAT selection criteria for an ongoing neuroprotectant clinical trial. METHODS The study drug, 3K3A-APC, is a pleiotropic cytoprotectant and may reduce thrombolysis-associated hemorrhage. The NeuroNEXT trial NN104 (RHAPSODY) is designed to establish a maximally tolerated dose of 3K3A-APC. Each trial site provided their IAT selection criteria. An expert panel reviewed site criteria and published evidence. Finally, the trial leadership designed IAT selection criteria. RESULTS Derived selection criteria reflected consistency among the sites and comparability to published IAT trials. A protocol amendment allowing IAT (and relaxed age, National Institutes of Health Stroke Scale, and time limits) in the RHAPSODY trial was implemented on June 15, 2015. Recruitment before and after the amendment improved from 8 enrolled patients (601 screened, 1.3%) to 51 patients (821 screened, 6.2%; odds ratio [95% confidence limit] of 4.9 [2.3-10.4]; P<0.001). Gross recruitment was 0.11 patients per site month versus 0.43 patients per site per month, respectively, before and after the amendment. CONCLUSIONS It is feasible to include IAT in a neuroprotectant trial for acute ischemic stroke. Criteria are presented for including such patients in a manner that is consistent with published evidence for IAT while still preserving the ability to test the role of the putative neuroprotectant. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT02222714.
Collapse
Affiliation(s)
- Patrick Lyden
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.).
| | - Sara Weymer
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - Chris Coffey
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - Merit Cudkowicz
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - Samantha Berg
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - Sarah O'Brien
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - Marc Fisher
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - E Clarke Haley
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - Pooja Khatri
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - Jeff Saver
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - Steven Levine
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - Howard Levy
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - Marilyn Rymer
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - Lawrence Wechsler
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - Ashutosh Jadhav
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - Elizabeth McNeil
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - Salina Waddy
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| | - Kent Pryor
- From the Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA (P.L.); ZZ Biotech, LLC, Houston, TX (S.W., H.L., K.P.); Department of Biostatistics, University of Iowa, Iowa City (C.C., S.O.); Neurological Clinical Research Institute, Massachusetts General Hospital, Boston (M.C., S.B.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.); Department of Neurology, University of Virginia, Charlottesville (E.C.H.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (P.K.); Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles (J.S.); Department of Neurology, State University of New York Downstate Medical Center, Brooklyn (S.L.); Department of Neurology, University of Kansas Hospital, Kansas City (M.R.); Department of Neurology, University of Pittsburgh Medical School, PA (L.W., A.J.); and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (E.M., S.W.)
| |
Collapse
|
34
|
Griffin JH, Mosnier LO, Fernández JA, Zlokovic BV. 2016 Scientific Sessions Sol Sherry Distinguished Lecturer in Thrombosis: Thrombotic Stroke: Neuroprotective Therapy by Recombinant-Activated Protein C. Arterioscler Thromb Vasc Biol 2016; 36:2143-2151. [PMID: 27758767 PMCID: PMC5119536 DOI: 10.1161/atvbaha.116.308038] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 09/21/2016] [Indexed: 01/19/2023]
Abstract
APC (activated protein C), derived from the plasma protease zymogen, is antithrombotic and anti-inflammatory. In preclinical injury models, recombinant APC provides neuroprotection for multiple injuries, including ischemic stroke. APC acts directly on brain endothelial cells and neurons by initiating cell signaling that requires multiple receptors. Two or more major APC receptors mediate APC's neuroprotective cell signaling. When bound to endothelial cell protein C receptor, APC can cleave protease-activated receptor 1, causing biased cytoprotective signaling that reduces ischemia-induced injury. Pharmacological APC alleviates bleeding induced by tissue-type plasminogen activator in murine ischemic stroke studies. Remarkably, APC's signaling promotes neurogenesis. The signaling-selective recombinant variant of APC, 3K3A-APC, was engineered to lack most of the APC's anticoagulant activity but retain APC's cell signaling actions. Recombinant 3K3A-APC is in ongoing National Institutes of Health (NIH)-funded clinical trials for ischemic stroke.
Collapse
Affiliation(s)
- John H Griffin
- From the Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA (J.H.G., L.O.M., J.A.F.); Division of Hematology/Oncology, Department of Medicine, University of California, San Diego (J.H.G.); and Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles (B.V.Z.).
| | - Laurent O Mosnier
- From the Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA (J.H.G., L.O.M., J.A.F.); Division of Hematology/Oncology, Department of Medicine, University of California, San Diego (J.H.G.); and Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles (B.V.Z.)
| | - José A Fernández
- From the Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA (J.H.G., L.O.M., J.A.F.); Division of Hematology/Oncology, Department of Medicine, University of California, San Diego (J.H.G.); and Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles (B.V.Z.)
