1
|
Schaer DJ, Schaer CA, Humar R, Vallelian F, Henderson R, Tanaka KA, Levy JH, Buehler PW. Navigating Hemolysis, Hemoglobin Toxicity, and Its Renal Implications in Cardiac Surgery. Anesthesiology 2024:141893. [PMID: 39159287 DOI: 10.1097/aln.0000000000005109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Cardiopulmonary bypass–induced hemolysis is linked to acute kidney injury in cardiac surgery. Emerging therapies targeting cell-free hemoglobin (CFHb), like haptoglobin, nitric oxide (NO), and antioxidants, show promise in reducing kidney injury, highlighting the need for further research.
Collapse
Affiliation(s)
- Dominik J Schaer
- Department of Internal Medicine, University Hospital and University of Zurich, Zurich, Switzerland
| | - Christian A Schaer
- Institute of Anesthesiology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Rok Humar
- Department of Internal Medicine, University Hospital and University of Zurich, Zurich, Switzerland
| | - Florence Vallelian
- Department of Internal Medicine, University Hospital and University of Zurich, Zurich, Switzerland
| | - Reney Henderson
- Division of Cardiovascular Anesthesia, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Kenichi A Tanaka
- Department of Anesthesiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Jerrold H Levy
- Department of Anesthesiology, Critical Care and Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Paul W Buehler
- Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, and Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
2
|
Gomes FL, Jeong SH, Shin SR, Leijten J, Jonkheijm P. Engineering Synthetic Erythrocytes as Next-Generation Blood Substitutes. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2315879. [PMID: 39386164 PMCID: PMC11460667 DOI: 10.1002/adfm.202315879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Indexed: 10/12/2024]
Abstract
Blood scarcity is one of the main causes of healthcare disruptions worldwide, with blood shortages occurring at an alarming rate. Over the last decades, blood substitutes has aimed at reinforcing the supply of blood, with several products (e.g., hemoglobin-based oxygen carriers, perfluorocarbons) achieving a limited degree of success. Regardless, there is still no widespread solution to this problem due to persistent challenges in product safety and scalability. In this Review, we describe different advances in the field of blood substitution, particularly in the development of artificial red blood cells, otherwise known as engineered erythrocytes. We categorize the different strategies into natural, synthetic, or hybrid approaches, and discuss their potential in terms of safety and scalability. We identify synthetic engineered erythrocytes as the most powerful approach, and describe erythrocytes from a materials engineering perspective. We review their biological structure and function, as well as explore different methods of assembling a material-based cell. Specifically, we discuss how to recreate size, shape, and deformability through particle fabrication, and how to recreate the functional machinery through synthetic biology and nanotechnology. We conclude by describing the versatile nature of synthetic erythrocytes in medicine and pharmaceuticals and propose specific directions for the field of erythrocyte engineering.
Collapse
Affiliation(s)
- Francisca L Gomes
- Department of Molecules and Materials, Laboratory of Biointerface Chemistry, Faculty of Science and Technology, Technical Medical Centre and MESA+ Institute, University of Twente, Drienerlolaan 5, Enschede, 7522NB,The Netherlands
- Department of Developmental BioEngineering, Leijten Laboratory, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, Enschede, 7522NB, The Netherlands
| | - Seol-Ha Jeong
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA, 02139, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA, 02139, USA
| | - Jeroen Leijten
- Department of Developmental BioEngineering, Leijten Laboratory, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, Enschede, 7522NB, The Netherlands
| | - Pascal Jonkheijm
- Department of Molecules and Materials, Laboratory of Biointerface Chemistry, Faculty of Science and Technology, Technical Medical Centre and MESA+ Institute, University of Twente, Drienerlolaan 5, Enschede, 7522NB,The Netherlands
| |
Collapse
|
3
|
Muller CR, Williams AT, Eaker AM, Walser C, Dos Santos F, Cuddington CT, Wolfe SR, Palmer AF, Cabrales P. Novel high molecular weight polymerized hemoglobin in a non-obese model of cardiovascular and metabolic dysfunction. Biomed Pharmacother 2024; 176:116789. [PMID: 38815289 DOI: 10.1016/j.biopha.2024.116789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 06/01/2024] Open
Abstract
The widespread adoption of high-calorie, high-fat, high-sucrose diets (HFHSD) has become a global health concern, particularly due to their association with cardiovascular diseases and metabolic disorders. These comorbidities increase susceptibility to severe outcomes from viral infections and trauma, with trauma-related incidents significantly contributing to global mortality rates. This context underscores the critical need for a reliable blood supply. Recent research has focused on high molecular weight (MW) polymerized human hemoglobin (PolyhHb) as a promising alternative to red blood cells (RBCs), showing encouraging outcomes in previous studies. Given the overlap of metabolic disorders and trauma-related health issues, it is crucial to assess the potential toxicity of PolyhHb transfusions, particularly in models that represent these vulnerable populations. This study evaluated the effects of PolyhHb exchange transfusion in guinea pigs that had developed metabolic disorders due to a 12-week HFHSD regimen. The guinea pigs, underwent a 20 % blood volume exchange transfusion with either PolyhHb or the lower molecular weight polymerized bovine hemoglobin, Oxyglobin. Results revealed that both PolyhHb and Oxyglobin transfusions led to liver damage, with a more pronounced effect observed in HFHSD-fed animals. Additionally, markers of cardiac dysfunction indicated signs of cardiac injury in both the HFHSD and normal diet groups following the Oxyglobin transfusion. This study highlights how pre-existing metabolic disorders can exacerbate the potential side effects of hemoglobin-based oxygen carriers (HBOCs). Importantly, the newer generation of high MW PolyhHb showed lower cardiac toxicity compared to the earlier generation low MW PolyhHb, known as Oxyglobin, even in models with pre-existing endothelial and metabolic challenges.
