90 results on '"Belcher JD"'
Search Results
2. Mast cell extracellular trap formation underlies vascular and neural injury and hyperalgesia in sickle cell disease.
- Author
-
Argueta DA, Tran H, Goel Y, Nguyen A, Nguyen J, Kiven SB, Chen C, Abdulla F, Vercellotti GM, Belcher JD, and Gupta K
- Subjects
- Animals, Mice, Humans, Male, Disease Models, Animal, Inflammation metabolism, Mice, Inbred C57BL, Neutrophils metabolism, Anemia, Sickle Cell complications, Anemia, Sickle Cell metabolism, Hyperalgesia metabolism, Hyperalgesia etiology, Extracellular Traps metabolism, Mast Cells metabolism, Protein-Arginine Deiminase Type 4 metabolism
- Abstract
Sickle cell disease (SCD) is the most common inherited monogenetic disorder. Chronic and acute pain are hallmark features of SCD involving neural and vascular injury and inflammation. Mast cells reside in the vicinity of nerve fibers and vasculature, but how they influence these structures remains unknown. We therefore examined the mechanism of mast cell activation in a sickle microenvironment replete with cell-free heme and inflammation. Mast cells exposed to this environment showed an explosion of nuclear contents with the release of citrullinated histones, suggestive of mast cell extracellular trap (MCET) release. MCETs interacted directly with the vasculature and nerve fibers, a cause of vascular and neural injury in sickle cell mice. MCET formation was dependent upon peptidylarginine deiminase 4 (PAD4). Inhibition of PAD4 ameliorated vasoocclusion, chronic and acute hyperalgesia, and inflammation in sickle mice. PAD4 activation may also underlie neutrophil trap formation in SCD, thus providing a novel target to treat the sequelae of vascular and neural injury in SCD., (© 2024 Argueta et al.)
- Published
- 2024
- Full Text
- View/download PDF
3. Cold exposure induces vaso-occlusion and pain in sickle mice that depend on complement activation.
- Author
-
Ivy ZK, Belcher JD, Khasabova IA, Chen C, Juliette JP, Abdulla F, Ruan C, Allen K, Nguyen J, Rogness VM, Beckman JD, Khasabov SG, Gupta K, Taylor RP, Simone DA, and Vercellotti GM
- Subjects
- Mice, Humans, Animals, Hyperalgesia etiology, Hyperalgesia metabolism, Mice, Transgenic, Pain, Complement Activation, Anemia, Sickle Cell complications, Anemia, Sickle Cell genetics, Anemia, Sickle Cell metabolism, Sickle Cell Trait complications
- Abstract
Vaso-occlusive pain episodes (VOE) cause severe pain in patients with sickle cell disease (SCD). Vaso-occlusive events promote ischemia/reperfusion pathobiology that activates complement. We hypothesized that complement activation is linked to VOE. We used cold to induce VOE in the Townes sickle homozygous for hemoglobin S (HbSS) mouse model and complement inhibitors to determine whether anaphylatoxin C5a mediates VOE. We used a dorsal skinfold chamber to measure microvascular stasis (vaso-occlusion) and von Frey filaments applied to the plantar surface of the hind paw to assess mechanical hyperalgesia in HbSS and control Townes mice homozygous for hemoglobin A (HbAA) mice after cold exposure at 10°C/50°F for 1 hour. Cold exposure induced more vaso-occlusion in nonhyperalgesic HbSS mice (33%) than in HbAA mice (11%) or HbSS mice left at room temperature (1%). Cold exposure also produced mechanical hyperalgesia as measured by paw withdrawal threshold in HbSS mice compared with that in HbAA mice or HbSS mice left at room temperature. Vaso-occlusion and hyperalgesia were associated with an increase in complement activation fragments Bb and C5a in plasma of HbSS mice after cold exposure. This was accompanied by an increase in proinflammatory NF-κB activation and VCAM-1 and ICAM-1 expression in the liver. Pretreatment of nonhyperalgesic HbSS mice before cold exposure with anti-C5 or anti-C5aR monoclonal antibodies (mAbs) decreased vaso-occlusion, mechanical hyperalgesia, complement activation, and liver inflammatory markers compared with pretreatment with control mAb. Anti-C5 or -C5aR mAb infusion also abrogated mechanical hyperalgesia in HbSS mice with ongoing hyperalgesia at baseline. These findings suggest that C5a promotes vaso-occlusion, pain, and inflammation during VOE and may play a role in chronic pain., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
4. Sickle red blood cell-derived extracellular vesicles activate endothelial cells and enhance sickle red cell adhesion mediated by von Willebrand factor.
- Author
-
An R, Man Y, Cheng K, Zhang T, Chen C, Wang F, Abdulla F, Kucukal E, Wulftange WJ, Goreke U, Bode A, Nayak LV, Vercellotti GM, Belcher JD, Little JA, and Gurkan UA
- Subjects
- Humans, Animals, Mice, von Willebrand Factor metabolism, Cell Adhesion, Erythrocytes metabolism, Endothelial Cells pathology, Anemia, Sickle Cell
- Abstract
Endothelial activation and sickle red blood cell (RBC) adhesion are central to the pathogenesis of sickle cell disease (SCD). Quantitatively, RBC-derived extracellular vesicles (REVs) are more abundant from SS RBCs compared with healthy RBCs (AA RBCs). Sickle RBC-derived REVs (SS REVs) are known to promote endothelial cell (EC) activation through cell signalling and transcriptional regulation at longer terms. However, the SS REV-mediated short-term non-transcriptional response of EC is unclear. Here, we examined the impact of SS REVs on acute microvascular EC activation and RBC adhesion at 2 h. Compared with AA REVs, SS REVs promoted human pulmonary microvascular ECs (HPMEC) activation indicated by increased von Willebrand factor (VWF) expression. Under microfluidic conditions, we found abnormal SS RBC adhesion to HPMECs exposed to SS REVs. This enhanced SS RBC adhesion was reduced by haeme binding protein haemopexin or VWF cleaving protease ADAMTS13 to a level similar to HPMECs treated with AA REVs. Consistent with these observations, haemin- or SS REV-induced microvascular stasis in SS mice with implanted dorsal skin-fold chambers that was inhibited by ADAMTS13. The adhesion induced by SS REVs was variable and was higher with SS RBCs from patients with increased markers of haemolysis (lactate dehydrogenase and reticulocyte count) or a concomitant clinical diagnosis of deep vein thrombosis. Our results emphasise the critical contribution made by REVs to the pathophysiology of SCD by triggering acute microvascular EC activation and abnormal RBC adhesion. These findings may help to better understand acute pathophysiological mechanism of SCD and thereby the development of new treatment strategies using VWF as a potential target., (© 2023 The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
5. The BACH1 inhibitor ASP8731 inhibits inflammation and vaso-occlusion and induces fetal hemoglobin in sickle cell disease.
- Author
-
Belcher JD, Nataraja S, Abdulla F, Zhang P, Chen C, Nguyen J, Ruan C, Singh M, Demes S, Olson L, Stickens D, Stanwix J, Clarke E, Huang Y, Biddle M, and Vercellotti GM
- Abstract
In sickle cell disease (SCD), heme released during intravascular hemolysis promotes oxidative stress, inflammation, and vaso-occlusion. Conversely, free heme can also activate expression of antioxidant and globin genes. Heme binds to the transcription factor BACH1, which represses NRF2-mediated gene transcription. ASP8731, is a selective small molecule inhibitor of BACH1. We investigated the ability of ASP8731 to modulate pathways involved in SCD pathophysiology. In HepG2 liver cells, ASP8731 increased HMOX1 and FTH1 mRNA. In pulmonary endothelial cells, ASP8731 decreased VCAM1 mRNA in response to TNF-α and blocked a decrease in glutathione in response to hemin. Townes-SS mice were gavaged once per day for 4 weeks with ASP8731, hydroxyurea (HU) or vehicle. Both ASP8731 and HU inhibited heme-mediated microvascular stasis and in combination, ASP8731 significantly reduced microvascular stasis compared to HU alone. In Townes-SS mice, ASP8731 and HU markedly increased heme oxygenase-1 and decreased hepatic ICAM-1, NF-kB phospho-p65 protein expression in the liver, and white blood cell counts. In addition, ASP8731 increased gamma-globin expression and HbF+ cells (F-cells) as compared to vehicle-treated mice. In human erythroid differentiated CD34+ cells, ASP8731 increased HGB mRNA and increased the percentage of F-cells 2-fold in manner similar to HU. ASP8731 and HU when given together induced more HbF+ cells compared to either drug alone. In CD34+ cells from one donor that was non-responsive to HU, ASP8731 induced HbF+ cells ~2-fold. ASP8731 and HU also increased HBG and HBA , but not HBB mRNA in erythroid differentiated CD34+ cells derived from SCD patients. These data indicate that BACH1 may offer a new therapeutic target to treat SCD., Competing Interests: JB and GV are consultants and receive research funding from Astellas Pharma/Mitobridge. SN, SD, LO, and JS are employees of Astellas Pharma US Inc. MS, DS, and MB were employees of Astellas Pharma US Inc./Mitobridge at the time the study was conducted. EC and YH were employed by company ReachBio. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The authors declare that this study received funding from Astellas-Mitobridge. The funder had the following involvement: study design, data collection, analysis, interpretation of data, and the writing of this article., (Copyright © 2023 Belcher, Nataraja, Abdulla, Zhang, Chen, Nguyen, Ruan, Singh, Demes, Olson, Stickens, Stanwix, Clark, Huang, Biddle and Vercellotti.)
- Published
- 2023
- Full Text
- View/download PDF
6. Factor XII contributes to thrombotic complications and vaso-occlusion in sickle cell disease.
- Author
-
Sparkenbaugh EM, Henderson MW, Miller-Awe M, Abrams C, Ilich A, Trebak F, Ramadas N, Vital S, Bohinc D, Bane KL, Chen C, Patel M, Wallisch M, Renné T, Gruber A, Cooley B, Gailani D, Kasztan M, Vercellotti GM, Belcher JD, Gavins FE, Stavrou EX, Key NS, and Pawlinski R
- Subjects
- Animals, Mice, Inflammation, Stroke, Thrombosis metabolism, Anemia, Sickle Cell complications, Anemia, Sickle Cell metabolism, Factor XII metabolism
- Abstract
A hypercoagulable state, chronic inflammation, and increased risk of venous thrombosis and stroke are prominent features in patients with sickle cell disease (SCD). Coagulation factor XII (FXII) triggers activation of the contact system that is known to be involved in both thrombosis and inflammation, but not in physiological hemostasis. Therefore, we investigated whether FXII contributes to the prothrombotic and inflammatory complications associated with SCD. We found that when compared with healthy controls, patients with SCD exhibit increased circulating biomarkers of FXII activation that are associated with increased activation of the contact pathway. We also found that FXII, but not tissue factor, contributes to enhanced thrombin generation and systemic inflammation observed in sickle cell mice challenged with tumor necrosis factor α. In addition, FXII inhibition significantly reduced experimental venous thrombosis, congestion, and microvascular stasis in a mouse model of SCD. Moreover, inhibition of FXII attenuated brain damage and reduced neutrophil adhesion to the brain vasculature of sickle cell mice after ischemia/reperfusion induced by transient middle cerebral artery occlusion. Finally, we found higher FXII, urokinase plasminogen activator receptor, and αMβ2 integrin expression in neutrophils of patients with SCD compared with healthy controls. Our data indicate that targeting FXII effectively reduces experimental thromboinflammation and vascular complications in a mouse model of SCD, suggesting that FXII inhibition may provide a safe approach for interference with inflammation, thrombotic complications, and vaso-occlusion in patients with SCD.
- Published
- 2023
- Full Text
- View/download PDF
7. MASP-2 and MASP-3 inhibitors block complement activation, inflammation, and microvascular stasis in a murine model of vaso-occlusion in sickle cell disease.
- Author
-
Belcher JD, Nguyen J, Chen C, Abdulla F, Conglin R, Ivy ZK, Cummings J, Dudler T, and Vercellotti GM
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Complement Activation, Disease Models, Animal, E-Selectin, Heme, Hemoglobins, Hemolysis, Hypoxia, Inflammation, Intercellular Adhesion Molecule-1, Mannose-Binding Lectins, Mannose-Binding Protein-Associated Serine Proteases metabolism, Mice, NF-kappa B, Vascular Cell Adhesion Molecule-1 metabolism, Anemia, Sickle Cell complications, Volatile Organic Compounds
- Abstract
Patients with sickle cell disease (SCD) have ongoing hemolysis that promotes endothelial injury, complement activation, inflammation, vaso-occlusion, ischemia-reperfusion pathophysiology, and pain. Complement activation markers are increased in SCD in steady-state and further increased during vaso-occlusive crisis (VOC). However, the mechanisms driving complement activation in SCD have not been completely elucidated. Ischemia-reperfusion and heme released from hemoglobin during hemolysis, events that characterize SCD pathophysiology, can activate the lectin pathway (LP) and alternative pathway (AP), respectively. Here we evaluated the role of LP and AP in Townes sickle (SS) mice using inhibitory monoclonal antibodies (mAb) to mannose binding lectin (MBL)-associated serine protease (MASP)-2 or MASP-3, respectively. Townes SS mice were pretreated with MASP-2 mAb, MASP-3 mAb, isotype control mAb, or PBS before they were challenged with hypoxia-reoxygenation or hemoglobin. Pretreatment of SS mice with MASP-2 or MASP-3 mAb, markedly reduced Bb fragments, C4d and C5a in plasma and complement deposition in the liver, kidneys, and lungs collected 4 hours after challenge compared to control mAb-treated mice. Consistent with complement inhibition, hepatic inflammation markers NF-ĸB phospho-p65, VCAM-1, ICAM-1, and E-selectin were significantly reduced in SS mice pretreated with MASP-2 or MASP-3 mAb. Importantly, MASP-2 or MASP-3 mAb pretreatment significantly inhibited microvascular stasis (vaso-occlusion) induced by hypoxia-reoxygenation or hemoglobin. These studies suggest that the LP and the AP are both playing a role in promoting inflammation and vaso-occlusion in SCD. Inhibiting complement activation via the LP or the AP might inhibit inflammation and prevent VOC in SCD patients., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
8. A model of painful vaso-occlusive crisis in mice with sickle cell disease.
- Author
-
Khasabova II, Juliette J, Rogness VM, Khasabov SG, Golovko MY, Golovko SA, Kiven S, Gupta K, Belcher JD, Vercellotti GM, Seybold VS, and Simone DA
- Subjects
- Mice, Animals, Pain etiology, Anemia, Sickle Cell complications, Hemoglobinopathies
- Published
- 2022
- Full Text
- View/download PDF
9. Plasma-Derived Hemopexin as a Candidate Therapeutic Agent for Acute Vaso-Occlusion in Sickle Cell Disease: Preclinical Evidence.