| | - Berislav V Zlokovic
- From the Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA (J.H.G., L.O.M., J.A.F.); Division of Hematology/Oncology, Department of Medicine, University of California, San Diego (J.H.G.); and Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles (B.V.Z.)
| |
Collapse
|
35
|
Abstract
Protease signaling in cells elicits multiple physiologically important responses via protease-activated receptors (PARs). There are 4 members of this family of G-protein-coupled receptors (PAR1-4). PARs are activated by proteolysis of the N terminus to reveal a tethered ligand. The rate-limiting step of PAR signaling is determined by the efficiency of proteolysis of the N terminus, which is regulated by allosteric binding sites, cofactors, membrane localization, and receptor dimerization. This ultimately controls the initiation of PAR signaling. In addition, these factors also control the cellular response by directing signaling toward G-protein or β-arrestin pathways. PAR1 signaling on endothelial cells is controlled by the activating protease and heterodimerization with PAR2 or PAR3. As a consequence, the genetic and epigenetic control of PARs and their cofactors in physiologic and pathophysiologic conditions have the potential to influence cellular behavior. Recent studies have uncovered polymorphisms that result in PAR4 sequence variants with altered reactivity that interact to influence platelet response. This further demonstrates how interactions within the plasma membrane can control the physiological output. Understanding the structural rearrangement following PAR activation and how PARs are allosterically controlled within the plasma membrane will determine how best to target this family of receptors therapeutically. The purpose of this article is to review how signaling from PARs is influenced by alternative cleavage sites and the physical interactions within the membrane. Going forward, it will be important to relate the altered signaling to the molecular arrangement of PARs in the cell membrane and to determine how these may be influenced genetically.
Collapse
|
36
|
Bhat V, von Drygalski A, Gale AJ, Griffin JH, Mosnier LO. Improved coagulation and haemostasis in haemophilia with inhibitors by combinations of superFactor Va and Factor VIIa. Thromb Haemost 2015; 115:551-61. [PMID: 26466980 DOI: 10.1160/th15-07-0525] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/23/2015] [Indexed: 01/14/2023]
Abstract
Bypassing inhibitors in haemophilia patients is limited to activated (a) Factor(F)VII products. We introduced "FVa activity augmentation" as another bypassing strategy and studied effects of an engineered FVa variant designated superFVa. Procoagulant and clot stabilising properties of superFVa and recombinant human (rh)FVIIa, either alone or in combination, were studied in thrombin generation and clot lysis assays in normal human plasma (NHP) with or without anti-FVIII inhibitors, in haemophilia plasma, and in FVIII-deficient mice or in wild-type mice with anti-FVIII inhibitors. SuperFVa was as effective as rhFVIIa to improve thrombin generation or clot lysis. Furthermore, procoagulant effects were significantly enhanced when these compounds were combined. RhFVIIa at 40 nM (a therapeutic concentration) improved thrombin generation mildly, but markedly improved thrombin generation when combined with a low concentration (e. g. 3 nM) of superFVa. In clot lysis studies, the concentration of rhFVIIa to normalise clot lysis times could be reduced by 100-fold (e. g. from 40 nM to 0.4 nM) when combined with a low concentration (0.37 nM) of superFVa. In haemostasis studies of FVIII-deficient mice, blood loss was dose-dependently reduced by either superFVa or rhFVIIa. SuperFVa (200 U/kg) corrected mean blood loss indistinguishably from rhFVIII. Blood loss correction by rhFVIIa was greatly improved when combined with superFVa. Similar blood loss correction results were observed for therapies in wild-type mice after infusion with anti-FVIII inhibitors. Thus, superFVa may be an effective procoagulant agent in the setting of haemophilia with inhibitors and it merits further evaluation for new bypassing strategies.