Collapse
Affiliation(s)
- Cynthia R Muller
- Department of Bioengineering, University of California, San Diego, CA, USA
| | | | - Allyn M Eaker
- Department of Bioengineering, University of California, San Diego, CA, USA
| | - Cynthia Walser
- Department of Bioengineering, University of California, San Diego, CA, USA
| | - Fernando Dos Santos
- Department of Anesthesiology & Critical Care, University of California, San Diego, CA, USA
| | - Clayton T Cuddington
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Savannah R Wolfe
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Andre F Palmer
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Pedro Cabrales
- Department of Bioengineering, University of California, San Diego, CA, USA.
| |
Collapse
|
4
|
Lamb DR, Greenfield A, Thangaraju K, Setua S, Eiker G, Wang Q, Vahedi A, Khan MA, Yahya A, Cabrales P, Palmer AF, Buehler PW. The Molecular Size of Bioengineered Oxygen Carriers Determines Tissue Oxygenation in a Hypercholesterolemia Guinea Pig Model of Hemorrhagic Shock and Resuscitation. Mol Pharm 2023; 20:5739-5752. [PMID: 37843033 DOI: 10.1021/acs.molpharmaceut.3c00611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Polymerized human hemoglobin (PolyhHb) has shown promise in preclinical hemorrhagic shock settings. Different synthetic and purification schemes can control the size of PolyhHbs, yet research is lacking on the impact of polymerized hemoglobin size on tissue oxygenation following hemorrhage and resuscitation in specialized animal models that challenge their resuscitative capabilities. Pre-existing conditions that compromise the vasculature and end organs, such as the liver, may limit the effectiveness of resuscitation and exacerbate the toxicity of these molecules, which is an important but minimally explored therapeutic dimension. In this study, we compared the effective oxygen delivery of intermediate molecular weight PolyhHb (PolyhHb-B3; 500-750 kDa) to high molecular weight PolyhHb (PolyhHb-B4; 750 kDa-0.2 μm) for resuscitative effectiveness in guinea pig models subjected to hemorrhagic shock. We evaluated how the size of PolyhHb impacts hemodynamics and tissue oxygenation in normal guinea pigs and guinea pigs on an atherogenic diet. We observed that while PolyhHb-B3 and -B4 equivalently restore hemodynamic parameters of normal-dieted guinea pigs, high-fat-dieted guinea pigs resuscitated with PolyhHb-B4 have lower mean arterial pressures, impaired tissue oxygenation, and higher plasma lactate levels than those receiving PolyhHb-B3. We characterized the plasma of these animals following resuscitation and found that despite similar oxygen delivery kinetics, circulating PolyhHb-B3 and -B4 demonstrated a size-dependent increase in the plasma viscosity, consistent with impaired perfusion in the PolyhHb-B4 transfusion group. We conclude that intermediate-sized PolyhHbs (such as -B3) are ideal for further research given the effective resuscitation of hemorrhagic shock based on tissue oxygenation in hypercholesterolemic guinea pigs.