- Author
-
Gentinetta T, Belcher JD, Brügger-Verdon V, Adam J, Ruthsatz T, Bain J, Schu D, Ventrici L, Edler M, Lioe H, Patel K, Chen C, Nguyen J, Abdulla F, Zhang P, Wassmer A, Jain M, Mischnik M, Pelzing M, Martin K, Davis R, Didichenko S, Schaub A, Brinkman N, Herzog E, Zürcher A, Vercellotti GM, Kato GJ, and Höbarth G
- Abstract
People living with sickle cell disease (SCD) face intermittent acute pain episodes due to vaso-occlusion primarily treated palliatively with opioids. Hemolysis of sickle erythrocytes promotes release of heme, which activates inflammatory cell adhesion proteins on endothelial cells and circulating cells, promoting vaso-occlusion. In this study, plasma-derived hemopexin inhibited heme-mediated cellular externalization of P-selectin and von Willebrand factor, and expression of IL-8, VCAM-1, and heme oxygenase-1 in cultured endothelial cells in a dose-responsive manner. In the Townes SCD mouse model, intravenous injection of free hemoglobin induced vascular stasis (vaso-occlusion) in nearly 40% of subcutaneous blood vessels visualized in a dorsal skin-fold chamber. Hemopexin administered intravenously prevented or relieved stasis in a dose-dependent manner. Hemopexin showed parallel activity in relieving vascular stasis induced by hypoxia-reoxygenation. Repeated IV administration of hemopexin was well tolerated in rats and non-human primates with no adverse findings that could be attributed to human hemopexin. Hemopexin had a half-life in wild-type mice, rats, and non-human primates of 80-102 h, whereas a reduced half-life of hemopexin in Townes SCD mice was observed due to ongoing hemolysis. These data have led to a Phase 1 clinical trial of hemopexin in adults with SCD, which is currently ongoing.
- Published
- 2022
- Full Text
- View/download PDF
10. Vasculo-toxic and pro-inflammatory action of unbound haemoglobin, haem and iron in transfusion-dependent patients with haemolytic anaemias.
- Author
-
Vinchi F, Sparla R, Passos ST, Sharma R, Vance SZ, Zreid HS, Juaidi H, Manwani D, Yazdanbakhsh K, Nandi V, Silva AMN, Agarvas AR, Fibach E, Belcher JD, Vercellotti GM, Ghoti H, and Muckenthaler MU
- Subjects
- Adolescent, Adult, Anemia, Sickle Cell therapy, Blood Transfusion, Child, Child, Preschool, Endothelium, Vascular metabolism, Female, Humans, Inflammation blood, Intercellular Adhesion Molecule-1 blood, Interleukin-6 blood, Male, Spherocytosis, Hereditary therapy, Tumor Necrosis Factor-alpha blood, Vascular Cell Adhesion Molecule-1 blood, Vascular Endothelial Growth Factor A blood, beta-Thalassemia therapy, Anemia, Sickle Cell blood, Heme metabolism, Hemoglobins metabolism, Iron blood, Spherocytosis, Hereditary blood, beta-Thalassemia blood
- Abstract
Increasing evidence suggests that free haem and iron exert vasculo-toxic and pro-inflammatory effects by activating endothelial and immune cells. In the present retrospective study, we compared serum samples from transfusion-dependent patients with β-thalassaemia major and intermedia, hereditary spherocytosis and sickle cell disease (SCD). Haemolysis, transfusions and ineffective erythropoiesis contribute to haem and iron overload in haemolytic patients. In all cohorts we observed increased systemic haem and iron levels associated with scavenger depletion and toxic 'free' species formation. Endothelial dysfunction, oxidative stress and inflammation markers were significantly increased compared to healthy donors. In multivariable logistic regression analysis, oxidative stress markers remained significantly associated with both haem- and iron-related parameters, while soluble vascular cell adhesion molecule 1 (sVCAM-1), soluble endothelial selectin (sE-selectin) and tumour necrosis factor α (TNFα) showed the strongest association with haem-related parameters and soluble intercellular adhesion molecule 1 (sICAM-1), sVCAM-1, interleukin 6 (IL-6) and vascular endothelial growth factor (VEGF) with iron-related parameters. While hereditary spherocytosis was associated with the highest IL-6 and TNFα levels, β-thalassaemia major showed limited inflammation compared to SCD. The sVCAM1 increase was significantly lower in patients with SCD receiving exchange compared to simple transfusions. The present results support the involvement of free haem/iron species in the pathogenesis of vascular dysfunction and sterile inflammation in haemolytic diseases, irrespective of the underlying haemolytic mechanism, and highlight the potential therapeutic benefit of iron/haem scavenging therapies in these conditions., (© 2021 The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
11. Noncanonical Roles of Caspase-4 and Caspase-5 in Heme-Driven IL-1β Release and Cell Death.
- Author
-
Bolívar BE, Brown-Suedel AN, Rohrman BA, Charendoff CI, Yazdani V, Belcher JD, Vercellotti GM, Flanagan JM, and Bouchier-Hayes L
- Subjects
- Alarmins metabolism, Cell Death, Cells, Cultured, Heme metabolism, Hemolysis, Humans, Interleukin-1beta metabolism, Up-Regulation, Anemia, Sickle Cell metabolism, Caspases metabolism, Caspases, Initiator metabolism, Erythrocytes physiology, Inflammasomes metabolism, Inflammation metabolism, Macrophages immunology
- Abstract
Excessive release of heme from RBCs is a key pathophysiological feature of several disease states, including bacterial sepsis, malaria, and sickle cell disease. This hemolysis results in an increased level of free heme that has been implicated in the inflammatory activation of monocytes, macrophages, and the endothelium. In this study, we show that extracellular heme engages the human inflammatory caspases, caspase-1, caspase-4, and caspase-5, resulting in the release of IL-1β. Heme-induced IL-1β release was further increased in macrophages from patients with sickle cell disease. In human primary macrophages, heme activated caspase-1 in an inflammasome-dependent manner, but heme-induced activation of caspase-4 and caspase-5 was independent of canonical inflammasomes. Furthermore, we show that both caspase-4 and caspase-5 are essential for heme-induced IL-1β release, whereas caspase-4 is the primary contributor to heme-induced cell death. Together, we have identified that extracellular heme is a damage-associated molecular pattern that can engage canonical and noncanonical inflammasome activation as a key mediator of inflammation in macrophages., (Copyright © 2021 by The American Association of Immunologists, Inc.)
- Published
- 2021
- Full Text
- View/download PDF
12. Soluble MD-2 and Heme in Sickle Cell Disease Plasma Promote Pro-Inflammatory Signaling in Endothelial Cells.
- Author
-
Zhang P, Nguyen J, Abdulla F, Nelson AT, Beckman JD, Vercellotti GM, and Belcher JD
- Subjects
- Anemia, Sickle Cell blood, Animals, Hemopexin metabolism, Human Umbilical Vein Endothelial Cells metabolism, Humans, Inflammation, Interleukin-8 metabolism, Lymphocyte Antigen 96 blood, Mice, Toll-Like Receptor 4 metabolism, Anemia, Sickle Cell metabolism, Endothelial Cells metabolism, Heme metabolism, Lymphocyte Antigen 96 metabolism, Signal Transduction
- Abstract
Recent evidence indicates that hemolysis in sickle cell disease (SCD) promotes inflammation via innate immune signaling through toll-like receptor 4 (TLR4). Free heme released by hemolyzed red blood cells can bind to myeloid differentiation factor-2 (MD-2) and activate TLR4 pro-inflammatory signaling on endothelium to promote vaso-occlusion and acute chest syndrome in murine models of SCD. MD-2 is co-expressed with TLR4 on cell membranes, but in inflammatory conditions, soluble MD-2 (sMD-2) is elevated in plasma. sMD-2 levels were significantly increased in human and murine sickle (SS) plasma as compared to normal (AA) plasma. Human umbilical vein endothelial cells (HUVEC) and human lung microvascular endothelial cells incubated with human SS plasma had significant increases in pro-inflammatory IL-8, IL-6, and soluble VCAM-1 secretion compared to endothelial cells incubated with AA plasma. The increase in HUVEC IL-8 secretion was blocked by depletion of sMD-2 from SS plasma and enhanced by the addition of sMD-2 to AA plasma. The TLR4 signaling inhibitor, TAK-242, inhibited HUVEC IL-8 secretion in response to SS plasma by 85%. Heme-agarose pull-down assays and UV/Vis spectroscopy demonstrated that heme binds to sMD-2. Hemopexin, a high affinity heme-binding protein, inhibited HUVEC IL-8 secretion induced by SS plasma or SS and AA plasma supplemented with sMD-2. These data suggest that sMD-2 bound to heme might play an important role in pro-inflammatory signaling by endothelium in SCD., Competing Interests: JDBec receives funding from Bayer not related to work herein. JDBel and GMV receive research funding from CSL Behring and Mitobridge (Astellas). The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Zhang, Nguyen, Abdulla, Nelson, Beckman, Vercellotti and Belcher.)
- Published
- 2021
- Full Text
- View/download PDF
13. Endothelial TLR4 Expression Mediates Vaso-Occlusive Crisis in Sickle Cell Disease.
- Author
-
Beckman JD, Abdullah F, Chen C, Kirchner R, Rivera-Rodriguez D, Kiser ZM, Nguyen A, Zhang P, Nguyen J, Hebbel RP, Belcher JD, and Vercellotti GM
- Subjects
- Animals, Erythrocytes metabolism, Female, Hematopoiesis physiology, Heme metabolism, Hemoglobin, Sickle metabolism, Humans, Inflammation metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Microvessels, NF-kappa B metabolism, Signal Transduction physiology, Transcription Factor RelA metabolism, Anemia, Sickle Cell metabolism, Endothelium metabolism, Hemoglobin A metabolism, Toll-Like Receptor 4 metabolism
- Abstract
Heme, released from red blood cells in sickle cell disease (SCD), interacts with toll-like receptor 4 (TLR4) to activate NF-κB leading to the production of cytokines and adhesion molecules which promote inflammation, pain, and vaso-occlusion. In SCD, TLR4 inhibition has been shown to modulate heme-induced microvascular stasis and lung injury. We sought to delineate the role of endothelial verses hematopoietic TLR4 in SCD by developing a TLR4 null transgenic sickle mouse. We bred a global Tlr4
-/- deficiency state into Townes-AA mice expressing normal human adult hemoglobin A and Townes-SS mice expressing sickle hemoglobin S. SS- Tlr4-/- had similar complete blood counts and serum chemistries as SS- Tlr4+/+ mice. However, SS- Tlr4-/- mice developed significantly less microvascular stasis in dorsal skin fold chambers than SS- Tlr4+/+ mice in response to challenges with heme, lipopolysaccharide (LPS), and hypoxia/reoxygenation (H/R). To define a potential mechanism for decreased microvascular stasis in SS- Tlr4-/- mice, we measured pro-inflammatory NF-κB and adhesion molecules in livers post-heme challenge. Compared to heme-challenged SS- Tlr4+/+ livers, SS- Tlr4-/- livers had lower adhesion molecule and cytokine mRNAs, NF-κB phospho-p65, and adhesion molecule protein expression. Furthermore, lung P-selectin and von Willebrand factor immunostaining was reduced. Next, to establish if endothelial or hematopoietic cell TLR4 signaling is critical to vaso-occlusive physiology, we created chimeric mice by transplanting SS- Tlr4-/- or SS- Tlr4+/+ bone marrow into AA- Tlr4-/- or AA- Tlr4+/+ recipients. Hemin-stimulated microvascular stasis was significantly decreased when the recipient was AA- Tlr4-/- . These data demonstrate that endothelial, but not hematopoietic, TLR4 expression is necessary to initiate vaso-occlusive physiology in SS mice., Competing Interests: JBec receives funding from Bayer not related to work herein. JBel and GV receive research funding from CSL Behring and Mitobridge (Astellas). The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Beckman, Abdullah, Chen, Kirchner, Rivera-Rodriguez, Kiser, Nguyen, Zhang, Nguyen, Hebbel, Belcher and Vercellotti.)- Published
- 2021
- Full Text
- View/download PDF
14. Identification of a Heme Activation Site on the MD-2/TLR4 Complex.
- Author
-
Belcher JD, Zhang P, Nguyen J, Kiser ZM, Nath KA, Hu J, Trent JO, and Vercellotti GM
- Subjects
- Anemia, Sickle Cell metabolism, HEK293 Cells, Humans, Heme metabolism, Lymphocyte Antigen 96 metabolism, Toll-Like Receptor 4 metabolism
- Abstract
Myeloid differentiation factor-2 (MD-2) binds lipopolysaccharide (LPS) and initiates toll-like receptor-4 (TLR4) pro-inflammatory signaling. Heme also activates TLR4 signaling, but it is unknown if heme interacts with MD-2. Therefore, we examined MD-2 for a potential heme activation site. Heme-agarose and biotin-heme/streptavidin-agarose pulled down recombinant MD-2, which was inhibited by excess free heme. UV/visible spectroscopy confirmed MD-2-heme binding. To determine whether MD-2 was required for heme-mediated TLR4 signaling, HEK293 cells were transfected with MD-2, TLR4, CD14, and an NF-κB luciferase reporter, and then stimulated with heme or LPS. Heme or LPS treatment elicited robust reporter activity. Absence of MD-2, TLR4 or CD14 plasmid abolished NF-κB reporter responses to heme or LPS. In silico analysis identified two potential heme docking sites on MD-2 near conserved amino acids W23/S33/Y34 and Y36/C37/I44. Heme-induced NF-κB activity was reduced by 39 and 78% in HEK293 cells transfected with MD-2 mutants W23A and Y34A, respectively, compared to WT-MD-2. NF-κB activation by LPS was not affected by the same mutants. Biotinyl-heme/streptavidin-agarose pulled down 68% less W23A and 80% less W23A/S33A/Y34A mutant MD-2 than WT-MD-2. In contrast, at the Y36/C37/I44 MD-2 site, heme-induced NF-κB activity was significantly increased by mutants Y36A (191% of WT-MD-2) and unchanged by mutants C37A and I44A (95 and 92%, respectively, of WT-MD-2). In conclusion, these data suggest that heme binds and activates TLR4 signaling at amino acids W23 and Y34 on MD-2., (Copyright © 2020 Belcher, Zhang, Nguyen, Kiser, Nath, Hu, Trent and Vercellotti.)