Collapse
Affiliation(s)
| | - Annette von Drygalski
- Annette von Drygalski, MD, Pharm D, The Scripps Research Institute, Department of Molecular and Experimental Medicine, MEM 180, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA, Tel.: +1 858 784 8220, Fax: +1 858 784 2243, E-mail:
| | | | | | | |
Collapse
|
37
|
Bouwens EAM, Stavenuiter F, Mosnier LO. Cell painting with an engineered EPCR to augment the protein C system. Thromb Haemost 2015; 114:1144-55. [PMID: 26272345 DOI: 10.1160/th15-01-0079] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/28/2015] [Indexed: 11/05/2022]
Abstract
The protein C (PC) system conveys beneficial anticoagulant and cytoprotective effects in numerous in vivo disease models. The endothelial protein C receptor (EPCR) plays a central role in these pathways as cofactor for PC activation and by enhancing activated protein C (APC)-mediated protease-activated receptor (PAR) activation. During inflammatory disease, expression of EPCR on cell membranes is often diminished thereby limiting PC activation and APC's effects on cells. Here a caveolae-targeting glycosylphosphatidylinositol (GPI)-anchored EPCR (EPCR-GPI) was engineered to restore EPCR's bioavailability via "cell painting." The painting efficiency of EPCR-GPI on EPCR-depleted endothelial cells was time- and dose-dependent. The EPCR-GPI bioavailability after painting was long lasting since EPCR surface levels reached 400 % of wild-type cells after 2 hours and remained > 200 % for 24 hours. EPCR-GPI painting conveyed APC binding to EPCR-depleted endothelial cells where EPCR was lost due to shedding or shRNA. EPCR painting normalised PC activation on EPCR-depleted cells indicating that EPCR-GPI is functional active on painted cells. Caveolin-1 lipid rafts were enriched in EPCR after painting due to the GPI-anchor targeting caveolae. Accordingly, EPCR painting supported PAR1 and PAR3 cleavage by APC and augmented PAR1-dependent Akt phosphorylation by APC. Thus, EPCR-GPI painting achieved physiological relevant surface levels on endothelial cells, restored APC binding to EPCR-depleted cells, supported PC activation, and enhanced APC-mediated PAR cleavage and cytoprotective signalling. Therefore, EPCR-GPI provides a novel tool to restore the bioavailability and functionality of EPCR on EPCR- depleted and -deficient cells.
Collapse
Affiliation(s)
| | | | - Laurent O Mosnier
- Laurent O. Mosnier, Department of Molecular and Experimental Medicine (MEM-180), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA, Tel.: +1 858 784 8220, Fax: +1 858 784 2243, E-mail:
| |
Collapse
|
38
|
Engineering activated protein C to maximize therapeutic efficacy. Biochem Soc Trans 2015; 43:691-5. [DOI: 10.1042/bst20140312] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Indexed: 11/17/2022]
Abstract
The anticoagulant-activated protein C (APC) acts not solely as a crucial regulator of thrombus formation following vascular injury, but also as a potent signalling enzyme with important functions in the control of both acute and chronic inflammatory disease. These properties have been exploited to therapeutic effect in diverse animal models of inflammatory disease, wherein recombinant APC administration has proven to effectively limit disease progression. Subsequent clinical trials led to the use of recombinant APC (Xigris) for the treatment of severe sepsis. Although originally deemed successful, Xigris was ultimately withdrawn due to lack of efficacy and an unacceptable bleeding risk. Despite this apparent failure, the problems that beset Xigris usage may be tractable using protein engineering approaches. In this review, we detail the protein engineering approaches that have been utilized to improve the therapeutic characteristics of recombinant APC, from early studies in which the distinct anti-coagulant and signalling activities of APC were separated to reduce bleeding risk, to current attempts to enhance APC cytoprotective signalling output for increased therapeutic efficacy at lower APC dosage. These novel engineered variants represent the next stage in the development of safer, more efficacious APC therapy in disease settings in which APC plays a protective role.
Collapse
|
39
|
Eliwan HO, Watson RWG, Aslam S, Regan I, Philbin B, O'Hare FM, O'Neill A, Preston R, Blanco A, Grant T, Nolan B, Smith O, Molloy EJ. Neonatal brain injury and systemic inflammation: modulation by activated protein C ex vivo. Clin Exp Immunol 2015; 179:477-84. [PMID: 25204207 DOI: 10.1111/cei.12453] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2014] [Indexed: 01/04/2023] Open
Abstract
Infection and inflammation can be antecedents of neonatal encephalopathy (NE) and increase the risk of neurological sequelae. Activated protein C (APC) has anti-coagulant and anti-inflammatory effects and provides neuroprotection in brain and spinal cord injury. We examined neutrophil and monocyte responses to lipopolysaccharide (LPS) in infants with NE compared with healthy adult and neonatal controls, and also studied the effect of APC. Whole blood was incubated with LPS and APC and Toll-like receptor (TLR)-4 (LPS recognition), CD11b expression (activation) and intracellular reactive oxygen intermediate (ROI; function) release from neutrophils and monocytes was examined by flow cytometry serially from days 1 to 7. We found a significant increase in neutrophil ROI in infants with NE on day 3 following LPS compared to neonatal controls and this augmented response was reduced significantly by APC. Neutrophil and monocyte CD11b expression was increased significantly on day 1 in infants with NE compared to neonatal controls. LPS-induced neutrophil TLR-4 expression was increased significantly in infants with NE on days 3 and 7 and was reduced by APC. LPS-induced monocyte TLR-4 was increased significantly in infants with NE on day 7. Neutrophil and monocyte activation and production of ROIs may mediate tissue damage in infants with NE. APC modified LPS responses in infants with NE. APC may reduce the inflammatory responses in NE and may ameliorate multi-organ dysfunction. Further study of the immunomodulatory effects of protein C may be warranted using mutant forms with decreased bleeding potential.