Collapse
Affiliation(s)
- Derek R Lamb
- Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, HSF III, 670 West Baltimore St., Baltimore, Maryland 21202, United States
| | - Alisyn Greenfield
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 452 CBEC, 151 West Woodruff Avenue, Columbus, Ohio 43210, United States
| | - Kiruphagaran Thangaraju
- Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, HSF III, 670 West Baltimore St., Baltimore, Maryland 21202, United States
| | - Saini Setua
- Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, HSF III, 670 West Baltimore St., Baltimore, Maryland 21202, United States
| | - Gena Eiker
- Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, HSF III, 670 West Baltimore St., Baltimore, Maryland 21202, United States
| | - Qihong Wang
- Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, HSF III, 670 West Baltimore St., Baltimore, Maryland 21202, United States
| | - Amid Vahedi
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 452 CBEC, 151 West Woodruff Avenue, Columbus, Ohio 43210, United States
| | - Mohd Asim Khan
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 452 CBEC, 151 West Woodruff Avenue, Columbus, Ohio 43210, United States
| | - Ahmad Yahya
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 452 CBEC, 151 West Woodruff Avenue, Columbus, Ohio 43210, United States
| | - Pedro Cabrales
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093-0412, United States
| | - Andre F Palmer
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 452 CBEC, 151 West Woodruff Avenue, Columbus, Ohio 43210, United States
| | - Paul W Buehler
- Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, HSF III, 670 West Baltimore St., Baltimore, Maryland 21202, United States
- Department of Pathology, University of Maryland School of Medicine, 10 S Pine St # 700A, Baltimore, Maryland 21201, United States
| |
Collapse
|
5
|
Muller CR, Courelli V, Walser C, Cuddington CT, Wolfe SR, Palmer AF, Cabrales P. Polymerized human hemoglobin with low and high oxygen affinity in trauma models. Transl Res 2023; 260:83-92. [PMID: 37268039 DOI: 10.1016/j.trsl.2023.05.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/04/2023]
Abstract
The present study aimed to compare the ability of tense (T) and relaxed (R) quaternary state polymerized human hemoglobin (PolyhHb) to restore hemodynamics after severe trauma in a rat model, and to assess their relative toxicity in a guinea pigs (GPs). To assess the efficacy of these PolyhHbs in restoring hemodynamics, Wistar rats were subjected to traumatic brain injury (TBI) followed by hemorrhagic shock (HS). Animals were separated into 3 groups based on the resuscitation solution: Whole blood, T-state or R-state PolyhHb, and followed for 2 hours after resuscitation. For toxicity evaluation, GPs were subjected to HS and the hypovolemic state was maintained for 50 minutes. Then, the GPs were divided randomly into 2 groups, and reperfused with T- or R-state PolyhHb. Rats resuscitated with blood and T-state PolyhHb had a higher recovery of MAP at 30 min after resuscitation when compared to R-state PolyhHb, demonstrating the greater ability of T-state PolyhHb to restore hemodynamics compared to R-state PolyhHb. Resuscitation with R-state PolyhHb in GPs increased markers of liver damage and inflammation, kidney injury and systemic inflammation compared to the T-state PolyhHb group. Finally, increased levels of cardiac damage markers, such as troponin were observed, indicating greater cardiac injury in GPs resuscitated with R-state PolyhHb. Therefore, our results showed that T-state PolyhHb exhibited superior efficacy in a model of TBI followed by HS in rats, and presented reduced vital organ toxicity in GPs, when compared to R-state PolyhHb.
Collapse
Affiliation(s)
- Cynthia R Muller
- Department of Bioengineering, University of California San Diego, San Diego, CA.
| | - Vasiliki Courelli
- Department of Bioengineering, University of California San Diego, San Diego, CA
| | - Cynthia Walser
- Department of Bioengineering, University of California San Diego, San Diego, CA
| | - Clayton T Cuddington
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH
| | - Savannah R Wolfe
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH
| | - Andre F Palmer
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH
| | - Pedro Cabrales
- Department of Bioengineering, University of California San Diego, San Diego, CA
| |
Collapse
|
6
|
Williams MC, Zhang X, Baek JH, D’Agnillo F. Renal glomerular and tubular responses to glutaraldehyde- polymerized human hemoglobin. Front Med (Lausanne) 2023; 10:1158359. [PMID: 37384048 PMCID: PMC10293615 DOI: 10.3389/fmed.2023.1158359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/30/2023] [Indexed: 06/30/2023] Open
Abstract
Hemoglobin-based oxygen carriers (HBOCs) are being developed as oxygen and volume replacement therapeutics, however, their molecular and cellular effects on the vasculature and different organ systems are not fully defined. Using a guinea pig transfusion model, we examined the renal glomerular and tubular responses to PolyHeme, a highly characterized glutaraldehyde-polymerized human hemoglobin with low tetrameric hemoglobin content. PolyHeme-infused animals showed no major changes in glomerular histology or loss of specific markers of glomerular podocytes (Wilms tumor 1 protein, podocin, and podocalyxin) or endothelial cells (ETS-related gene and claudin-5) after 4, 24, and 72 h. Relative to sham controls, PolyHeme-infused animals also showed similar expression and subcellular distribution of N-cadherin and E-cadherin, two key epithelial junctional proteins of proximal and distal tubules, respectively. In terms of heme catabolism and iron-handling responses, PolyHeme induced a moderate but transient expression of heme oxygenase-1 in proximal tubular epithelium and tubulointerstitial macrophages that was accompanied by increased iron deposition in tubular epithelium. Contrary to previous findings with other modified or acellular hemoglobins, the present data show that PolyHeme does not disrupt the junctional integrity of the renal glomerulus and tubular epithelium, and triggers moderate activation of heme catabolic and iron sequestration systems likely as part of a renal adaptive response.