- Published
- 2020
- Full Text
- View/download PDF
15. Thrombin activation of PAR-1 contributes to microvascular stasis in mouse models of sickle cell disease.
- Author
-
Sparkenbaugh EM, Chen C, Brzoska T, Nguyen J, Wang S, Vercellotti GM, Key NS, Sundd P, Belcher JD, and Pawlinski R
- Subjects
- Anemia, Sickle Cell complications, Anemia, Sickle Cell genetics, Anemia, Sickle Cell pathology, Animals, Blood Coagulation Disorders genetics, Blood Coagulation Disorders metabolism, Blood Platelets metabolism, Constriction, Pathologic genetics, Constriction, Pathologic metabolism, Disease Models, Animal, Female, Hemoglobin, Sickle genetics, Humans, Male, Mice, Mice, Transgenic, Microvessels metabolism, Microvessels pathology, Receptor, PAR-1 genetics, Vascular Diseases etiology, Vascular Diseases metabolism, Anemia, Sickle Cell metabolism, Blood Coagulation Disorders etiology, Receptor, PAR-1 metabolism, Thrombin metabolism
- Abstract
Vaso-occlusive crisis (VOC) is the primary cause of morbidity and hospitalization in sickle cell disease (SCD); however, only 4 therapies (hydroxyurea, l-glutamine, crizanlizumab, and voxeletor) are currently approved in SCD. These agents limit the duration, severity, and frequency of crises. Activation of coagulation is a hallmark of SCD. Studies in animal models of SCD have shown that coagulation contributes to the chronic inflammation and end-organ damage associated with the disease; however, it is unknown whether coagulation directly contributes to the microvascular stasis that causes VOC. Herein, we demonstrate that inhibition of tissue factor (TF) and the downstream coagulation proteases factor Xa and thrombin significantly attenuates heme-induced microvascular stasis in mouse models of VOC. Pharmacologic inhibition of the principal thrombin receptor, protease activated receptor-1 (PAR-1), as well as deficiency of PAR-1 in all nonhematopoietic cells, also reduces stasis in sickle mice. PAR-1 deficiency was associated with reduced endothelial von Willebrand factor expression, which has been shown to mediate microvascular stasis. In addition, TF inhibition reduces lung vaso-occlusion in sickle mice mediated by arteriolar neutrophil-platelet microemboli. In sum, these results suggest that prophylactic anticoagulation might attenuate the incidence of VOC., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
16. The multifaceted role of ischemia/reperfusion in sickle cell anemia.
- Author
-
Hebbel RP, Belcher JD, and Vercellotti GM
- Subjects
- Animals, Disease Models, Animal, Inflammation classification, Inflammation genetics, Inflammation metabolism, Inflammation physiopathology, Mice, Mice, Transgenic, Reperfusion Injury classification, Reperfusion Injury genetics, Reperfusion Injury metabolism, Reperfusion Injury physiopathology, Anemia, Sickle Cell genetics, Anemia, Sickle Cell metabolism, Anemia, Sickle Cell physiopathology, Endothelium, Vascular metabolism, Endothelium, Vascular physiopathology
- Abstract
Sickle cell anemia is a unique disease dominated by hemolytic anemia and vaso-occlusive events. The latter trigger a version of ischemia/reperfusion (I/R) pathobiology that is singular in its origin, cyclicity, complexity, instability, perpetuity, and breadth of clinical consequences. Specific clinical features are probably attributable to local I/R injury (e.g., stroke syndromes) or remote organ injury (e.g., acute chest syndrome) or the systematization of inflammation (e.g., multifocal arteriopathy). Indeed, by fashioning an underlying template of endothelial dysfunction and vulnerability, the robust inflammatory systematization no doubt contributes to all sickle pathology. In this Review, we highlight I/R-targeting therapeutics shown to improve microvascular blood flow in sickle transgenic mice undergoing I/R, and we suggest how such insights might be translated into human therapeutic strategies.
- Published
- 2020
- Full Text
- View/download PDF
17. Decreased erythrocyte binding of Siglec-9 increases neutrophil activation in sickle cell disease.
- Author
-
Kiser ZM, Lizcano A, Nguyen J, Becker GL, Belcher JD, Varki AP, and Vercellotti GM
- Subjects
- Cells, Cultured, Humans, Oxidative Stress, Protein Binding, Reactive Oxygen Species metabolism, Respiratory Burst, Anemia, Sickle Cell blood, Antigens, CD metabolism, Erythrocytes metabolism, Neutrophil Activation, Sialic Acid Binding Immunoglobulin-like Lectins metabolism
- Abstract
Oxidative stress and inflammation promote vaso-occlusion in sickle cell disease (SCD). CD33-related Sialic acid-binding immunoglobulin-type lectins (CD33rSiglecs) are cell surface proteins that recognize sialic acids inhibit innate immune cell functions. We have shown that Siglec-9 on human neutrophils interact with erythrocyte sialic acids (prominently glycophorin-A (GYPA) to suppress neutrophil reactive oxygen species (ROS). We hypothesized that altered sickle erythrocyte membrane sialic acid leads to decreased Siglec-9 binding capability, and thus a decreased neutrophil oxidative burst. SS erythrocytes express significantly more sialic acid than AA erythrocytes (p = 0.02). SS erythrocytes displayed significantly less Siglec-9-Fc binding 39% ± 11 (mean ± SEM) compared to AA erythrocytes 78% ± 5 (p = 0.009). Treatment of AA erythrocytes with sialidase to remove sialic acid decreased binding to 3% ± 7.9 (p ≤ 0.001). When freshly isolated neutrophils were incubated with AA erythrocytes, neutrophils achieved 16% ± 6 of the oxidative burst exhibited by a stimulated neutrophil without erythrocytes. In contrast, neutrophils incubated with SS erythrocytes achieved 47% ± 6 of the oxidative burst (AA versus SS, p = 0.03). Stimulated neutrophils incubated with AA erythrocytes showed minimal NET formation while with SS erythrocytes NETs increased. SS erythrocytes are deficient in binding to neutrophil Siglec-9 which may contribute to the increased oxidative stress in SCD., Competing Interests: Declaration of competing interest Drs Belcher and Vercellotti receive research funding to their laboratory from CSL Behring (King of Prussia, PA, USA), Hillhurst (Montrose, CA, USA) and Astellas (Northbrook, IL, USA). Other authors have no conflicts of interest., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
18. Antithrombotic effects of heme-degrading and heme-binding proteins.
- Author
-
Nath KA, Grande JP, Belcher JD, Garovic VD, Croatt AJ, Hillestad ML, Barry MA, Nath MC, Regan RF, and Vercellotti GM
- Subjects
- Animals, Disease Models, Animal, Heme Oxygenase-1 genetics, Heme-Binding Proteins genetics, Hemin pharmacology, Mice, Mice, Knockout, Venous Thrombosis genetics, Heme Oxygenase-1 metabolism, Heme-Binding Proteins metabolism, Up-Regulation, Venous Thrombosis metabolism
- Abstract
In the murine venous thrombosis model induced by ligation of the inferior vena cava (IVCL), genetic deficiency of heme oxygenase-1 (HO-1) increases clot size. This study examined whether induction of HO-1 or administration of its products reduces thrombosis. Venous HO-1 upregulation by gene delivery reduced clot size, as did products of HO activity, biliverdin, and carbon monoxide. Induction of HO-1 by hemin reduced clot formation, clot size, and upregulation of plasminogen activator inhibitor-1 (PAI-1) that occurs in the IVCL model, while leaving urokinase plasminogen activator (uPA) and tissue plasminogen activator (tPA) expression unaltered. The reductive effect of hemin on clot size required HO activity. The IVCL model exhibited relatively high concentrations of heme that peaked just before maximum clot size, then declined as clot size decreased. Administration of hemin decreased heme concentration in the IVCL model. HO-2 mRNA was induced twofold in the IVCL model (vs. 40-fold HO-1 induction), but clot size was not increased in HO-2
-/- mice compared with HO-2+/+ mice. Hemopexin, the major heme-binding protein, was induced in the IVCL model, and clot size was increased in hemopexin-/- mice compared with hemopexin+/+ mice. We conclude that in the IVCL model, the heme-degrading protein HO-1 and HO products inhibit thrombus formation, as does the heme-binding protein, hemopexin. The reductive effects of hemin administration require HO activity and are mediated, in part, by reducing PAI-1 upregulation in the IVCL model. We speculate that HO-1, HO, and hemopexin reduce clot size by restraining the increase in clot concentration of heme (now recognized as a procoagulant) that otherwise occurs. NEW & NOTEWORTHY This study provides conclusive evidence that two proteins, one heme-degrading and the other heme-binding, inhibit clot formation. This may serve as a new therapeutic strategy in preventing and treating venous thromboembolic disease.- Published
- 2020
- Full Text
- View/download PDF
19. A novel, highly potent and selective phosphodiesterase-9 inhibitor for the treatment of sickle cell disease.
- Author
-
McArthur JG, Svenstrup N, Chen C, Fricot A, Carvalho C, Nguyen J, Nguyen P, Parachikova A, Abdulla F, Vercellotti GM, Hermine O, Edwards D, Ribeil JA, Belcher JD, and Maciel TT
- Subjects
- Animals, Fetal Hemoglobin, Humans, Hydroxyurea pharmacology, K562 Cells, Mice, Phosphoric Diester Hydrolases, Anemia, Sickle Cell drug therapy, Phosphodiesterase Inhibitors therapeutic use
- Abstract
The most common treatment for patients with sickle cell disease (SCD) is the chemotherapeutic hydroxyurea, a therapy with pleiotropic effects, including increasing fetal hemoglobin (HbF) in red blood cells and reducing adhesion of white blood cells to the vascular endothelium. Hydroxyurea has been proposed to mediate these effects through a mechanism of increasing cellular cGMP levels. An alternative path to increasing cGMP levels in these cells is through the use of phosphodiesterase-9 inhibitors that selectively inhibit cGMP hydrolysis and increase cellular cGMP levels. We have developed a novel, potent and selective phosphodiesterase-9 inhibitor (IMR-687) specifically for the treatment of SCD. IMR-687 increased cGMP and HbF in erythroid K562 and UT-7 cells and increased the percentage of HbF positive erythroid cells generated in vitro using a two-phase liquid culture of CD34
+ progenitors from sickle cell blood or bone marrow. Oral daily dosing of IMR-687 in the Townes transgenic mouse SCD model, increased HbF and reduced red blood cell sickling, immune cell activation and microvascular stasis. The IMR-687 reduction in red blood cell sickling and immune cell activation was greater than that seen with physiological doses of hydroxyurea. In contrast to other described phosphodiesterase-9 inhibitors, IMR-687 did not accumulate in the central nervous system, where it would inhibit phosphodiesterase-9 in neurons, or alter rodent behavior. IMR-687 was not genotoxic or myelotoxic and did not impact fertility or fetal development in rodents. These data suggest that IMR-687 may offer a safe and effective oral alternative for hydroxyurea in the treatment of SCD., (Copyright© 2020 Ferrata Storti Foundation.)- Published
- 2020
- Full Text
- View/download PDF
20. Heme oxygenase-2 protects against ischemic acute kidney injury: influence of age and sex.
- Author
-
Nath KA, Garovic VD, Grande JP, Croatt AJ, Ackerman AW, Farrugia G, Katusic ZS, Belcher JD, and Vercellotti GM
- Subjects
- Acute Kidney Injury enzymology, Acute Kidney Injury pathology, Acute Kidney Injury physiopathology, Age Factors, Animals, Disease Models, Animal, Female, Heme Oxygenase (Decyclizing) deficiency, Heme Oxygenase (Decyclizing) genetics, Kidney pathology, Kidney physiopathology, Male, Mice, Knockout, Phosphorylation, Reperfusion Injury enzymology, Reperfusion Injury pathology, Reperfusion Injury physiopathology, STAT3 Transcription Factor metabolism, Sex Factors, Signal Transduction, Acute Kidney Injury prevention & control, Heme Oxygenase (Decyclizing) metabolism, Kidney enzymology, Reperfusion Injury prevention & control
- Abstract
Heme oxygenase (HO) activity is exhibited by inducible (HO-1) and constitutive (HO-2) proteins. HO-1 protects against ischemic and nephrotoxic acute kidney injury (AKI). We have previously demonstrated that HO-2 protects against heme protein-induced AKI. The present study examined whether HO-2 is protective in ischemic AKI. Renal ischemia was imposed on young and aged HO-2
+/+ and HO-2-/- mice. On days 1 and 2 after renal ischemia, there were no significant differences in renal function between young male HO-2+/+ and HO-2-/- mice, between young female HO-2+/+ and HO-2-/- mice, or between aged female HO-2+/+ and HO-2-/- mice. However, in aged male mice, HO-2 deficiency worsened renal function on days 1 and 2 after ischemic AKI, and, on day 2 after ischemia, such deficiency augmented upregulation of injury-related genes and worsened histological injury. Renal HO activity was markedly decreased in unstressed aged male HO-2-/- mice and remained so after ischemia, despite exaggerated HO-1 induction in HO-2-/- mice after ischemia. Such exacerbation of deficiency of HO-2 protein and HO activity may reflect phosphorylated STAT3, as activation of this proinflammatory transcription factor was accentuated early after ischemia in aged male HO-2-/- mice. This exacerbation may not reflect impaired induction of nephroprotectant genes, since the induction of HO-1, sirtuin 1, and β-catenin was accentuated in aged male HO-2-/- mice after ischemia. We conclude that aged male mice are hypersensitive to ischemic AKI and that HO-2 mitigates such sensitivity. We speculate that this protective effect of HO-2 may be mediated, at least in part, by suppression of phosphorylated STAT3-dependent signaling.- Published