Collapse
Affiliation(s)
- H O Eliwan
- Paediatrics, National Maternity Hospital, Dublin, Ireland; UCD School of Medicine and Medical Science and Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; National Children Research Centre, Dublin, Ireland; Paediatrics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Andreou AP, Efthymiou M, Yu Y, Watts HR, Noormohamed FH, Ma D, Lane DA, Crawley JTB. Protective effects of non-anticoagulant activated protein C variant (D36A/L38D/A39V) in a murine model of ischaemic stroke. PLoS One 2015; 10:e0122410. [PMID: 25830552 PMCID: PMC4382112 DOI: 10.1371/journal.pone.0122410] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/20/2015] [Indexed: 11/30/2022] Open
Abstract
Ischaemic stroke is caused by occlusive thrombi in the cerebral vasculature. Although tissue-plasminogen activator (tPA) can be administered as thrombolytic therapy, it has major limitations, which include disruption of the blood-brain barrier and an increased risk of bleeding. Treatments that prevent or limit such deleterious effects could be of major clinical importance. Activated protein C (APC) is a natural anticoagulant that regulates thrombin generation, but also confers endothelial cytoprotective effects and improved endothelial barrier function mediated through its cell signalling properties. In murine models of stroke, although APC can limit the deleterious effects of tPA due to its cell signalling function, its anticoagulant actions can further elevate the risk of bleeding. Thus, APC variants such as APC(5A), APC(Ca-ins) and APC(36-39) with reduced anticoagulant, but normal signalling function may have therapeutic benefit. Human and murine protein C (5A), (Ca-ins) and (36-39) variants were expressed and characterised. All protein C variants were secreted normally, but 5-20% of the protein C (Ca-ins) variants were secreted as disulphide-linked dimers. Thrombin generation assays suggested reductions in anticoagulant function of 50- to 57-fold for APC(36-39), 22- to 27-fold for APC(Ca-ins) and 14- to 17-fold for APC(5A). Interestingly, whereas human wt APC, APC(36-39) and APC(Ca-ins) were inhibited similarly by protein C inhibitor (t½ - 33 to 39 mins), APC(5A) was inactivated ~9-fold faster (t½ - 4 mins). Using the murine middle cerebral artery occlusion ischaemia/repurfusion injury model, in combination with tPA, APC(36-39), which cannot be enhanced by its cofactor protein S, significantly improved neurological scores, reduced cerebral infarct area by ~50% and reduced oedema ratio. APC(36-39) also significantly reduced bleeding in the brain induced by administration of tPA, whereas wt APC did not. If our data can be extrapolated to clinical settings, then APC(36-39) could represent a feasible adjunctive therapy for ischaemic stroke.
Collapse
Affiliation(s)
- Anna P. Andreou
- Centre for Haematology, Imperial College London, London, United Kingdom
- Section of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London, United Kingdom
- * E-mail:
| | - Maria Efthymiou
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - Yao Yu
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - Helena R. Watts
- Section of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London, United Kingdom
| | - Faruq H. Noormohamed
- Section of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London, United Kingdom
| | - Daqing Ma
- Section of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London, United Kingdom
| | - David A. Lane
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - James TB Crawley
- Centre for Haematology, Imperial College London, London, United Kingdom
| |
Collapse
|
41
|
Abstract
The homeostatic blood protease, activated protein C (APC), can function as (1) an antithrombotic on the basis of inactivation of clotting factors Va and VIIIa; (2) a cytoprotective on the basis of endothelial barrier stabilization and anti-inflammatory and antiapoptotic actions; and (3) a regenerative on the basis of stimulation of neurogenesis, angiogenesis, and wound healing. Pharmacologic therapies using recombinant human and murine APCs indicate that APC provides effective acute or chronic therapies for a strikingly diverse range of preclinical injury models. APC reduces the damage caused by the following: ischemia/reperfusion in brain, heart, and kidney; pulmonary, kidney, and gastrointestinal inflammation; sepsis; Ebola virus; diabetes; and total lethal body radiation. For these beneficial effects, APC alters cell signaling networks and gene expression profiles by activating protease-activated receptors 1 and 3. APC's activation of these G protein-coupled receptors differs completely from thrombin's activation mechanism due to biased signaling via either G proteins or β-arrestin-2. To reduce APC-associated bleeding risk, APC variants were engineered to lack >90% anticoagulant activity but retain normal cell signaling. Such a neuroprotective variant, 3K3A-APC (Lys191-193Ala), has advanced to clinical trials for ischemic stroke. A rich data set of preclinical knowledge provides a solid foundation for potential translation of APC variants to future novel therapies.