Collapse
|
7
|
Chen N, Belcher DA, Savla C, Palmer AF, Berthiaume F. Biocompatibility of the oxygen carrier polymerized human hemoglobin towards HepG2/C3A cells. Heliyon 2023; 9:e15878. [PMID: 37215914 PMCID: PMC10192743 DOI: 10.1016/j.heliyon.2023.e15878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/24/2023] Open
Abstract
Hemoglobin (Hb) based oxygen carriers (HBOCs) are designed to minimize the toxicity of extracellular Hb, while preserving its high oxygen-carrying capacity for oxygen delivery to cells. Polymerized human Hb (PolyHb) is a novel type of nanosized HBOC synthesized via glutaraldehyde-mediated crosslinking of free Hb, and which preserves the predominant quaternary state during the crosslinking reaction (low oxygen affinity tense (T) quaternary state PolyHb is synthesized at 0% Hb oxygen saturation, and high oxygen affinity relaxed (R) quaternary state PolyHb is synthesized at 100% Hb oxygen saturation). Major potential applications for PolyHbs, and HBOCs in general, include oxygenation of bioreactor systems containing large liver cell masses, and ex-vivo perfusion preservation of explanted liver grafts. The toxicity of these compounds toward liver cells must be evaluated before testing their use in these complex systems for oxygen delivery. Herein, we characterized the effect of PolyHbs on the hepatoma cell line HepG2/C3A, used as a model hepatocyte and as a cell line used in some investigational bioartificial liver support devices. HepG2/C3A cells were incubated in cell culture media containing PolyHbs or unmodified Hb at concentrations up to 50 mg/mL and for up to 6 days. PolyHbs were well tolerated at a dose of 10 mg/mL, with no significant decrease in cell viability; however, proliferation was inhibited as much as 10-fold after 6 days of exposure at 50 mg/mL. Secretion of albumin, and urea, as well as glucose and ammonia removal were measured in presence of 10 mg/mL of PolyHbs or unmodified Hb. In addition, methoxy- and ethoxy-resorufin deacetylase (MROD and EROD) activities, which reflect cytochrome P450 metabolism, were measured. R-state PolyHb displayed improved or intact activity in 3 out of 7 functions compared to unmodified Hb. T-state PolyHb displayed improved or intact activity in 4 out of 7 functions compared to unmodified Hb. Thus, PolyHbs, both in the R-state and T-state, are safer to use at a concentration of 10 mg/mL as compared to unmodified Hb in static culture liver-related applications.
Collapse
Affiliation(s)
- Nuozhou Chen
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Donald A. Belcher
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Chintan Savla
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Andre F. Palmer
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Francois Berthiaume
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
8
|
Greenfield A, Lamb DR, Gu X, Thangaraju K, Setua S, Yahya A, Vahedi A, Khan MA, Wang Q, Buehler PW, Palmer AF. Biophysical Analysis and Preclinical Pharmacokinetics-Pharmacodynamics of Tangential Flow Filtration Fractionated Polymerized Human Hemoglobin as a Red Blood Cell Substitute. Biomacromolecules 2023; 24:1855-1870. [PMID: 36877888 DOI: 10.1021/acs.biomac.3c00051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Red blood cell (RBC) substitutes tested in late-phase clinical trials contained low-molecular-weight hemoglobin species (<500 kDa), resulting in vasoconstriction, hypertension, and oxidative tissue injury; therefore, contributing to poor clinical outcomes. This work aims to improve the safety profile of the RBC substitute, polymerized human hemoglobin (PolyhHb), via in vitro and in vivo screening of PolyhHb fractionated into four molecular weight brackets (50-300 kDa [PolyhHb-B1]; 100-500 kDa [PolyhHb-B2]; 500-750 kDa [PolyhHb-B3]; and 750 kDa to 0.2 μm [PolyhHb-B4]) using a two-stage tangential flow filtration purification process. Analysis showed that PolyhHb's oxygen affinity, and haptoglobin binding kinetics decreased with increasing bracket size. A 25% blood-for-PolyhHb exchange transfusion guinea pig model suggests that hypertension and tissue extravasation decreased with increasing bracket size. PolyhHb-B3 demonstrated extended circulatory pharmacokinetics, no renal tissue distribution, no aberrant blood pressure, or cardiac conduction effects, and may therefore be appropriate material for further evaluation.