- 2019
- Full Text
- View/download PDF
21. Critical role of C5a in sickle cell disease.
- Author
-
Vercellotti GM, Dalmasso AP, Schaid TR Jr, Nguyen J, Chen C, Ericson ME, Abdulla F, Killeen T, Lindorfer MA, Taylor RP, and Belcher JD
- Subjects
- Anemia, Sickle Cell drug therapy, Anemia, Sickle Cell genetics, Anemia, Sickle Cell pathology, Animals, Cerebrovascular Disorders drug therapy, Cerebrovascular Disorders genetics, Cerebrovascular Disorders pathology, Complement C3 genetics, Complement C5a antagonists & inhibitors, Complement C5a genetics, Complement Membrane Attack Complex genetics, Complement Membrane Attack Complex immunology, Disease Models, Animal, E-Selectin genetics, E-Selectin immunology, Gene Expression Regulation, Humans, Immunity, Innate, Intercellular Adhesion Molecule-1 genetics, Intercellular Adhesion Molecule-1 immunology, Kidney blood supply, Kidney drug effects, Kidney immunology, Kidney pathology, Liver blood supply, Liver drug effects, Liver immunology, Liver pathology, Lung blood supply, Lung drug effects, Lung immunology, Lung pathology, Male, Mice, Mice, Transgenic, NF-kappa B genetics, NF-kappa B immunology, P-Selectin antagonists & inhibitors, P-Selectin genetics, P-Selectin immunology, Receptor, Anaphylatoxin C5a antagonists & inhibitors, Receptor, Anaphylatoxin C5a genetics, Signal Transduction, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 immunology, Vascular Cell Adhesion Molecule-1 genetics, Vascular Cell Adhesion Molecule-1 immunology, Anemia, Sickle Cell immunology, Antibodies, Neutralizing pharmacology, Cerebrovascular Disorders immunology, Complement C3 immunology, Complement C5a immunology, Receptor, Anaphylatoxin C5a immunology
- Abstract
Innate immune complement activation may contribute to sickle cell disease (SCD) pathogenesis. Ischemia-reperfusion physiology is a key component of the inflammatory and vaso-occlusive milieu in SCD and is associated with complement activation. C5a is an anaphylatoxin, a potent pro-inflammatory mediator that can activate leukocytes, platelets, and endothelial cells, all of which play a role in vaso-occlusion. We hypothesize that hypoxia-reoxygenation (H/R) in SCD mice activates complement, promoting inflammation and vaso-occlusion. At baseline and after H/R, sickle Townes-SS mice had increased C3 activation fragments and C5b-9 deposition in kidneys, livers and lungs and alternative pathway Bb fragments in plasma compared to control AA-mice. Activated complement promoted vaso-occlusion (microvascular stasis) in SS-mice; infusion of zymosan-activated, but not heat-inactivated serum, induced substantial vaso-occlusion in the skin venules of SS-mice. Infusion of recombinant C5a induced stasis in SS, but not AA-mice that was blocked by anti-C5a receptor (C5aR) IgG. C5a-mediated stasis was accompanied by inflammatory responses in SS-mice including NF-κB activation and increased expression of TLR4 and adhesion molecules VCAM-1, ICAM-1, and E-selectin in the liver. Anti-C5aR IgG blocked these inflammatory responses. Also, C5a rapidly up-regulated Weibel-Palade body P-selectin and von Willebrand factor on the surface of human umbilical vein endothelial cells in vitro and on vascular endothelium in vivo. In SS-mice, a blocking antibody to P-selectin inhibited C5a-induced stasis. Similarly, an antibody to C5 that blocks murine C5 cleavage or an antibody that blocks C5aR inhibited H/R-induced stasis in SS-mice. These results suggest that inhibition of C5a may be beneficial in SCD., (© 2018 Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
22. Hemoglobin oxidation-dependent reactions promote interactions with band 3 and oxidative changes in sickle cell-derived microparticles.
- Author
-
Jana S, Strader MB, Meng F, Hicks W, Kassa T, Tarandovskiy I, De Paoli S, Simak J, Heaven MR, Belcher JD, Vercellotti GM, and Alayash AI
- Subjects
- Anemia, Sickle Cell drug therapy, Animals, Antisickling Agents pharmacology, Cell-Derived Microparticles metabolism, Endothelial Cells drug effects, Energy Metabolism, Hemoglobin, Sickle drug effects, Hemoglobin, Sickle metabolism, Hemoglobins metabolism, Humans, Hydroxyurea administration & dosage, Mice genetics, Oxidation-Reduction drug effects, Oxidative Stress physiology, Proteomics, Cell-Derived Microparticles drug effects, Hemoglobins drug effects, Hydroxyurea pharmacology
- Abstract
The contribution of intracellular hemoglobin (Hb) oxidation to RBC-derived microparticle (MP) formation is poorly defined in sickle cell disease (SCD). Here we report that sickle Hb (HbS) oxidation, coupled with changes in cytosolic antioxidative proteins, is associated with membrane alterations and MP formation in homozygous Townes-sickle cell (Townes-SS) mice. Photometric and proteomic analyses confirmed the presence of high levels of Hb oxidation intermediates (ferric/ferryl) and consequent β-globin posttranslational modifications, including the irreversible oxidation of βCys93 and the ubiquitination of βLys96 and βLys145. This is the first report to our knowledge to link the UPS (via ubiquitinated Hb and other proteins) to oxidative stress. Ferryl Hb also induced complex formation with band 3 and RBC membrane proteins. Incubation of Townes-SS MPs with human endothelial cells caused greater loss of monolayer integrity, apoptotic activation, heme oxygenase-1 induction, and concomitant bioenergetic imbalance compared with control Townes-AA MPs. MPs obtained from Townes-SS mice treated with hydroxyurea produced fewer posttranslational Hb modifications. In vitro, hydroxyurea reduced the levels of ferryl Hb and shielded its target residue, βCys93, by a process of S-nitrosylation. These mechanistic analyses suggest potential antioxidative therapeutic modalities that may interrupt MP heme-mediated pathophysiology in SCD patients.
- Published
- 2018
- Full Text
- View/download PDF
23. Oral carbon monoxide therapy in murine sickle cell disease: Beneficial effects on vaso-occlusion, inflammation and anemia.
- Author
-
Belcher JD, Gomperts E, Nguyen J, Chen C, Abdulla F, Kiser ZM, Gallo D, Levy H, Otterbein LE, and Vercellotti GM
- Subjects
- Administration, Oral, Anemia, Sickle Cell blood, Anemia, Sickle Cell genetics, Animals, Antisickling Agents pharmacology, Carbon Monoxide pharmacology, Disease Models, Animal, Erythrocyte Count, Female, Hematocrit, Heme Oxygenase-1 metabolism, Hemoglobins analysis, Hemolysis drug effects, Humans, Inflammation blood, Leukocyte Count, Liver drug effects, Liver metabolism, Male, Membrane Proteins metabolism, Mice, Mice, Transgenic, Microvessels drug effects, NF-E2-Related Factor 2 metabolism, Treatment Outcome, Anemia, Sickle Cell drug therapy, Antisickling Agents therapeutic use, Carbon Monoxide therapeutic use, Inflammation drug therapy
- Abstract
Carbon monoxide (CO) at low, non-toxic concentrations has been previously demonstrated to exert anti-inflammatory protection in murine models of sickle cell disease (SCD). However CO delivery by inhalation, CO-hemoglobin infusion or CO-releasing molecules presents problems for daily CO administration. Oral administration of a CO-saturated liquid avoids many of these issues and potentially provides a platform for self-administration to SCD patients. To test if orally-delivered CO could modulate SCD vaso-occlusion and inflammation, a liquid CO formulation (HBI-002) was administered by gavage (10 ml/kg) once-daily to NY1DD and Townes-SS transgenic mouse models of SCD. Baseline CO-hemoglobin (CO-Hb) levels were 1.6% and 1.8% in NY1DD and Townes-SS sickle mice and 0.6% in Townes-AS control mice. CO-Hb levels reached 5.4%, 4.7% and 3.0% within 5 minutes in NY1DD, SS and AS mice respectively after gavage with HBI-002. After ten treatments, each once-daily, hemoglobin levels rose from 5.3g/dL in vehicle-treated Townes-SS mice to 6.3g/dL in HBI-002-treated. Similarly, red blood cell (RBC) counts rose from 2.36 x 106/μL in vehicle-treated SS mice to 2.89 x 106/μL in HBI-002-treated mice. In concordance with these findings, hematocrits rose from 26.3% in vehicle-treated mice to 30.0% in HBI-002-treated mice. Reticulocyte counts were not significantly different between vehicle and HBI-002-treated SS mice implying less hemolysis and not an increase in RBC production. White blood cell counts decreased from 29.1 x 103/μL in vehicle-treated versus 20.3 x 103/μL in HBI-002-treated SS mice. Townes-SS mice treated with HBI-002 had markedly increased Nrf2 and HO-1 expression and decreased NF-κB activation compared to vehicle-treated mice. These anti-inflammatory effects were examined for the ability of HBI-002 (administered orally once-daily for up to 5 days) to inhibit vaso-occlusion induced by hypoxia-reoxygenation. In NY1DD and Townes-SS sickle mice, HBI-002 decreased microvascular stasis in a duration-dependent manner. Collectively, these findings support HBI-002 as a useful anti-inflammatory agent to treat SCD and warrants further development as a therapeutic., Competing Interests: Drs. John Belcher and Greg Vercellotti are employed by the University of Minnesota. Dr. John Belcher is a PI on the SBIR grant. Dr. Edward Gomperts is PI on the SBIR grant, is Executive Chairman of Hillhurst Biopharmaceuticals Inc., is an inventor on a patent and a patent application regarding HBI-002, and has shares in Hillhurst Biopharmaceuticals, Inc. Drs. Leo Otterbein and Howard Levy are consultants to and have shares in Hillhurst Biopharmaceuticals, Inc. HBI-002 is a therapeutic product under development by Hillhurst Biopharmaceuticals, Inc., and is not licensed at this time. This commercial affiliation does not alter our adherence to PLOS ONE policies on sharing data and materials. The specific roles of these authors are articulated with their initials in the 'author contributions' section.
- Published
- 2018
- Full Text
- View/download PDF
24. Serum haptoglobin and hemopexin levels are depleted in pediatric sickle cell disease patients.
- Author
-
Santiago RP, Guarda CC, Figueiredo CVB, Fiuza LM, Aleluia MM, Adanho CSA, Carvalho MOS, Pitanga TN, Zanette DL, Lyra IM, Nascimento VML, Vercellotti GM, Belcher JD, and Goncalves MS
- Subjects
- Child, Humans, Anemia, Sickle Cell metabolism, Haptoglobins analysis, Hemopexin analysis
- Published
- 2018
- Full Text
- View/download PDF
25. Bivalent ligand MCC22 potently attenuates nociception in a murine model of sickle cell disease.
- Author
-
Cataldo G, Lunzer MM, Olson JK, Akgün E, Belcher JD, Vercellotti GM, Portoghese PS, and Simone DA
- Subjects
- Analgesics therapeutic use, Analgesics, Opioid therapeutic use, Animals, Disease Models, Animal, Hyperalgesia physiopathology, Male, Mice, Mice, Transgenic, Analgesics pharmacology, Analgesics, Opioid pharmacology, Anemia, Sickle Cell physiopathology, Hyperalgesia drug therapy, Nociception drug effects
- Abstract
Sickle cell disease (SCD) is a chronic inflammatory disorder accompanied by chronic pain. In addition to ongoing pain and hyperalgesia, vaso-occlusive crises-induced pain can be chronic or episodic. Because analgesics typically used to treat pain are not very effective in SCD, opioids, including morphine, are a primary treatment for managing pain in SCD but are associated with many serious side effects, including constipation, tolerance, addiction, and respiratory depression. Thus, there is a need for the development of novel treatments for pain in SCD. In this study, we used the Townes transgenic mouse model of SCD to investigate the antinociceptive efficacy of the bivalent ligand, MCC22, and compared its effectiveness with morphine. MCC22 consists of a mu-opioid receptor agonist and a chemokine receptor-5 (CCR5) antagonist that are linked through a 22-atom spacer. Our results show that intraperitoneal administration of MCC22 produced exceptionally potent dose-dependent antihyperalgesia as compared to morphine, dramatically decreased evoked responses of nociceptive dorsal horn neurons, and decreased expression of proinflammatory cytokines in the spinal cord. Moreover, tolerance did not develop to its analgesic effects after repeated administration. In view of the extraordinary potency of MCC22 without tolerance, MCC22 and similar compounds may vastly improve the management of pain associated with SCD.
- Published
- 2018
- Full Text
- View/download PDF
26. Role of TLR4 signaling in the nephrotoxicity of heme and heme proteins.
- Author
-
Nath KA, Belcher JD, Nath MC, Grande JP, Croatt AJ, Ackerman AW, Katusic ZS, and Vercellotti GM
- Subjects
- Acute Kidney Injury chemically induced, Acute Kidney Injury pathology, Acute Kidney Injury prevention & control, Animals, Cell Line, Chemokine CCL2 metabolism, Disease Models, Animal, Epithelial Cells drug effects, Epithelial Cells pathology, Glycerol, Kidney drug effects, Kidney pathology, Male, Mice, Inbred C57BL, Mice, Knockout, NF-kappa B metabolism, Rats, Sulfonamides pharmacology, Toll-Like Receptor 4 deficiency, Toll-Like Receptor 4 genetics, Acute Kidney Injury metabolism, Epithelial Cells metabolism, Hemin, Kidney blood supply, Kidney metabolism, Renal Circulation drug effects, Signal Transduction drug effects, Toll-Like Receptor 4 antagonists & inhibitors, Toll-Like Receptor 4 metabolism, Vasoconstriction drug effects
- Abstract
Destabilized heme proteins release heme, and free heme is toxic. Heme is now recognized as an agonist for the Toll-like receptor-4 (TLR4) receptor. This study examined whether the TLR4 receptor mediates the nephrotoxicity of heme, specifically, the effects of heme on renal blood flow and inflammatory responses. We blocked TLR4 signaling by the specific antagonist TAK-242. Intravenous administration of heme to mice promptly reduced renal blood flow, an effect attenuated by TAK-242. In vitro, TAK-242 reduced heme-elicited activation of NF-κB and its downstream gene monocyte chemoattractant protein-1(MCP-1); in contrast, TAK-242 failed to reduce heme-induced activation of the anti-inflammatory transcription factor Nrf2 and its downstream gene heme oxygenase-1 (HO-1). TAK-242 did not reduce heme-induced renal MCP-1 upregulation in vivo. TAK-242 did not reduce dysfunction and histological injury in the glycerol model of heme protein-induced acute kidney injury (AKI), findings corroborated by studies in TLR4
+/+ and TLR4-/- mice. We conclude that 1) acute heme-mediated renal vasoconstriction occurs through TLR4 signaling; 2) proinflammatory effects of heme in renal epithelial cells involve TLR4 signaling, whereas the anti-inflammatory effects of heme do not; 3) TLR4 signaling does not mediate the proinflammatory effects of heme in the kidney; and 4) major mechanisms underlying glycerol-induced, heme protein-mediated AKI do not involve TLR4 signaling. These findings in the glycerol model are in stark contrast with findings in virtually all other AKI models studied to date and emphasize the importance of TLR4-independent pathways of heme protein-mediated injury in this model. Finally, these studies urge caution when using observations derived in vitro to predict what occurs in vivo.- Published
- 2018
- Full Text
- View/download PDF
27. Haptoglobin and hemopexin inhibit vaso-occlusion and inflammation in murine sickle cell disease: Role of heme oxygenase-1 induction.