Collapse
|
42
|
Jin SJ, Liu Y, Deng SH, Liao LH, Lin TL, Ning Q, Luo XP. Neuroprotective effects of activated protein C on intrauterine inflammation-induced neonatal white matter injury are associated with the downregulation of fibrinogen-like protein 2/fibroleukin prothrombinase and the inhibition of pro-inflammatory cytokine expression. Int J Mol Med 2015; 35:1199-212. [PMID: 25777531 PMCID: PMC4380123 DOI: 10.3892/ijmm.2015.2136] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 03/10/2015] [Indexed: 01/31/2023] Open
Abstract
Maternal intrauterine inflammation or infection is an important risk factor for neonatal cerebral white matter injury (WMI) and future neurological deficits. Activated protein C (APC), a natural anticoagulant, has been shown to exhibit anti-inflammatory, anti-apoptotic, profibrinolytic and cytoprotective activities. Recent studies have demonstrated that the novel prothrombinase, fibrinogen-like protein 2 (fgl2), contributes to the pathogenesis of a number of inflammatory diseases through the generation of fibrin. Thus, we hypothesized that APC may regulate coagulant and inflammatory processes and improve brain injury in an experimental rat model of intrauterine inflammation-induced WMI. The animal model was established by the administration of an intraperitoneal injection of lipopolysaccharide (LPS) to pregnant Sprague-Dawley rats on embryonic day (E)17 and E18. APC was administered intraperitoneally 30 min after the second LPS injection. The expression of fgl2 and the pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α), interleukin (IL)-6 and IL-1β expression in the placentas and fetal brains was determined on E19. Nerve cell death, the brain water content and protease-activated receptor 1 (PAR1) and nuclear factor κB (NF-κB) p65 expression was detected in the fetal brains. WMI in the neonatal rat brains was evaluated by hematoxylin and eosin (H&E) staining and immunohistochemistry for myelin basic protein (MBP). The results revealed that APC markedly reduced the LPS-induced increase in fgl2 expression and fibrin deposition, as well as the production of the pro-inflammatory cytokines, TNF-α, IL-6 and IL-1β, in the placentas and fetal brains. In addition, APC attenuated cerebral apoptosis and brain edema, downregulated PAR1 and NF-κB p65 expression in the fetal brains, and improved hypomyelination and structural disturbances in the periventricular area of the neonatal rat brains. Our observations provide evidence that APC attenuates fetal neuroinflammation and the associated secondary WMI in the developing brain by inhibiting the expression of fgl2 and pro-inflammatory mediators, suggesting that APC may be a potential therapeutic approach for intrauterine inflammation-induced neonatal brain injury.
Collapse
Affiliation(s)
- Sheng-Juan Jin
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yan Liu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Shi-Hua Deng
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Li-Hong Liao
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Tu-Lian Lin
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qin Ning
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiao-Ping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
43
|
Wood DC, Pelc LA, Pozzi N, Wallisch M, Verbout NG, Tucker EI, Gruber A, Di Cera E. WEDGE: an anticoagulant thrombin mutant produced by autoactivation. J Thromb Haemost 2015; 13:111-4. [PMID: 25369995 PMCID: PMC4368433 DOI: 10.1111/jth.12774] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND The production of therapeutically relevant proteases typically involves activation of a zymogen precursor by external enzymes, which may raise regulatory issues about availability and purity. Recent studies of thrombin precursors have shown how to engineer constructs that spontaneously convert to the mature protease by autoactivation, without the need for external enzymes. OBJECTIVES Autoactivation is an innovative strategy that promises to simplify the production of proteases of therapeutic relevance, but has not been tested in practical applications. The aim of this study was to provide a direct test of this strategy. METHODS An autoactivating version of the thrombin mutant W215A/E217A (WE), which is currently in preclinical development as an anticoagulant, was engineered. RESULTS AND CONCLUSIONS The autoactivating version of WE can be produced in large quantities, like WE made in BHK cells or Escherichia coli, and retains all significant functional properties in vitro and in vivo. The results serve as proof of principle that autoactivation is an innovative and effective strategy for the production of trypsin-like proteases of therapeutic relevance.
Collapse
Affiliation(s)
- D C Wood
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Mosnier LO, Zlokovic BV, Griffin JH. Cytoprotective-selective activated protein C therapy for ischaemic stroke. Thromb Haemost 2014; 112:883-92. [PMID: 25230930 DOI: 10.1160/th14-05-0448] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 08/05/2014] [Indexed: 12/14/2022]
Abstract
Despite years of research and efforts to translate stroke research to clinical therapy, ischaemic stroke remains a major cause of death, disability, and diminished quality of life. Primary and secondary preventive measures combined with improved quality of care have made significant progress. However, no novel drug for ischaemic stroke therapy has been approved in the past decade. Numerous studies have shown beneficial effects of activated protein C (APC) in rodent stroke models. In addition to its natural anticoagulant functions, APC conveys multiple direct cytoprotective effects on many different cell types that involve multiple receptors including protease activated receptor (PAR) 1, PAR3, and the endothelial protein C receptor (EPCR). Application of molecular engineered APC variants with altered selectivity profiles to rodent stroke models demonstrated that the beneficial effects of APC primarily require its cytoprotective activities but not its anticoagulant activities. Extensive basic, preclinical, and clinical research provided a compelling rationale based on strong evidence for translation of APC therapy that has led to the clinical development of the cytoprotective-selective APC variant, 3K3A-APC, for ischaemic stroke. Recent identification of non-canonical PAR1 and PAR3 activation by APC that give rise to novel tethered-ligands capable of inducing biased cytoprotective signalling as opposed to the canonical signalling provides a mechanistic explanation for how APC-mediated PAR activation can selectively induce cytoprotective signalling pathways. Collectively, these paradigm-shifting discoveries provide detailed insights into the receptor targets and the molecular mechanisms for neuroprotection by cytoprotective-selective 3K3A-APC, which is currently a biologic drug in clinical trials for ischaemic stroke.