Collapse
Affiliation(s)
- Alisyn Greenfield
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 452 CBEC, 151 West Woodruff Avenue, Columbus, Ohio 43210, United States
| | - Derek R Lamb
- The Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, The University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Xiangming Gu
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 452 CBEC, 151 West Woodruff Avenue, Columbus, Ohio 43210, United States
| | - Kiruphagaran Thangaraju
- The Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, The University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Saini Setua
- The Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, The University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Ahmad Yahya
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 452 CBEC, 151 West Woodruff Avenue, Columbus, Ohio 43210, United States
| | - Amid Vahedi
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 452 CBEC, 151 West Woodruff Avenue, Columbus, Ohio 43210, United States
| | - Mohd Asim Khan
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 452 CBEC, 151 West Woodruff Avenue, Columbus, Ohio 43210, United States
| | - Qihong Wang
- The Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, The University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Paul W Buehler
- The Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, The University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- Department of Pathology, The University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Andre F Palmer
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 452 CBEC, 151 West Woodruff Avenue, Columbus, Ohio 43210, United States
| |
Collapse
|
9
|
Luo Z, Sun L, Bian F, Wang Y, Yu Y, Gu Z, Zhao Y. Erythrocyte-Inspired Functional Materials for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206150. [PMID: 36581585 PMCID: PMC9951328 DOI: 10.1002/advs.202206150] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/03/2022] [Indexed: 05/30/2023]
Abstract
Erythrocytes are the most abundant cells in the blood. As the results of long-term natural selection, their specific biconcave discoid morphology and cellular composition are responsible for gaining excellent biological performance. Inspired by the intrinsic features of erythrocytes, various artificial biomaterials emerge and find broad prospects in biomedical applications such as therapeutic delivery, bioimaging, and tissue engineering. Here, a comprehensive review from the fabrication to the applications of erythrocyte-inspired functional materials is given. After summarizing the biomaterials mimicking the biological functions of erythrocytes, the synthesis strategies of particles with erythrocyte-inspired morphologies are presented. The emphasis is on practical biomedical applications of these bioinspired functional materials. The perspectives for the future possibilities of the advanced erythrocyte-inspired biomaterials are also discussed. It is hoped that the summary of existing studies can inspire researchers to develop novel biomaterials; thus, accelerating the progress of these biomaterials toward clinical biomedical applications.
Collapse
Affiliation(s)
- Zhiqiang Luo
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Lingyu Sun
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Feika Bian
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yu Wang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yunru Yu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhou325001China
| | - Zhuxiao Gu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhou325001China
| |
Collapse
|
10
|
Alayash AI. Oxidation reactions of cellular and acellular hemoglobins: Implications for human health. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:1068972. [DOI: 10.3389/fmedt.2022.1068972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 10/31/2022] [Indexed: 11/29/2022] Open
Abstract
Oxygen reversibly binds to the redox active iron, a transition metal in human Hemoglobin (Hb), which subsequently undergoes oxidation in air. This process is akin to iron rusting in non-biological systems. This results in the formation of non-oxygen carrying methemoglobin (ferric) (Fe3+) and reactive oxygen species (ROS). In circulating red blood cells (RBCs), Hb remains largely in the ferrous functional form (HbF2+) throughout the RBC's lifespan due to the presence of effective enzymatic and non-enzymatic proteins that keep the levels of metHb to a minimum (1%–3%). In biological systems Hb is viewed as a Fenton reagent where oxidative toxicity is attributed to the formation of a highly reactive hydroxyl radical (OH•) generated by the reaction between Hb's iron (Fe2+) and hydrogen peroxide (H2O2). However, recent research on both cellular and acellular Hbs revealed that the protein engages in enzymatic-like activity when challenged with H2O2, resulting in the formation of a highly reactive ferryl heme (Fe4+) that can target other biological molecules before it self-destructs. Accumulating evidence from several in vitro and in vivo studies are summarized in this review to show that Hb's pseudoperoxidase activity is physiologically more dominant than the Fenton reaction and it plays a pivotal role in the pathophysiology of several blood disorders, storage lesions associated with old blood, and in the toxicity associated with the infusion of Hb-derived oxygen therapeutics.