- Author
-
Belcher JD, Chen C, Nguyen J, Abdulla F, Zhang P, Nguyen H, Nguyen P, Killeen T, Miescher SM, Brinkman N, Nath KA, Steer CJ, and Vercellotti GM
- Subjects
- Aldehydes analysis, Anemia, Sickle Cell pathology, Animals, Carbon Monoxide pharmacology, Cytokines analysis, Disease Models, Animal, Female, Gene Expression drug effects, Haptoglobins pharmacology, Hemopexin pharmacology, Intercellular Adhesion Molecule-1, Male, Metalloporphyrins pharmacology, Mice, Microsomes, Liver metabolism, Protoporphyrins pharmacology, Skin metabolism, Skin pathology, Transcription Factor RelA metabolism, Vascular Cell Adhesion Molecule-1 metabolism, Anemia, Sickle Cell prevention & control, Haptoglobins therapeutic use, Heme Oxygenase-1 metabolism, Hemopexin therapeutic use, Inflammation prevention & control
- Abstract
During hemolysis, hemoglobin and heme released from red blood cells promote oxidative stress, inflammation and thrombosis. Plasma haptoglobin and hemopexin scavenge free hemoglobin and heme, respectively, but can be depleted in hemolytic states. Haptoglobin and hemopexin supplementation protect tissues, including the vasculature, liver and kidneys. It is widely assumed that these protective effects are due primarily to hemoglobin and heme clearance from the vasculature. However, this simple assumption does not account for the consequent cytoprotective adaptation seen in cells and organs. To further address the mechanism, we used a hyperhemolytic murine model (Townes-SS) of sickle cell disease to examine cellular responses to haptoglobin and hemopexin supplementation. A single infusion of haptoglobin or hemopexin (± equimolar hemoglobin) in SS-mice increased heme oxygenase-1 (HO-1) in the liver, kidney and skin several fold within 1 hour and decreased nuclear NF-ĸB phospho-p65, and vaso-occlusion for 48 hours after infusion. Plasma hemoglobin and heme levels were not significantly changed 1 hour after infusion of haptoglobin or hemopexin. Haptoglobin and hemopexin also inhibited hypoxia/reoxygenation and lipopolysaccharide-induced vaso-occlusion in SS-mice. Inhibition of HO-1 activity with tin protoporphyrin blocked the protections afforded by haptoglobin and hemopexin in SS-mice. The HO-1 reaction product carbon monoxide, fully restored the protection, in part by inhibiting Weibel-Palade body mobilization of P-selectin and von Willebrand factor to endothelial cell surfaces. Thus, the mechanism by which haptoglobin and hemopexin supplementation in hyperhemolytic SS-mice induces cytoprotective cellular responses is linked to increased HO-1 activity.
- Published
- 2018
- Full Text
- View/download PDF
28. Case series supporting heme detoxification via therapeutic plasma exchange in acute multiorgan failure syndrome resistant to red blood cell exchange in sickle cell disease.
- Author
-
Louie JE, Anderson CJ, Fayaz M Fomani K, Henry A, Killeen T, Mohandas N, Yazdanbakhsh K, Belcher JD, Vercellotti GM, and Shi PA
- Subjects
- Adult, Erythrocyte Transfusion, Female, Humans, Male, Middle Aged, Heme metabolism, Multiple Organ Failure blood, Multiple Organ Failure therapy, Plasma, Plasma Exchange
- Abstract
Background: Depletion of haptoglobin (Hp) and hemopexin (Hx) with increase in free hemoglobin and heme are important etiologies of vaso-occlusive complications in sickle cell disease (SCD). This study is the first to show an association between clinical improvement in SCD and repletion of Hp and Hx by therapeutic plasma exchange (TPE) using plasma replacement., Study Design and Methods: Thirteen fresh-frozen plasma (FFP) units derived from consecutive whole blood donations were thawed at 37°C after 10 months of storage; Hp and Hx concentrations immediately postthaw and after 5 days of refrigerated storage were analyzed by enzyme-linked immunosorbent assay (ELISA). All SCD patients presenting to a single institution over a 2-year period with acute multiorgan failure syndrome resistant to red blood cell exchange (RCE) were treated with TPE with FFP replacement; concentrations of Hp, Hx, and heme were evaluated before and after TPE by ELISA., Results: Plasma concentrations of Hp and Hx decreased approximately 20% (p ≤ 0.002) after 5 days of refrigerated storage. Significant mean fold increases after TPE of 10 for Hp (p < 0.005) and seven for Hx (p < 0.003) and a 30% mean decrease in heme concentrations (p = 0.07) were noted in association with clinical improvement (three patients), whereas minimal increases in Hp and Hx were associated with continued clinical deterioration in one patient., Conclusion: Fresh-frozen plasma rather than thawed plasma is optimal for Hp and Hx replacement. Patient data are consistent with Hp and Hx increases via TPE limiting clinical toxicity of worsened hemolysis associated with severe vaso-occlusive complications refractory to RCE in SCD., (© 2017 AABB.)
- Published
- 2018
- Full Text
- View/download PDF
29. Inflammation in sickle cell disease.
- Author
-
Conran N and Belcher JD
- Subjects
- Anemia, Sickle Cell pathology, Humans, Inflammation drug therapy, Anemia, Sickle Cell complications, Inflammation etiology
- Abstract
The primary β-globin gene mutation that causes sickle cell disease (SCD) has significant pathophysiological consequences that result in hemolytic events and the induction of the inflammatory processes that ultimately lead to vaso-occlusion. In addition to their role in the initiation of the acute painful vaso-occlusive episodes that are characteristic of SCD, inflammatory processes are also key components of many of the complications of the disease including autosplenectomy, acute chest syndrome, pulmonary hypertension, leg ulcers, nephropathy and stroke. We, herein, discuss the events that trigger inflammation in the disease, as well as the mechanisms, inflammatory molecules and cells that propagate these inflammatory processes. Given the central role that inflammation plays in SCD pathophysiology, many of the therapeutic approaches currently under pre-clinical and clinical development for the treatment of SCD endeavor to counter aspects or specific molecules of these inflammatory processes and it is possible that, in the future, we will see anti-inflammatory drugs being used either together with, or in place of, hydroxyurea in those SCD patients for whom hematopoietic stem cell transplants and evolving gene therapies are not a viable option.
- Published
- 2018
- Full Text
- View/download PDF
30. A monocyte-TNF-endothelial activation axis in sickle transgenic mice: Therapeutic benefit from TNF blockade.
- Author
-
Solovey A, Somani A, Belcher JD, Milbauer L, Vincent L, Pawlinski R, Nath KA, Kelm RJ Jr, Mackman N, O'Sullivan MG, Gupta K, Vercellotti GM, and Hebbel RP
- Subjects
- Anemia, Sickle Cell diagnosis, Anemia, Sickle Cell drug therapy, Anemia, Sickle Cell genetics, Animals, Antibodies, Monoclonal pharmacology, Biomarkers, Bone Marrow Transplantation, Cell Aggregation genetics, Cell Aggregation immunology, Disease Models, Animal, Early Growth Response Protein 1 genetics, Early Growth Response Protein 1 metabolism, Endothelium, Vascular metabolism, Etanercept pharmacology, Etanercept therapeutic use, Heart Function Tests, Humans, Inflammation Mediators, Leukocytes, Mononuclear drug effects, Leukocytes, Mononuclear immunology, Leukocytes, Mononuclear metabolism, Mice, Mice, Knockout, Mice, Transgenic, Molecular Targeted Therapy, Monocytes drug effects, Monocytes immunology, NF-kappa B deficiency, NF-kappa B genetics, Phenotype, Protein Kinase Inhibitors pharmacology, Thromboplastin metabolism, Tumor Necrosis Factor-alpha antagonists & inhibitors, Vascular Cell Adhesion Molecule-1 metabolism, Anemia, Sickle Cell metabolism, Endothelial Cells metabolism, Monocytes metabolism, Signal Transduction drug effects, Tumor Necrosis Factor-alpha metabolism
- Abstract
Elaboration of tumor necrosis factor (TNF) is a very early event in development of ischemia/reperfusion injury pathophysiology. Therefore, TNF may be a prominent mediator of endothelial cell and vascular wall dysfunction in sickle cell anemia, a hypothesis we addressed using NY1DD, S+S
Antilles , and SS-BERK sickle transgenic mice. Transfusion experiments revealed participation of abnormally activated blood monocytes exerting an endothelial activating effect, dependent upon Egr-1 in both vessel wall and blood cells, and upon NFκB(p50) in a blood cell only. Involvement of TNF was identified by beneficial impact from TNF blockers, etanercept and infliximab, with less benefit from an IL-1 blocker, anakinra. In therapeutic studies, etanercept ameliorated multiple disturbances of the murine sickle condition: monocyte activation, blood biomarkers of inflammation, low platelet count and Hb, vascular stasis triggered by hypoxia/reoxygenation (but not if triggered by hemin infusion), tissue production of neuro-inflammatory mediators, endothelial activation (monitored by tissue factor and VCAM-1 expression), histopathologic liver injury, and three surrogate markers of pulmonary hypertension (perivascular inflammatory aggregates, arteriolar muscularization, and right ventricular mean systolic pressure). In aggregate, these studies identify a prominent-and possibly dominant-role for an abnormal monocyte-TNF-endothelial activation axis in the sickle context. Its presence, plus the many benefits of etanercept observed here, argue that pilot testing of TNF blockade should be considered for human sickle cell anemia, a challenging but achievable translational research goal., (© 2017 The Authors American Journal of Hematology Published by Wiley Periodicals, Inc.)- Published
- 2017
- Full Text
- View/download PDF
31. The role of carbon monoxide and heme oxygenase in the prevention of sickle cell disease vaso-occlusive crises.
- Author
-
Gomperts E, Belcher JD, Otterbein LE, Coates TD, Wood J, Skolnick BE, Levy H, and Vercellotti GM
- Subjects
- Anemia, Sickle Cell genetics, Anemia, Sickle Cell therapy, Animals, Carbon Monoxide administration & dosage, Carbon Monoxide adverse effects, Carbon Monoxide blood, Clinical Trials as Topic, Disease Models, Animal, Heme Oxygenase (Decyclizing) blood, Hemoglobins chemistry, Hemoglobins genetics, Hemoglobins metabolism, Humans, Signal Transduction, Treatment Outcome, Anemia, Sickle Cell complications, Anemia, Sickle Cell metabolism, Carbon Monoxide metabolism, Heme Oxygenase (Decyclizing) metabolism, Vascular Diseases etiology, Vascular Diseases prevention & control
- Abstract
Sickle Cell Disease (SCD) is a painful, lifelong hemoglobinopathy inherited as a missense point mutation in the hemoglobin (Hb) beta-globin gene. This disease has significant impact on quality of life and mortality, thus a substantial medical need exists to reduce the vaso-occlusive crises which underlie the pathophysiology of the disease. The concept that a gaseous molecule may exert biological function has been well known for over one hundred years. Carbon monoxide (CO), although studied in SCD for over 50 years, has recently emerged as a powerful cytoprotective biological response modifier capable of regulating a host of physiologic and therapeutic processes that, at low concentrations, exerts key physiological functions in various models of tissue inflammation and injury. CO is physiologically generated by the metabolism of heme by the heme oxygenase enzymes and is measurable in blood. A substantial amount of preclinical and clinical data with CO have been generated, which provide compelling support for CO as a potential therapeutic in a number of pathological conditions. Data underlying the therapeutic mechanisms of CO, including in SCD, have been generated by a plethora of in vitro and preclinical studies including multiple SCD mouse models. These data show CO to have key signaling impacts on a host of metallo-enzymes as well as key modulating genes that in sum, result in significant anti-inflammatory, anti-oxidant and anti-apoptotic effects as well as vasodilation and anti-adhesion of cells to the endothelium resulting in preservation of vascular flow. CO may also have a role as an anti-polymerization HbS agent. In addition, considerable scientific data in the non-SCD literature provide evidence for a beneficial impact of CO on cerebrovascular complications, suggesting that in SCD, CO could potentially limit these highly problematic neurologic outcomes. Research is needed and hopefully forthcoming, to carefully elucidate the safety and benefits of this potential therapy across the age spectrum of patients impacted by the host of pathophysiological complications of this devastating disease., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
32. Control of Oxidative Stress and Inflammation in Sickle Cell Disease with the Nrf2 Activator Dimethyl Fumarate.
- Author
-
Belcher JD, Chen C, Nguyen J, Zhang P, Abdulla F, Nguyen P, Killeen T, Xu P, O'Sullivan G, Nath KA, and Vercellotti GM
- Subjects
- Administration, Oral, Animals, Dimethyl Fumarate administration & dosage, Dimethyl Fumarate chemistry, Dose-Response Relationship, Drug, Female, Male, Mice, Mice, Transgenic, Anemia, Sickle Cell drug therapy, Dimethyl Fumarate pharmacology, Inflammation drug therapy, NF-E2-Related Factor 2 metabolism, Oxidative Stress drug effects
- Abstract
Aims: Heme derived from hemolysis is pro-oxidative and proinflammatory and promotes vaso-occlusion in murine models of sickle cell disease (SCD), suggesting that enhanced detoxification of heme may be beneficial. Nuclear factor erythroid-2-related factor-2 (Nrf2) transcription pathway is the principal cellular defense system responding to pro-oxidative and proinflammatory stress. Dimethyl fumarate (DMF), a drug approved for treatment of multiple sclerosis, provides neuroprotection by activating Nrf2-responsive genes. We hypothesized that induction of Nrf2 with DMF would be beneficial in murine SCD models., Results: DMF (30 mg/kg/day) or vehicle (0.08% methyl cellulose) was administered for 3-7 days to NY1DD and HbSS-Townes SCD mice. Vaso-occlusion, a hallmark of SCD, measured in sickle mice with dorsal skinfold chambers, was inhibited by DMF. The inhibitory effect of DMF was abrogated by the heme oxygenase-1 (HO-1) inhibitor tin protoporphyrin. DMF increased nuclear Nrf2 and cellular mRNA of Nrf2-responsive genes in livers and kidneys. DMF increased heme defenses, including HO-1, haptoglobin, hemopexin, and ferritin heavy chain, although plasma hemoglobin and heme levels were unchanged. DMF decreased markers of inflammation, including nuclear factor-kappa B phospho-p65, adhesion molecules, and toll-like receptor 4. DMF administered for 24 weeks to HbSS-Townes mice decreased hepatic necrosis, inflammatory cytokines, and irregularly shaped erythrocytes and increased hemoglobin F, but did not alter hematocrits, reticulocyte counts, lactate dehydrogenase, plasma heme, or spleen weights, indicating that the beneficial effects of DMF were not attributable to decreased hemolysis., Innovation: These studies identify Nrf2 activation as a new therapeutic target for the treatment of SCD., Conclusion: DMF activates Nrf2, enhances antioxidant defenses, and inhibits inflammation and vaso-occlusion in SCD mice. Antioxid. Redox Signal. 26, 748-762.