Collapse
Affiliation(s)
- Laurent O Mosnier
- Laurent O. Mosnier, PhD, Department of Molecular and Experimental Medicine (MEM-180), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, USA, Tel.: +1 858 784 2227, Fax: +1 858 784 2243, E-mail:
| | | | | |
Collapse
|
45
|
Lyden P, Levy H, Weymer S, Pryor K, Kramer W, Griffin JH, Davis TP, Zlokovic B. Phase 1 safety, tolerability and pharmacokinetics of 3K3A-APC in healthy adult volunteers. Curr Pharm Des 2014; 19:7479-85. [PMID: 24372304 DOI: 10.2174/1381612819666131230131454] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 11/19/2013] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND PURPOSE Activated Protein C (APC) stimulates multiple cytoprotective pathways via the protease activated receptor-1 (PAR-1) and promotes anticoagulation. 3K3A-APC was designed for preserved activity at PAR-1 with reduced anticoagulation. This Phase 1 trial characterized pharmacokinetics and anticoagulation effects of 3K3A-APC. METHODS Subjects (n=64) were randomly assigned to receive 3K3A-APC (n=4) at 6, 30, 90, 180, 360, 540 or 720 µg/kg or placebo (n=6) and were observed for 24 hr. After safety review additional subjects received drug every 12 hr for 5 doses (n=6 per group) at 90, 180, 360, or 540 µg/kg or placebo (n=8) and were observed for 24 hr. RESULTS All subjects returned for safety assessments at 72 hours and 15 days. We found few adverse events in all groups. Systolic blood pressure increased in both active and placebo groups. Moderately severe headache, nausea and vomiting were reported in one of two subjects treated with 720 µg/kg so 540 µg/kg was considered the highest tolerated dose. Mean plasma concentrations increased in proportion to dose. Clearance ranged from 11,693 ± 807 to 18,701 ± 4,797 mL/hr, volume of distribution ranged from 4,873±828 to 6,971 ± 1,169 mL, and elimination half-life ranged from 0.211 ± 0.097 to 0.294 ± 0.054 hours. Elevations in aPTT were minimal. CONCLUSIONS 3K3A-APC was well tolerated at multiple doses as high as 540 µg/kg. These results should be confirmed in stroke patients with relevant co-morbidities. Clinical Trial Registration-URL: http://www.clinicaltrials.gov. Unique identifier: NCT01660230.
Collapse
|
46
|
von Drygalski A, Bhat V, Gale AJ, Burnier L, Cramer TJ, Griffin JH, Mosnier LO. An engineered factor Va prevents bleeding induced by anticoagulant wt activated protein C. PLoS One 2014; 9:e104304. [PMID: 25127130 PMCID: PMC4134195 DOI: 10.1371/journal.pone.0104304] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 07/07/2014] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE An increased risk of bleeding is observed in patients receiving activated protein C (APC), which may be a limiting factor for the application of novel APC therapies. Since APC's therapeutic effects often require its cytoprotective activities on cells but not APC's anticoagulant activities, an agent that specifically antagonizes APC's anticoagulant effects but not its cytoprotective effects could provide an effective means to control concerns for risk of bleeding. We hypothesized that superFVa, an engineered activated FVa-variant that restores hemostasis in hemophilia could reduce APC-induced bleeding. APPROACH AND RESULTS SuperFVa was engineered with mutations of the APC cleavage sites (Arg506/306/679Gln) and a disulfide bond (Cys609-Cys1691) between the A2 and A3 domains, which augment its biological activity and cause high resistance to APC. SuperFVa normalized APC-prolonged clotting times and restored APC-suppressed thrombin generation in human and murine plasma at concentrations where wild-type (wt) FVa did not show effects. Following intravenous injection of APC into BALB/c mice, addition to whole blood ex vivo of superFVa but not wt-FVa significantly normalized whole blood clotting. Blood loss following tail clip or liver laceration was significantly reduced when superFVa was administered intravenously to BALB/c mice prior to intravenous APC-treatment. Furthermore, superFVa abolished mortality (∼50%) associated with excessive bleeding following liver laceration in mice treated with APC. CONCLUSIONS Our results provide proof of concept that superFVa is effective in preventing APC-induced bleeding and may provide therapeutic benefits as a prohemostatic agent in various situations where bleeding is a serious risk.