Collapse
|
11
|
Muller CR, Williams AT, Walser C, Eaker AM, Sandoval JL, Cuddington CT, Wolfe SR, Palmer AF, Cabrales P. Safety and efficacy of human polymerized hemoglobin on guinea pig resuscitation from hemorrhagic shock. Sci Rep 2022; 12:20480. [PMID: 36443351 PMCID: PMC9703428 DOI: 10.1038/s41598-022-23926-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 11/07/2022] [Indexed: 11/29/2022] Open
Abstract
For the past thirty years, hemoglobin-based oxygen carriers (HBOCs) have been under development as a red blood cell substitute. Side-effects such as vasoconstriction, oxidative injury, and cardiac toxicity have prevented clinical approval of HBOCs. Recently, high molecular weight (MW) polymerized human hemoglobin (PolyhHb) has shown positive results in rats. Studies have demonstrated that high MW PolyhHb increased O2 delivery, with minimal effects on blood pressure, without vasoconstriction, and devoid of toxicity. In this study, we used guinea pigs to evaluate the efficacy and safety of high MW PolyhHb, since like humans guinea pigs cannot produce endogenous ascorbic acid, which limits the capacity of both species to deal with oxidative stress. Hence, this study evaluated the efficacy and safety of resuscitation from severe hemorrhagic shock with high MW PolyhHb, fresh blood, and blood stored for 2 weeks. Animals were randomly assigned to each experimental group, and hemorrhage was induced by the withdrawal of 40% of the blood volume (BV, estimated as 7.5% of body weight) from the carotid artery catheter. Hypovolemic shock was maintained for 50 min. Resuscitation was implemented by infusing 25% of the animal's BV with the different treatments. Hemodynamics, blood gases, total hemoglobin, and lactate were not different before hemorrhage and during shock between groups. The hematocrit was lower for the PolyhHb group compared to the fresh and stored blood groups after resuscitation. Resuscitation with stored blood had lower blood pressure compared to fresh blood at 2 h. There was no difference in mean arterial pressure between groups at 24 h. Resuscitation with PolyhHb was not different from fresh blood for most parameters. Resuscitation with PolyhHb did not show any remarkable change in liver injury, inflammation, or cardiac damage. Resuscitation with stored blood showed changes in liver function and inflammation, but no kidney injury or systemic inflammation. Resuscitation with stored blood after 24 h displayed sympathetic hyper-activation and signs of cardiac injury. These results suggest that PolyhHb is an effective resuscitation alternative to blood. The decreased toxicities in terms of cardiac injury markers, vital organ function, and inflammation following PolyhHb resuscitation in guinea pigs indicate a favorable safety profile. These results are promising and support future studies with this new generation of PolyhHb as alternative to blood when blood is unavailable.
Collapse
Affiliation(s)
- Cynthia R Muller
- Department of Bioengineering, University of California, 0412, 9500 Gilman Dr. La Jolla, San Diego, CA, 92093-0412, USA
| | - Alexander T Williams
- Department of Bioengineering, University of California, 0412, 9500 Gilman Dr. La Jolla, San Diego, CA, 92093-0412, USA
| | - Cynthia Walser
- Department of Bioengineering, University of California, 0412, 9500 Gilman Dr. La Jolla, San Diego, CA, 92093-0412, USA
| | - Allyn M Eaker
- Department of Bioengineering, University of California, 0412, 9500 Gilman Dr. La Jolla, San Diego, CA, 92093-0412, USA
| | - Jose Luis Sandoval
- Department of Bioengineering, University of California, 0412, 9500 Gilman Dr. La Jolla, San Diego, CA, 92093-0412, USA
| | - Clayton T Cuddington
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Savannah R Wolfe
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Andre F Palmer
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Pedro Cabrales
- Department of Bioengineering, University of California, 0412, 9500 Gilman Dr. La Jolla, San Diego, CA, 92093-0412, USA.
| |
Collapse
|
12
|
Drvenica IT, Stančić AZ, Maslovarić IS, Trivanović DI, Ilić VL. Extracellular Hemoglobin: Modulation of Cellular Functions and Pathophysiological Effects. Biomolecules 2022; 12:1708. [PMID: 36421721 PMCID: PMC9688122 DOI: 10.3390/biom12111708] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 08/05/2023] Open
Abstract
Hemoglobin is essential for maintaining cellular bioenergetic homeostasis through its ability to bind and transport oxygen to the tissues. Besides its ability to transport oxygen, hemoglobin within erythrocytes plays an important role in cellular signaling and modulation of the inflammatory response either directly by binding gas molecules (NO, CO, and CO2) or indirectly by acting as their source. Once hemoglobin reaches the extracellular environment, it acquires several secondary functions affecting surrounding cells and tissues. By modulating the cell functions, this macromolecule becomes involved in the etiology and pathophysiology of various diseases. The up-to-date results disclose the impact of extracellular hemoglobin on (i) redox status, (ii) inflammatory state of cells, (iii) proliferation and chemotaxis, (iv) mitochondrial dynamic, (v) chemoresistance and (vi) differentiation. This review pays special attention to applied biomedical research and the use of non-vertebrate and vertebrate extracellular hemoglobin as a promising candidate for hemoglobin-based oxygen carriers, as well as cell culture medium additive. Although recent experimental settings have some limitations, they provide additional insight into the modulatory activity of extracellular hemoglobin in various cellular microenvironments, such as stem or tumor cells niches.