- Published
- 2017
- Full Text
- View/download PDF
33. Catalytic Determinants of Alkene Production by the Cytochrome P450 Peroxygenase OleT JE .
- Author
-
Matthews S, Belcher JD, Tee KL, Girvan HM, McLean KJ, Rigby SE, Levy CW, Leys D, Parker DA, Blankley RT, and Munro AW
- Subjects
- Amino Acid Sequence, Catalytic Domain, Crystallography, X-Ray, Cytochrome P-450 Enzyme System chemistry, Cytochrome P-450 Enzyme System genetics, Fatty Acids metabolism, Hydroxylation, Models, Molecular, Mutation, Peroxidases chemistry, Peroxidases genetics, Sequence Alignment, Staphylococcaceae chemistry, Staphylococcaceae genetics, Staphylococcaceae metabolism, Substrate Specificity, Alkenes metabolism, Cytochrome P-450 Enzyme System metabolism, Peroxidases metabolism, Staphylococcaceae enzymology
- Abstract
The Jeotgalicoccus sp. peroxygenase cytochrome P450 OleT
JE (CYP152L1) is a hydrogen peroxide-driven oxidase that catalyzes oxidative decarboxylation of fatty acids, producing terminal alkenes with applications as fine chemicals and biofuels. Understanding mechanisms that favor decarboxylation over fatty acid hydroxylation in OleTJE could enable protein engineering to improve catalysis or to introduce decarboxylation activity into P450s with different substrate preferences. In this manuscript, we have focused on OleTJE active site residues Phe79 , His85 , and Arg245 to interrogate their roles in substrate binding and catalytic activity. His85 is a potential proton donor to reactive iron-oxo species during substrate decarboxylation. The H85Q mutant substitutes a glutamine found in several peroxygenases that favor fatty acid hydroxylation. H85Q OleTJE still favors alkene production, suggesting alternative protonation mechanisms. However, the mutant undergoes only minor substrate binding-induced heme iron spin state shift toward high spin by comparison with WT OleTJE , indicating the key role of His85 in this process. Phe79 interacts with His85 , and Phe79 mutants showed diminished affinity for shorter chain (C10-C16) fatty acids and weak substrate-induced high spin conversion. F79A OleTJE is least affected in substrate oxidation, whereas the F79W/Y mutants exhibit lower stability and cysteine thiolate protonation on reduction. Finally, Arg245 is crucial for binding the substrate carboxylate, and R245E/L mutations severely compromise activity and heme content, although alkene products are formed from some substrates, including stearic acid (C18:0). The results identify crucial roles for the active site amino acid trio in determining OleTJE catalytic efficiency in alkene production and in regulating protein stability, heme iron coordination, and spin state., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2017
- Full Text
- View/download PDF
34. Sustained treatment of sickle cell mice with haptoglobin increases HO-1 and H-ferritin expression and decreases iron deposition in the kidney without improvement in kidney function.
- Author
-
Shi PA, Choi E, Chintagari NR, Nguyen J, Guo X, Yazdanbakhsh K, Mohandas N, Alayash AI, Manci EA, Belcher JD, and Vercellotti GM
- Subjects
- Anemia, Sickle Cell complications, Anemia, Sickle Cell drug therapy, Anemia, Sickle Cell genetics, Animals, Apoferritins genetics, Blood Cell Count, Disease Models, Animal, Female, Gene Expression, Haptoglobins administration & dosage, Haptoglobins adverse effects, Haptoglobins pharmacokinetics, Heme Oxygenase-1 genetics, Heme Oxygenase-1 metabolism, Kidney Diseases etiology, Kidney Diseases physiopathology, Male, Mice, Mice, Transgenic, Treatment Outcome, Anemia, Sickle Cell metabolism, Apoferritins metabolism, Haptoglobins pharmacology, Iron metabolism, Kidney Diseases metabolism, Kidney Diseases pathology
- Abstract
There is growing evidence that extracellular haemoglobin and haem mediate inflammatory and oxidative damage in sickle cell disease. Haptoglobin (Hp), the scavenger for free haemoglobin, is depleted in most patients with sickle cell disease due to chronic haemolysis. Although single infusions of Hp can ameliorate vaso-occlusion in mouse models of sickle cell disease, prior studies have not examined the therapeutic benefits of more chronic Hp dosing on sickle cell disease manifestations. In the present study, we explored the effect of Hp treatment over a 3-month period in sickle mice at two dosing regimens: the first at a moderate dose of 200 mg/kg thrice weekly and the second at a higher dose of 400 mg/kg thrice weekly. We found that only the higher dosing regimen resulted in increased haem-oxygenase-1 and heavy chain ferritin (H-ferritin) expression and decreased iron deposition in the kidney. Despite the decreased kidney iron deposition following Hp treatment, there was no significant improvement in kidney function. However, there was a nearly significant trend towards decreased liver infarction., (© 2016 John Wiley & Sons Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
35. Hepatic Overexpression of Hemopexin Inhibits Inflammation and Vascular Stasis in Murine Models of Sickle Cell Disease.
- Author
-
Vercellotti GM, Zhang P, Nguyen J, Abdulla F, Chen C, Nguyen P, Nowotny C, Steer CJ, Smith A, and Belcher JD
- Abstract
Sickle cell disease (SCD) patients have low serum hemopexin (Hpx) levels due to chronic hemolysis. We hypothesize that in SCD mice, hepatic overexpression of hemopexin will scavenge the proximal mediator of vascular activation, heme, and will inhibit inflammation and microvascular stasis. To examine the protective role of Hpx in SCD, we transplanted bone marrow from NY1DD SCD mice into Hpx
™/™ or Hpx+/+ C57BL/6 mice. Dorsal skin fold chambers were implanted in week 13 post-transplant and microvascular stasis (% non-flowing venules) evaluated in response to heme infusion. Hpx™/™ sickle mice had significantly greater microvascular stasis in response to heme infusion than Hpx+/+ sickle mice (p<0.05), demonstrating the protective effect of Hpx in SCD. We utilized Sleeping Beauty (SB) transposon-mediated gene transfer to overexpress wild-type rat Hpx (wt-Hpx) in NY1DD and Townes-SS SCD mice. Control SCD mice were treated with lactated Ringer's solution (LRS) or a luciferase (Luc) plasmid. Plasma and hepatic Hpx were significantly increased compared to LRS and Luc controls. Microvascular stasis in response to heme infusion in NY1DD and Townes-SS mice overexpressing wt-Hpx had significantly less stasis than controls (p<0.05). Wt-Hpx overexpression markedly increased hepatic nuclear Nrf2 expression, HO-1 activity and protein, the heme-Hpx binding protein and scavenger receptor, CD91/LRP1 and decreased NF-κB activation. Two missense (ms)-Hpx SB-constructs that bound neither heme nor the Hpx receptor, CD91/LRP1, did not prevent heme-induced stasis. In conclusion, increasing Hpx levels in transgenic sickle mice via gene transfer activates the Nrf2/HO-1 anti-oxidant axis and ameliorates inflammation and vaso-occlusion.- Published
- 2016
- Full Text
- View/download PDF
36. Liver-targeted gene therapy: Approaches and challenges.
- Author
-
Aravalli RN, Belcher JD, and Steer CJ
- Subjects
- Animals, Genetic Vectors, Humans, Liver Diseases therapy, Targeted Gene Repair
- Abstract
The liver plays a major role in many inherited and acquired genetic disorders. It is also the site for the treatment of certain inborn errors of metabolism that do not directly cause injury to the liver. The advancement of nucleic acid-based therapies for liver maladies has been severely limited because of the myriad untoward side effects and methodological limitations. To address these issues, research efforts in recent years have been intensified toward the development of targeted gene approaches using novel genetic tools, such as zinc-finger nucleases, transcription activator-like effector nucleases, and clustered regularly interspaced short palindromic repeats as well as various nonviral vectors such as Sleeping Beauty transposons, PiggyBac transposons, and PhiC31 integrase. Although each of these methods uses a distinct mechanism of gene modification, all of them are dependent on the efficient delivery of DNA and RNA molecules into the cell. This review provides an overview of current and emerging therapeutic strategies for liver-targeted gene therapy and gene repair., (© 2015 American Association for the Study of Liver Diseases.)
- Published
- 2015
- Full Text
- View/download PDF
37. Haptoglobin attenuates hemoglobin-induced heme oxygenase-1 in renal proximal tubule cells and kidneys of a mouse model of sickle cell disease.
- Author
-
Chintagari NR, Nguyen J, Belcher JD, Vercellotti GM, and Alayash AI
- Subjects
- Animals, Cell Line, Disease Models, Animal, Female, Humans, Kidney metabolism, Kidney Tubules cytology, Kidney Tubules pathology, Male, Mice, Mice, Inbred C57BL, Oxidative Stress, Anemia, Sickle Cell complications, Anemia, Sickle Cell metabolism, Haptoglobins metabolism, Heme Oxygenase-1 metabolism, Hemoglobin A metabolism, Kidney pathology
- Abstract
Sickle cell disease (SCD), a hereditary hemolytic disorder is characterized by chronic hemolysis, oxidative stress, vaso-occlusion and end-organ damage. Hemolysis releases toxic cell-free hemoglobin (Hb) into circulation. Under physiologic conditions, plasma Hb binds to haptoglobin (Hp) and forms Hb-Hp dimers. The dimers bind to CD163 receptors on macrophages for further internalization and degradation. However, in SCD patients plasma Hp is depleted and free Hb is cleared primarily by proximal tubules of kidneys. Excess free Hb in plasma predisposes patients to renal damage. We hypothesized that administration of exogenous Hp reduces Hb-mediated renal damage. To test this hypothesis, human renal proximal tubular cells (HK-2) were exposed to HbA (50μM heme) for 24h. HbA increased the expression of heme oxygenase-1 (HO-1), an enzyme which degrades heme, reduces heme-mediated oxidative toxicity, and confers cytoprotection. Similarly, infusion of HbA (32μM heme/kg) induced HO-1 expression in kidneys of SCD mice. Immunohistochemistry confirmed the increased HO-1 expression in the proximal tubules of the kidney. Exogenous Hp attenuated the HbA-induced HO-1 expression in vitro and in SCD mice. Our results suggest that Hb-mediated oxidative toxicity may contribute to renal damage in SCD and that Hp treatment reduces heme/iron toxicity in the kidneys following hemolysis., (Published by Elsevier Inc.)
- Published
- 2015
- Full Text
- View/download PDF
38. The fucosylation inhibitor, 2-fluorofucose, inhibits vaso-occlusion, leukocyte-endothelium interactions and NF-ĸB activation in transgenic sickle mice.
- Author
-
Belcher JD, Chen C, Nguyen J, Abdulla F, Nguyen P, Nguyen M, Okeley NM, Benjamin DR, Senter PD, and Vercellotti GM
- Subjects
- Anemia, Sickle Cell drug therapy, Animals, Cell Adhesion drug effects, Cells, Cultured, Disease Models, Animal, Endothelial Cells drug effects, Endothelial Cells metabolism, Endothelium metabolism, Erythrocytes drug effects, Erythrocytes metabolism, Female, Heme metabolism, Hemoglobins metabolism, Humans, Inflammation drug therapy, Inflammation metabolism, Leukocyte Rolling drug effects, Leukocytes metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Neutrophils drug effects, Neutrophils metabolism, Vascular Cell Adhesion Molecule-1 metabolism, Venules drug effects, Venules metabolism, Anemia, Sickle Cell metabolism, Endothelium drug effects, Fucose pharmacology, Leukocytes drug effects, Microcirculation drug effects, NF-kappa B metabolism
- Abstract
2-Fluorofucose (2FF) blocks the fucosylation and the tethering of sialyl-Lewisx tetrasaccharide and structural variants on leukocytes and red blood cells to P- and E-selectins on activated endothelial cell surfaces. Because P- and E-selectin are required for vaso-occlusion in murine sickle cell disease (SCD), we investigated whether 2FF would inhibit vaso-occlusion in SCD mice. Microvascular stasis was measured in subcutaneous venules in NY1DD and HbSS-Townes SCD mice with dorsal skin-fold chambers after infusion of hemoglobin or exposure to hypoxia/reoxygenation. 2FF in drinking water or administered by gavage inhibited stasis in sickle mice in a dose-responsive manner. Significant inhibitory effects on stasis were seen 1 day post-treatment. 2FF treatment of SCD mice also significantly reduced leukocyte rolling and adhesion along the vessel walls of SCD mice and the static adhesion of neutrophils and sickle red blood cells isolated from 2FF-treated SCD mice to resting and activated endothelial cells. Total white blood cell counts increased in response to 2FF. NF-ĸB activation and VCAM-1 and E-selectin expression were inhibited in the livers of SCD mice consistent with an overall decrease in vascular inflammation and ischemia-reperfusion physiology. Pretreatment with 2FF completely eliminated heme-induced lethality in HbSS-Townes mice, consistent with the observed anti-inflammatory and anti-adhesive properties of 2FF in SCD mice. These data suggest that 2FF may be beneficial for preventing or treating vaso-occlusive crises in SCD patients.