Collapse
Affiliation(s)
- Annette von Drygalski
- The Scripps Research Institute, Dept of Molecular and Experimental Medicine, La Jolla, California, United States of America
- University of California San Diego, Dept of Medicine, Div of Hematology/Oncology, San Diego, California, United States of America
| | - Vikas Bhat
- The Scripps Research Institute, Dept of Molecular and Experimental Medicine, La Jolla, California, United States of America
- University of California San Diego, Dept of Medicine, Div of Hematology/Oncology, San Diego, California, United States of America
| | - Andrew J. Gale
- The Scripps Research Institute, Dept of Molecular and Experimental Medicine, La Jolla, California, United States of America
| | - Laurent Burnier
- The Scripps Research Institute, Dept of Molecular and Experimental Medicine, La Jolla, California, United States of America
| | - Thomas J. Cramer
- University of California San Diego, Dept of Medicine, Div of Hematology/Oncology, San Diego, California, United States of America
| | - John H. Griffin
- The Scripps Research Institute, Dept of Molecular and Experimental Medicine, La Jolla, California, United States of America
- University of California San Diego, Dept of Medicine, Div of Hematology/Oncology, San Diego, California, United States of America
| | - Laurent O. Mosnier
- The Scripps Research Institute, Dept of Molecular and Experimental Medicine, La Jolla, California, United States of America
| |
Collapse
|
47
|
Bock F, Shahzad K, Vergnolle N, Isermann B. Activated protein C based therapeutic strategies in chronic diseases. Thromb Haemost 2014; 111:610-7. [PMID: 24652581 DOI: 10.1160/th13-11-0967] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 03/07/2014] [Indexed: 01/03/2023]
Abstract
Activated protein C (aPC) is a natural anticoagulant and a potent anti-inflammatory and cytoprotective agent. At the expense of increased bleeding risk aPC has been used - with some success - in sepsis. The design of cytoprotective-selective aPC variants circumvents this limitation of increased bleeding, reviving the interest in aPC as a therapeutic agent. Emerging studies suggest that aPC`s beneficial effects are not restricted to acute illness, but likewise relevant in chronic diseases, such as diabetic nephropathy, neurodegeneration or wound healing. Epigenetic regulation of gene expression, reduction of oxidative stress, and regulation of ROS-dependent transcription factors are potential mechanisms of sustained cytoprotective effects of aPC in chronic diseases. Given the available data it seems questionable whether a unifying mechanism of aPC dependent cytoprotection in acute and chronic diseases exists. In addition, the signalling pathways employed by aPC are tissue and cell specific. The mechanistic insights gained from studies exploring aPC`s effects in various diseases may hence lay ground for tissue and disease specific therapeutic approaches. This review outlines recent investigations into the mechanisms and consequences of long-term modulation of aPC-signalling in models of chronic diseases.
Collapse
Affiliation(s)
| | | | | | - Berend Isermann
- Berend Isermann, MD, Otto-von-Guericke-University Magdeburg, Institute of Clinical Pathology and Pathobiochemistry, Leipziger Str. 44, D-39120 Magdeburg, Germany, Tel.: +49 391 67 13900, Fax: +49 391 67 13902, E-mail: ;
| |
Collapse
|
48
|
Blood-spinal cord barrier disruption contributes to early motor-neuron degeneration in ALS-model mice. Proc Natl Acad Sci U S A 2014; 111:E1035-42. [PMID: 24591593 DOI: 10.1073/pnas.1401595111] [Citation(s) in RCA: 188] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Humans with ALS and transgenic rodents expressing ALS-associated superoxide dismutase (SOD1) mutations develop spontaneous blood-spinal cord barrier (BSCB) breakdown, causing microvascular spinal-cord lesions. The role of BSCB breakdown in ALS disease pathogenesis in humans and mice remains, however, unclear, although chronic blood-brain barrier opening has been shown to facilitate accumulation of toxic blood-derived products in the central nervous system, resulting in secondary neurodegenerative changes. By repairing the BSCB and/or removing the BSCB-derived injurious stimuli, we now identify that accumulation of blood-derived neurotoxic hemoglobin and iron in the spinal cord leads to early motor-neuron degeneration in SOD1(G93A) mice at least in part through iron-dependent oxidant stress. Using spontaneous or warfarin-accelerated microvascular lesions, motor-neuron dysfunction and injury were found to be proportional to the degree of BSCB disruption at early disease stages in SOD1(G93A) mice. Early treatment with an activated protein C analog restored BSCB integrity that developed from spontaneous or warfarin-accelerated microvascular lesions in SOD1(G93A) mice and eliminated neurotoxic hemoglobin and iron deposits. Restoration of BSCB integrity delayed onset of motor-neuron impairment and degeneration. Early chelation of blood-derived iron and antioxidant treatment mitigated early motor-neuronal injury. Our data suggest that BSCB breakdown contributes to early motor-neuron degeneration in ALS mice and that restoring BSCB integrity during an early disease phase retards the disease process.