Collapse
Affiliation(s)
- Ivana T. Drvenica
- Group for Immunology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia
| | - Ana Z. Stančić
- Group for Immunology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia
| | - Irina S. Maslovarić
- Group for Immunology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia
| | - Drenka I. Trivanović
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia
| | - Vesna Lj. Ilić
- Group for Immunology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia
| |
Collapse
|
13
|
Scalable production and complete biophysical characterization of poly(ethylene glycol) surface conjugated liposome encapsulated hemoglobin (PEG-LEH). PLoS One 2022; 17:e0269939. [PMID: 35802716 PMCID: PMC9269976 DOI: 10.1371/journal.pone.0269939] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/31/2022] [Indexed: 11/19/2022] Open
Abstract
Particle encapsulated hemoglobin (Hb)-based oxygen (O2) carriers (HBOCs) have clear advantages over their acellular counterparts because of their larger molecular diameter and lack of vasoactivity upon transfusion. Poly(ethylene glycol) surface conjugated liposome encapsulated Hb (PEG-LEH) nanoparticles are considered a promising class of HBOC for use as a red blood cell (RBC) substitute. However, their widespread usage is limited by manufacturing processes which prevent material scale up. In this study, PEG-LEH nanoparticles were produced via a scalable and robust process using a high-pressure cell disruptor, and their biophysical properties were thoroughly characterized. Hb encapsulation, methemoglobin (metHb) level, O2-PEG-LEH equilibria, PEG-LEH gaseous (oxygen, carbon monoxide, nitric oxide) ligand binding/release kinetics, lipocrit, and long-term storage stability allowed us to examine their potential suitability and efficacy as an RBC replacement. Our results demonstrate that PEG-LEH nanoparticle suspensions manufactured via a high-pressure cell disruptor have Hb concentrations comparable to whole blood (~12 g/dL) and possess other desirable characteristics, which may permit their use as potential lifesaving O2 therapeutics.
Collapse
|
14
|
Alayash AI. Hemoglobin Oxidation Reactions in Stored Blood. Antioxidants (Basel) 2022; 11:antiox11040747. [PMID: 35453432 PMCID: PMC9027219 DOI: 10.3390/antiox11040747] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/02/2022] [Accepted: 04/04/2022] [Indexed: 02/01/2023] Open
Abstract
Hemoglobin (Hb) inside and outside the red blood cells (RBCs) undergoes constant transformation to an oxidized form in a process known as autoxidation. The ferrous heme iron (Fe2+) of the prosthetic group is spontaneously transformed into an oxidized ferric (Fe3+) form, but under oxidative stress conditions a higher oxidation ferryl heme (Fe4+) is also formed. Although Fe3+ is a non-functional form of Hb, the Fe4+ is also extremely reactive towards other biological molecules due to its high redox potential. The RBC contains an effective reductive machinery that maintains Hb in the functional form with little oxidation during its life span. The redox transformation of Hb occurs to a lesser extent in young RBCs; it may, however, have detrimental effects on the integrity of these cells during ex vivo storage or when RBCs are subjected to pathogen reduction processes. In this review, Hb oxidation reactions (“oxidative lesion”) will be described, including details of how these reactions might impact the clinical use of stored or processed blood for therapeutic purposes.
Collapse
Affiliation(s)
- Abdu I Alayash
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices (DBCD), Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), Silver Spring, MD 20993, USA
| |
Collapse
|
15
|
Alayash AI. The Impact of COVID-19 Infection on Oxygen Homeostasis: A Molecular Perspective. Front Physiol 2021; 12:711976. [PMID: 34690793 PMCID: PMC8532809 DOI: 10.3389/fphys.2021.711976] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 08/27/2021] [Indexed: 12/14/2022] Open
Abstract
The novel coronavirus (2019-nCoV/SARS-CoV-2) causes respiratory symptoms including a substantial pulmonary dysfunction with worsening arterial hypoxemia (low blood oxygenation), eventually leading to acute respiratory distress syndrome (ARDS). The impact of the viral infection on blood oxygenation and other elements of oxygen homeostasis, such as oxygen sensing and respiratory mitochondrial mechanisms, are not well understood. As a step toward understanding these mechanisms in the context of COVID-19, recent experiments revealed contradictory data on the impact of COVID-19 infection on red blood cells (RBCs) oxygenation parameters. However, structural protein damage and membrane lipid remodeling in RBCs from COVID-19 patients that may impact RBC function have been reported. Moreover, COVID-19 infection could potentially disrupt one, if not all, of the other major pathways of homeostasis. Understanding the nature of the crosstalk among normal homeostatic pathways; oxygen carrying, oxygen sensing (i.e., hypoxia inducible factor, HIF) proteins, and the mitochondrial respiratory machinery may provide a target for therapeutic interventions.