- Published
- 2015
- Full Text
- View/download PDF
39. H-ferritin ferroxidase induces cytoprotective pathways and inhibits microvascular stasis in transgenic sickle mice.
- Author
-
Vercellotti GM, Khan FB, Nguyen J, Chen C, Bruzzone CM, Bechtel H, Brown G, Nath KA, Steer CJ, Hebbel RP, and Belcher JD
- Abstract
Hemolysis, oxidative stress, inflammation, vaso-occlusion, and organ infarction are hallmarks of sickle cell disease (SCD). We have previously shown that increases in heme oxygenase-1 (HO-1) activity detoxify heme and inhibit vaso-occlusion in transgenic mouse models of SCD. HO-1 releases Fe(2+) from heme, and the ferritin heavy chain (FHC) ferroxidase oxidizes Fe(2+) to catalytically inactive Fe(3+) inside ferritin. FHC overexpression has been shown to be cytoprotective. In this study, we hypothesized that overexpression of FHC and its ferroxidase activity will inhibit inflammation and microvascular stasis in transgenic SCD mice in response to plasma hemoglobin. We utilized a Sleeping Beauty (SB) transposase plasmid to deliver a human wild-type-ferritin heavy chain (wt-hFHC) transposable element by hydrodynamic tail vein injections into NY1DD SCD mice. Control SCD mice were infused with the same volume of lactated Ringer's solution (LRS) or a human triple missense FHC (ms-hFHC) plasmid with no ferroxidase activity. 8 weeks later, LRS-injected mice had ~40% microvascular stasis (% non-flowing venules) 1 h after infusion of stroma-free hemoglobin, while mice overexpressing wt-hFHC had only 5% stasis (p < 0.05), and ms-hFHC mice had 33% stasis suggesting vascular protection by ferroxidase active wt-hFHC. The wt-hFHC SCD mice had marked increases in splenic hFHC mRNA and hepatic hFHC protein, ferritin light chain (FLC), 5-aminolevulinic acid synthase (ALAS), heme content, ferroportin, nuclear factor erythroid 2-related factor 2 (Nrf2), and HO-1 activity and protein. There was also a decrease in hepatic activated nuclear factor-kappa B (NF-κB) phospho-p65 and vascular cell adhesion molecule-1 (VCAM-1). Inhibition of HO-1 activity with tin protoporphyrin demonstrated HO-1 was not essential for the protection by wt-hFHC. We conclude that wt-hFHC ferroxidase activity enhances cytoprotective Nrf2-regulated proteins including HO-1, thereby resulting in decreased NF-κB-activation, adhesion molecules, and microvascular stasis in transgenic SCD mice.
- Published
- 2014
- Full Text
- View/download PDF
40. Not simply misshapen red cells: multimolecular and cellular events in sickle vaso-occlusion.
- Author
-
Vercellotti GM and Belcher JD
- Subjects
- Animals, Humans, Male, Neutrophils physiology, Proto-Oncogene Proteins c-akt physiology, Vasculitis physiopathology
- Abstract
Thromboinflammatory diseases result from the interactions of vascular endothelial cells, inflammatory cells, and platelets with cellular adhesion molecules, plasma proteins, and lipids. Tipping the balance toward a prothrombotic, proinflammatory phenotype results from multicellular activation signals. In this issue of the JCI, Li et al. explore the regulation of heterotypic neutrophil-platelet contacts in response to TNF-α-induced venular inflammation with relevance to sickle cell disease (SCD).
- Published
- 2014
- Full Text
- View/download PDF
41. Heme triggers TLR4 signaling leading to endothelial cell activation and vaso-occlusion in murine sickle cell disease.
- Author
-
Belcher JD, Chen C, Nguyen J, Milbauer L, Abdulla F, Alayash AI, Smith A, Nath KA, Hebbel RP, and Vercellotti GM
- Subjects
- Animals, Bone Marrow Cells cytology, Cell Adhesion, Haptoglobins metabolism, Heme chemistry, Hemoglobins chemistry, Hemolysis, Hemopexin metabolism, Human Umbilical Vein Endothelial Cells, Humans, Inflammation, Lipopolysaccharides metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, NF-kappa B p50 Subunit metabolism, Oxidative Stress, Phenotype, von Willebrand Factor metabolism, Anemia, Sickle Cell metabolism, Endothelial Cells metabolism, Signal Transduction, Toll-Like Receptor 4 metabolism, Vasoconstriction
- Abstract
Treatment of sickle cell disease (SCD) is hampered by incomplete understanding of pathways linking hemolysis to vaso-occlusion. We investigated these pathways in transgenic sickle mice. Infusion of hemoglobin or heme triggered vaso-occlusion in sickle, but not normal, mice. Methemoglobin, but not heme-stabilized cyanomethemoglobin, induced vaso-occlusion, indicating heme liberation is necessary. In corroboration, hemoglobin-induced vaso-occlusion was blocked by the methemoglobin reducing agent methylene blue, haptoglobin, or the heme-binding protein hemopexin. Untreated HbSS mice, but not HbAA mice, exhibited ∼10% vaso-occlusion in steady state that was inhibited by haptoglobin or hemopexin infusion. Antibody blockade of adhesion molecules P-selectin, von Willebrand factor (VWF), E-selectin, vascular cell adhesion molecule 1, intercellular adhesion molecule 1, platelet endothelial cell (EC) adhesion molecule 1, α4β1, or αVβ3 integrin prevented vaso-occlusion. Heme rapidly (5 minutes) mobilized Weibel-Palade body (WPB) P-selectin and VWF onto EC and vessel wall surfaces and activated EC nuclear factor κB (NF-κB). This was mediated by TLR4 as TAK-242 blocked WPB degranulation, NF-κB activation, vaso-occlusion, leukocyte rolling/adhesion, and heme lethality. TLR4(-/-) mice transplanted with TLR4(+/+) sickle bone marrow exhibited no heme-induced vaso-occlusion. The TLR4 agonist lipopolysaccharide (LPS) activated ECs and triggered vaso-occlusion that was inhibited by TAK-242, linking hemolysis- and infection-induced vaso-occlusive crises to TLR4 signaling. Heme and LPS failed to activate VWF and NF-κB in TLR4(-/-) ECs. Anti-LPS immunoglobulin G blocked LPS-induced, but not heme-induced, vaso-occlusion, illustrating LPS-independent TLR4 signaling by heme. Inhibition of protein kinase C, NADPH oxidase, or antioxidant treatment blocked heme-mediated stasis, WPB degranulation, and oxidant production. We conclude that intravascular hemolysis in SCD releases heme that activates endothelial TLR4 signaling leading to WPB degranulation, NF-κB activation, and vaso-occlusion.
- Published
- 2014
- Full Text
- View/download PDF
42. β-Globin sleeping beauty transposon reduces red blood cell sickling in a patient-derived CD34(+)-based in vitro model.
- Author
-
Sjeklocha LM, Wong PY, Belcher JD, Vercellotti GM, and Steer CJ
- Subjects
- Anemia, Sickle Cell metabolism, Bone Marrow metabolism, Bone Marrow physiology, Cell Differentiation genetics, Genetic Therapy methods, Genetic Vectors genetics, Hemoglobin, Sickle genetics, Hemoglobin, Sickle metabolism, Humans, Transgenes genetics, Anemia, Sickle Cell genetics, Anemia, Sickle Cell therapy, Antigens, CD34 metabolism, DNA Transposable Elements genetics, Erythrocytes metabolism, Erythrocytes physiology, beta-Globins genetics
- Abstract
The ultimate goal of gene therapy for sickle cell anemia (SCA) is an improved phenotype for the patient. In this study, we utilized bone marrow from a sickle cell patient as a model of disease in an in vitro setting for the hyperactive Sleeping Beauty transposon gene therapy system. We demonstrated that mature sickle red blood cells containing hemoglobin-S and sickling in response to metabisulfite can be generated in vitro from SCA bone marrow. These cells showed the characteristic morphology and kinetics of hemoglobin-S polymerization, which we quantified using video microscopy and imaging cytometry. Using video assessment, we showed that delivery of an IHK-β(T87Q) antisickling globin gene by Sleeping Beauty via nucleofection improves metrics of sickling, decreasing percent sickled from 53.2 ± 2.2% to 43.9 ± 2.0%, increasing the median time to sickling from 8.5 to 9.6 min and decreasing the maximum rate of sickling from 2.3 x 10(-3) sickling cells/total cells/sec in controls to 1.26 x 10(-3) sickling cells/total cells/sec in the IHK-β(T87Q)-globin group (p < 0.001). Using imaging cytometry, the percentage of elongated sickled cells decreased from 34.8 ± 4.5% to 29.5 ± 3.0% in control versus treated (p < 0.05). These results support the potential use of Sleeping Beauty as a clinical gene therapy vector and provide a useful tool for studying sickle red blood cells in vitro.
- Published
- 2013
- Full Text
- View/download PDF
43. MP4CO, a pegylated hemoglobin saturated with carbon monoxide, is a modulator of HO-1, inflammation, and vaso-occlusion in transgenic sickle mice.
- Author
-
Belcher JD, Young M, Chen C, Nguyen J, Burhop K, Tran P, and Vercellotti GM
- Subjects
- Anemia, Sickle Cell metabolism, Anemia, Sickle Cell mortality, Animals, Carbon Monoxide metabolism, Disease Models, Animal, Female, Guaiacol analogs & derivatives, Hemin metabolism, Hemin pharmacology, Hemoglobins metabolism, Humans, Inflammation drug therapy, Inflammation metabolism, Inflammation mortality, Male, Maleimides metabolism, Mice, Mice, Transgenic, Microcirculation drug effects, Microcirculation physiology, NF-E2-Related Factor 2 metabolism, NF-kappa B metabolism, Polyethylene Glycols metabolism, Vasculitis metabolism, Vasculitis mortality, Anemia, Sickle Cell drug therapy, Carbon Monoxide pharmacology, Heme Oxygenase-1 metabolism, Hemoglobins pharmacology, Maleimides pharmacology, Membrane Proteins metabolism, Polyethylene Glycols pharmacology, Vasculitis drug therapy
- Abstract
Transgenic sickle mice expressing β(S) hemoglobin have activated vascular endothelium in multiple organs that exhibits enhanced expression of NF-ĸB and adhesion molecules and promotes microvascular stasis in sickle, but not normal, mice in response to hypoxia/reoxygenation (H/R), or heme. Induction of heme oxygenase-1 (HO-1) or administration of its products, carbon monoxide (CO) or biliverdin, inhibits microvascular stasis in sickle mice. Infusion of human hemoglobin conjugated with polyethylene glycol and saturated with CO (MP4CO) markedly induced hepatic HO-1 activity and inhibited NF-ĸB activation and H/R-induced microvascular stasis in sickle mice. These effects were mediated by CO; saline or MP4 saturated with O2 (MP4OX) had little to no effect on H/R-induced stasis, though unmodified oxyhemoglobin exacerbated stasis. The HO-1 inhibitor, tin protoporphyrin, blocked MP4CO protection, consistent with HO-1 involvement in the protection afforded by MP4CO. MP4CO also induced nuclear factor-erythroid 2 p45-related factor 2 (Nrf2), an important transcriptional regulator of HO-1 and other antioxidant genes. In a heterozygous (hemoglobin-AS) sickle mouse model, intravenous hemin induced cardiovascular collapse and mortality within 120 minutes, which was significantly reduced by MP4CO, but not MP4OX. These data demonstrate that MP4CO induces cytoprotective Nrf2 and HO-1 and decreases NF-ĸB activation, microvascular stasis, and mortality in transgenic sickle mouse models.
- Published
- 2013
- Full Text
- View/download PDF
44. Hemolysis and free hemoglobin revisited: exploring hemoglobin and hemin scavengers as a novel class of therapeutic proteins.
- Author
-
Schaer DJ, Buehler PW, Alayash AI, Belcher JD, and Vercellotti GM
- Subjects
- Animals, Haptoglobins metabolism, Hemin metabolism, Hemoglobins metabolism, Hemopexin metabolism, Humans, Haptoglobins therapeutic use, Hematologic Diseases blood, Hematologic Diseases drug therapy, Hemolysis drug effects, Hemolysis physiology, Hemopexin therapeutic use
- Abstract
Hemolysis occurs in many hematologic and nonhematologic diseases. Extracellular hemoglobin (Hb) has been found to trigger specific pathophysiologies that are associated with adverse clinical outcomes in patients with hemolysis, such as acute and chronic vascular disease, inflammation, thrombosis, and renal impairment. Among the molecular characteristics of extracellular Hb, translocation of the molecule into the extravascular space, oxidative and nitric oxide reactions, hemin release, and molecular signaling effects of hemin appear to be the most critical. Limited clinical experience with a plasma-derived haptoglobin (Hp) product in Japan and more recent preclinical animal studies suggest that the natural Hb and the hemin-scavenger proteins Hp and hemopexin have a strong potential to neutralize the adverse physiologic effects of Hb and hemin. This includes conditions that are as diverse as RBC transfusion, sickle cell disease, sepsis, and extracorporeal circulation. This perspective reviews the principal mechanisms of Hb and hemin toxicity in different disease states, updates how the natural scavengers efficiently control these toxic moieties, and explores critical issues in the development of human plasma-derived Hp and hemopexin as therapeutics for patients with excessive intravascular hemolysis.