Collapse
|
49
|
von Drygalski A, Cramer TJ, Bhat V, Griffin JH, Gale AJ, Mosnier LO. Improved hemostasis in hemophilia mice by means of an engineered factor Va mutant. J Thromb Haemost 2014; 12:363-72. [PMID: 24818532 PMCID: PMC4161283 DOI: 10.1111/jth.12489] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 12/02/2013] [Indexed: 11/30/2022]
Abstract
BACKGROUND Factor (F)VIIa-based bypassing not always provides sufficient hemostasis in hemophilia. OBJECTIVES To investigate the potential of engineered activated factor V (FVa) variants as bypassing agents in hemophilia A. METHODS Activity of FVa variants was studied in vitro using prothrombinase assays with purified components and in FV- and FVIII-deficient plasma using clotting and thrombin generation assays. In vivo bleed reduction after the tail clip was studied in hemophilia A mice. RESULTS AND CONCLUSIONS FVa mutations included a disulfide bond connecting the A2 and A3 domains and ones that rendered FVa resistant to inactivation by activated protein C (APC). '(super) FVa,' a combination of the A2-A3 disulfide (A2-SS-A3) to stabilize FVa and of APC-cleavage site mutations (Arg506/306/679Gln), had enhanced specific activity and complete APC resistance compared with wild-type FVa, FVL eiden (Arg506Gln), or FVaL eiden (A2-SS-A3). Furthermore, (super) FVa potently increased thrombin generation in vitro in FVIII-deficient plasma. In vivo, (super) FVa reduced bleeding in FVIII-deficient mice more effectively than wild-type FVa. Low-dose (super) FVa, but not wild-type FVa, decreased early blood loss during the first 10 min by more than two-fold compared with saline and provided bleed protection for the majority of mice, similar to treatments with FVIII. During the second 10 min after tail cut, (super) FVa at high dose, but not wild-type FVa, effectively reduced bleeding. These findings suggest that (super) FVa enhances not only clot formation but also clot stabilization. Thus, (super) FVa efficiently improved hemostasis in hemophilia in vitro and in vivo and may have potential therapeutic benefits as a novel bypassing agent in hemophilia.
Collapse
Affiliation(s)
- A von Drygalski
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA; Division of Hematology/Oncology, Department of Medicine, University California San Diego, San Diego, CA, USA
| | | | | | | | | | | |
Collapse
|
50
|
Xue M, Jackson CJ. Activated protein C and its potential applications in prevention of islet β-cell damage and diabetes. VITAMINS AND HORMONES 2014; 95:323-63. [PMID: 24559924 DOI: 10.1016/b978-0-12-800174-5.00013-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Activated protein C (APC) is derived from its precursor, protein C (PC). Originally thought to be synthesized exclusively by the liver, recent reports have shown that PC is also produced by many other cells including pancreatic islet β cells. APC functions as a physiological anticoagulant with anti-inflammatory, anti-apoptotic, and barrier-stabilizing properties. APC exerts its protective effects via an intriguing mechanism requiring combinations of endothelial PC receptor, protease-activated receptors, epidermal growth factor receptor, Tie2 or CD11b, depending on cell types. Diabetes is a chronic condition resulted from the body's inability to produce and/or properly use insulin. The prevalence of diabetes has risen dramatically and has become one of the major causes of premature mortality and morbidity worldwide. Diabetes prevention is an ideal approach to reduce this burden. Type 1 and type 2 diabetes are the major forms of diabetes mellitus, and both are characterized by an autoimmune response, intraislet inflammation, β-cell apoptosis, and progressive β-cell loss. Protecting β-cell from damage is critical in both prevention and treatment of diabetes. Recent in vitro and animal studies show that APC's strong anti-inflammatory and anti-apoptotic properties are beneficial in preventing β-cell destruction and diabetes in the NOD mouse model of type 1 diabetes. Future preventive and therapeutic uses of APC in diabetes look very promising.
Collapse
Affiliation(s)
- Meilang Xue
- Sutton Arthritis Research Laboratories, Kolling Institute of Medical Research, The University of Sydney at Royal North Shore Hospital, St Leonards, New South Wales, Australia.
| | - Christopher J Jackson
- Sutton Arthritis Research Laboratories, Kolling Institute of Medical Research, The University of Sydney at Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|