- Published
- 2013
- Full Text
- View/download PDF
45. Age sensitizes the kidney to heme protein-induced acute kidney injury.
- Author
-
Nath KA, Grande JP, Farrugia G, Croatt AJ, Belcher JD, Hebbel RP, Vercellotti GM, and Katusic ZS
- Subjects
- Acute Kidney Injury metabolism, Acute Kidney Injury physiopathology, Animals, Blood Urea Nitrogen, Creatinine blood, Female, Heme Oxygenase (Decyclizing) deficiency, Heme Oxygenase (Decyclizing) genetics, Heme Oxygenase (Decyclizing) metabolism, Heme Oxygenase-1 metabolism, Hemeproteins metabolism, Hemoglobins metabolism, Interleukin-6 metabolism, Kidney metabolism, Kidney Tubular Necrosis, Acute metabolism, Kidney Tubular Necrosis, Acute physiopathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Models, Animal, RNA, Messenger metabolism, Acute Kidney Injury chemically induced, Aging physiology, Hemeproteins adverse effects, Hemoglobins adverse effects, Kidney physiopathology, Kidney Tubular Necrosis, Acute chemically induced
- Abstract
Age increases the risk for ischemic acute kidney injury (AKI). We questioned whether a similar age-dependent injury occurs following exposure to hemoglobin, a known nephrotoxin. Old mice (~16 mo old), but not young mice (~6 mo old), when administered hemoglobin, exhibited marked elevation in blood urea nitrogen (BUN) and serum creatinine, and acute tubular necrosis with prominent tubular cast formation. The aged kidney exhibited induction of heme oxygenase-1 (HO-1) and other genes/proteins that may protect against heme-mediated renal injury, including ferritin, ferroportin, haptoglobin, and hemopexin. Old mice did not evince induction of HO-2 mRNA by hemoglobin, whereas a modest induction of HO-2 mRNA was observed in young mice. To determine the functional significance of HO-2 in heme protein-induced AKI, we administered hemoglobin to relatively young HO-2(+/+) and HO-2(-/-) mice: HO-2(-/-) mice, compared with HO-2(+/+) mice, exhibited greater renal dysfunction and histologic injury when administered hemoglobin. In addition to failing to elicit a protective system such as HO-2 in response to hemoglobin, old mice exhibited an exaggerated maladaptive response typified by markedly greater induction of the nephrotoxic cytokine IL-6 (130-fold increase vs. 10-fold increase in mRNA in young mice). We conclude that aged mice, unlike relatively younger mice, are exquisitely sensitive to the nephrotoxicity of hemoglobin, an effect attended by a failure to induce HO-2 mRNA and a fulminant upregulation of IL-6. Age thus markedly augments the sensitivity of the kidney to heme proteins, and HO-2 confers resistance to such insults.
- Published
- 2013
- Full Text
- View/download PDF
46. Vasculotoxic and Proinflammatory Effects of Plasma Heme: Cell Signaling and Cytoprotective Responses.
- Author
-
Belcher JD, Nath KA, and Vercellotti GM
- Abstract
The proinfammatory vasculotoxic effects of intravascular hemolysis are modulated by plasma hemoglobin and heme clearance via the haptoglobin/CD163 system and the hemopexin/CD91 system, respectively, and detoxification through the heme oxygenase/ferritin system. However, sudden or excessive hemolysis can overwhelm these protective systems leading to heme interacting with cells of the vasculature. Heme presents a damage-associated molecular pattern to the innate immune system. Heme is an extracellular inflammatory signaling molecule with strict binding specificity for TLR4 on monocyte/macrophages, endothelial, and other cells. The resulting TLR4 signaling cascade rapidly leads to intracellular oxidative stress and an inflammatory response. Heme also induces a cytoprotective response that includes Nrf2 responsive genes such as heme oxygenase-1, ferritin, haptoglobin, hemopexin, and other antioxidant response genes. It is the balance between the pro-inflammatory/vasculotoxic effects of plasma hemoglobin/heme and the cytoprotective responses that ultimately determines the pathophysiologic outcome in patients.
- Published
- 2013
- Full Text
- View/download PDF
47. Erythroid-specific expression of β-globin from Sleeping Beauty-transduced human hematopoietic progenitor cells.
- Author
-
Sjeklocha LM, Park CW, Wong PY, Roney MJ, Belcher JD, Kaufman DS, Vercellotti GM, Hebbel RP, and Steer CJ
- Subjects
- Cell Lineage, Gene Silencing, Humans, K562 Cells, RNA, Messenger genetics, Transgenes, beta-Globins genetics, Erythrocytes metabolism, Hematopoietic Stem Cells metabolism, Transposases physiology, beta-Globins metabolism
- Abstract
Gene therapy for sickle cell disease will require efficient delivery of a tightly regulated and stably expressed gene product to provide an effective therapy. In this study we utilized the non-viral Sleeping Beauty (SB) transposon system using the SB100X hyperactive transposase to transduce human cord blood CD34(+) cells with DsRed and a hybrid IHK-β-globin transgene. IHK transduced cells were successfully differentiated into multiple lineages which all showed transgene integration. The mature erythroid cells had an increased β-globin to γ-globin ratio from 0.66±0.08 to 1.05±0.12 (p=0.05), indicating expression of β-globin from the integrated SB transgene. IHK-β-globin mRNA was found in non-erythroid cell types, similar to native β-globin mRNA that was also expressed at low levels. Additional studies in the hematopoietic K562 cell line confirmed the ability of cHS4 insulator elements to protect DsRed and IHK-β-globin transgenes from silencing in long-term culture studies. Insulated transgenes had statistically significant improvement in the maintenance of long term expression, while preserving transgene regulation. These results support the use of Sleeping Beauty vectors in carrying an insulated IHK-β-globin transgene for gene therapy of sickle cell disease.
- Published
- 2011
- Full Text
- View/download PDF
48. Heme oxygenase-1 gene delivery by Sleeping Beauty inhibits vascular stasis in a murine model of sickle cell disease.
- Author
-
Belcher JD, Vineyard JV, Bruzzone CM, Chen C, Beckman JD, Nguyen J, Steer CJ, and Vercellotti GM
- Subjects
- Animals, Base Sequence, Female, Heme Oxygenase-1 metabolism, Humans, Liver cytology, Liver enzymology, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Molecular Sequence Data, Rats, Regional Blood Flow, Anemia, Sickle Cell genetics, Anemia, Sickle Cell physiopathology, Anemia, Sickle Cell therapy, Blood Vessels physiopathology, Disease Models, Animal, Gene Transfer Techniques, Genetic Therapy methods, Heme Oxygenase-1 genetics, Transposases genetics, Transposases metabolism
- Abstract
Increases in heme oxygenase-1 (HO-1) and administration of heme degradation products CO and biliverdin inhibit vascular inflammation and vasoocclusion in mouse models of sickle cell disease (SCD). In this study, an albumin (alb) promoter-driven Sleeping Beauty (SB) transposase plasmid with a wild-type rat hmox-1 (wt-HO-1) transposable element was delivered by hydrodynamic tail vein injections to SCD mice. Eight weeks after injection, SCD mice had three- to five-fold increases in HO-1 activity and protein expression in liver, similar to hemin-treated mice. Immunohistochemistry demonstrated increased perinuclear HO-1 staining in hepatocytes. Messenger RNA transcription of the hmox-1 transgene in liver was confirmed by quantitative real-time polymerase chain reaction restriction fragment length polymorphism (qRT-PCR RFLP) with no detectible transgene expression in other organs. The livers of all HO-1 overexpressing mice had activation of nuclear phospho-p38 mitogen-activated protein kinase (MAPK) and phospho-Akt, decreased nuclear expression of nuclear factor-kappa B (NF-kappaB) p65, and decreased soluble vascular cell adhesion molecule-1 (sVCAM-1) in serum. Hypoxia-induced stasis, a characteristic of SCD, but not normal mice, was inhibited in dorsal skin fold chambers in wt-HO-1 SCD mice despite the absence of hmox-1 transgene expression in the skin suggesting distal effects of HO activity on the vasculature. No protective effects were seen in SCD mice injected with nonsense (ns-) rat hmox-1 that encodes carboxy-truncated HO-1 with little or no enzyme activity. We speculate that HO-1 gene delivery to the liver is beneficial in SCD mice by degrading pro-oxidative heme, releasing anti-inflammatory heme degradation products CO and biliverdin/bilirubin into circulation, activating cytoprotective pathways and inhibiting vascular stasis at sites distal to transgene expression.
- Published
- 2010
- Full Text
- View/download PDF
49. Acadesine inhibits tissue factor induction and thrombus formation by activating the phosphoinositide 3-kinase/Akt signaling pathway.
- Author
-
Zhang W, Wang J, Wang H, Tang R, Belcher JD, Viollet B, Geng JG, Zhang C, Wu C, Slungaard A, Zhu C, and Huo Y
- Subjects
- AMP-Activated Protein Kinases antagonists & inhibitors, AMP-Activated Protein Kinases deficiency, AMP-Activated Protein Kinases genetics, Adenosine A2 Receptor Antagonists, Aminoimidazole Carboxamide pharmacology, Animals, Apolipoproteins E deficiency, Apolipoproteins E genetics, Atherosclerosis blood, Atherosclerosis drug therapy, Atherosclerosis enzymology, Blood Coagulation drug effects, Cells, Cultured, Disease Models, Animal, Dose-Response Relationship, Drug, Endothelial Cells enzymology, Enzyme Activation, Humans, Lipopolysaccharides pharmacology, Macrophages drug effects, Macrophages metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Monocytes enzymology, NF-kappa B metabolism, Protein Kinase Inhibitors pharmacology, Pyrazoles pharmacology, Pyrimidines pharmacology, RNA, Messenger metabolism, Receptor, Adenosine A2A deficiency, Receptor, Adenosine A2A genetics, Sepsis blood, Sepsis drug therapy, Sepsis enzymology, Thromboplastin genetics, Transcription Factor AP-1 metabolism, Triazines pharmacology, Triazoles pharmacology, Up-Regulation, Venous Thrombosis blood, Venous Thrombosis enzymology, Aminoimidazole Carboxamide analogs & derivatives, Endothelial Cells drug effects, Fibrinolytic Agents pharmacology, Monocytes drug effects, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Ribonucleosides pharmacology, Signal Transduction drug effects, Thromboplastin metabolism, Venous Thrombosis prevention & control
- Abstract
Objective: Acadesine, an adenosine-regulating agent and activator of AMP-activated protein kinase, has been shown to possess antiinflammatory activity. This study investigated whether and how acadesine inhibits tissue factor (TF) expression and thrombus formation., Methods and Results: Human umbilical vein endothelial cells and human peripheral blood monocytes were stimulated with lipopolysaccharide to induce TF expression. Pretreatment with acadesine dramatically suppressed the clotting activity and expression of TF (protein and mRNA). These inhibitory effects of acadesine were unchanged for endothelial cells treated with ZM241385 (a specific adenosine A(2A) receptor antagonist) or AMP-activated protein kinase inhibitor compound C, and in macrophages lacking adenosine A(2A) receptor or alpha1-AMP-activated protein kinase. In endothelial cells and macrophages, acadesine activated the phosphoinositide 3-kinase/Akt signaling pathway, reduced the activity of mitogen-activated protein kinases, and consequently suppressed TF expression by inhibiting the activator protein-1 and NF-kappaB pathways. In mice, acadesine suppressed lipopolysaccharide-mediated increases in blood coagulation, decreased TF expression in atherosclerotic lesions, and reduced deep vein thrombus formation., Conclusion: Acadesine inhibits TF expression and thrombus formation by activating the phosphoinositide 3-kinase/Akt pathway. This novel finding implicates acadesine as a potentially useful treatment for many disorders associated with thrombotic pathology, such as angina pain, deep vein thrombosis, and sepsis.
- Published
- 2010
- Full Text
- View/download PDF
50. The HDAC inhibitors trichostatin A and suberoylanilide hydroxamic acid exhibit multiple modalities of benefit for the vascular pathobiology of sickle transgenic mice.
- Author
-
Hebbel RP, Vercellotti GM, Pace BS, Solovey AN, Kollander R, Abanonu CF, Nguyen J, Vineyard JV, Belcher JD, Abdulla F, Osifuye S, Eaton JW, Kelm RJ Jr, and Slungaard A
- Subjects
- Anemia, Sickle Cell genetics, Anemia, Sickle Cell metabolism, Animals, Cells, Cultured, Disease Models, Animal, Endothelial Cells cytology, Endothelial Cells drug effects, Endothelial Cells metabolism, Enzyme Inhibitors pharmacology, Fetal Hemoglobin genetics, Hemoglobin A genetics, Hemoglobin, Sickle genetics, Humans, Intercellular Adhesion Molecule-1 metabolism, Iron Chelating Agents pharmacology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Pulmonary Veins cytology, Regional Blood Flow drug effects, Regional Blood Flow physiology, Thromboplastin metabolism, Vascular Cell Adhesion Molecule-1 metabolism, Venules cytology, Venules physiology, Vorinostat, beta-Thalassemia drug therapy, beta-Thalassemia genetics, beta-Thalassemia metabolism, Anemia, Sickle Cell drug therapy, Histone Deacetylase Inhibitors pharmacology, Hydroxamic Acids pharmacology
- Abstract
The vascular pathobiology of sickle cell anemia involves inflammation, coagulation, vascular stasis, reperfusion injury, iron-based oxidative biochemistry, deficient nitric oxide (NO) bioavailability, and red cell sickling. These disparate pathobiologies intersect and overlap, so it is probable that multimodality therapy will be necessary for this disease. We have, therefore, tested a histone deacetylase (HDAC) inhibitor, trichostatin A (TSA), for efficacy in reducing endothelial activation. We found that pulmonary vascular endothelial VCAM-1 and tissue factor (TF) expression (both are indicators of endothelial activation) are powerfully and significantly inhibited by TSA. This is seen both with pretreatment before the inducing stress of hypoxia/reoxygenation (NY1DD sickle transgenic mouse), and upon longer-term therapy after endothelial activation has already occurred (hBERK1 sickle mouse at ambient air). In addition, TSA prevented vascular stasis in sickle mice, it exhibited activity as an iron chelator, and it induced expression of the antisickling hemoglobin, hemoglobin F. Notably, the TSA analog SAHA (suberoylanilide hydroxaminc acid) that is already approved for human clinical use exhibits the same spectrum of biologic effects as TSA. We suggest that SAHA possibly could provide true, multimodality, salubrious effects for prevention and treatment of the chronic vasculopathy of sickle cell anemia.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.