Collapse
Affiliation(s)
- Abdu I Alayash
- Division of Blood and Devices (DBCD), United States Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
16
|
The Providence Mutation (βK82D) in Human Hemoglobin Substantially Reduces βCysteine 93 Oxidation and Oxidative Stress in Endothelial Cells. Int J Mol Sci 2020; 21:ijms21249453. [PMID: 33322551 PMCID: PMC7763657 DOI: 10.3390/ijms21249453] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 01/17/2023] Open
Abstract
The highly toxic oxidative transformation of hemoglobin (Hb) to the ferryl state (HbFe4+) is known to occur in both in vitro and in vivo settings. We recently constructed oxidatively stable human Hbs, based on the Hb Providence (βK82D) mutation in sickle cell Hb (βE6V/βK82D) and in a recombinant crosslinked Hb (rHb0.1/βK82D). Using High Resolution Accurate Mass (HRAM) mass spectrometry, we first quantified the degree of irreversible oxidation of βCys93 in these proteins, induced by hydrogen peroxide (H2O2), and compared it to their respective controls (HbA and HbS). Both Hbs containing the βK82D mutation showed considerably less cysteic acid formation, a byproduct of cysteine irreversible oxidation. Next, we performed a novel study aimed at exploring the impact of introducing βK82D containing Hbs on vascular endothelial redox homeostasis and energy metabolism. Incubation of the mutants carrying βK82D with endothelial cells resulted in altered bioenergetic function, by improving basal cellular glycolysis and glycolytic capacity. Treatment of cells with Hb variants containing βK82D resulted in lower heme oxygenase-1 and ferritin expressions, compared to native Hbs. We conclude that the presence of βK82D confers oxidative stability to Hb and adds significant resistance to oxidative toxicity. Therefore, we propose that βK82D is a potential gene-editing target in the treatment of sickle cell disease and in the design of safe and effective oxygen therapeutics.
Collapse
|
17
|
Muller CR, Williams AT, Munoz CJ, Eaker AM, Breton AN, Palmer AF, Cabrales P. Safety profile of high molecular weight polymerized hemoglobins. Transfusion 2020; 61:212-224. [PMID: 33104250 DOI: 10.1111/trf.16157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Hemoglobin (Hb)-based oxygen (O2 ) carriers (HBOCs) are being developed as alternatives to red blood cells and blood when these products are unavailable. Clinical trials of previous HBOC generations revealed side effects, including hypertension and vasoconstriction, that were not observed in preclinical studies. Large molecular weight (MW) polymerized bovine Hb (PolybHb) represents a new class of HBOC with promising results. We evaluated the safety profile of PolybHb after an exchange transfusion (ET) in guinea pigs (GPs). This study compares changes in indices of cardiac, inflammatory, and organ function after ET with high (R-state) and low (T-state) O2 affinity PolybHb with high MW. STUDY DESIGN AND METHODS Guinea pigs underwent a 20% ET with PolybHb. To assess the implication of PolybHb ET on the microcirculation, hamsters instrumented with a dorsal window chamber were subjected to a similar volume ET. RESULTS T and R-state PolybHb did not induce significant alterations in cardiac function. T-state PolybHb induced mild vasoconstriction shortly after transfusion, while R-state did not have acute effects on microvascular tone. CONCLUSION Large MW PolybHbs were found to be safe and efficacious in increasing O2 carrying capacity and the O2 affinity of the PolybHb did not affect O2 delivery or extraction by tissues in relevant preclinical models. In conclusion, these results suggest that both T-state and R-state PolybHb are safe and do not impair O2 delivery. The results are encouraging and support further evaluation of high MW PolybHbs and their future feasibility compared to allogenic blood in a trauma model.
Collapse
Affiliation(s)
- Cynthia R Muller
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Alexander T Williams
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Carlos J Munoz
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Allyn M Eaker
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Amanda N Breton
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Andre F Palmer
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Pedro Cabrales
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
18
|
Abstract
Several adverse events have been associated with the infusion of hemoglobin-based oxygen carriers (HBOCs), including transient hypertension, gastrointestinal, pancreatic/liver enzyme elevation, and cardiac/renal injury in humans. Although several mechanisms have been suggested, the basis of HBOC toxicity is still poorly understood. Scavenging of vascular endothelial nitric oxide (NO) and heme-mediated oxidative side reactions are thought to be the major causes of toxicity. However, based on more recent preclinical studies, oxidative pathways (driven by the heme prosthetic group) seem to play a more prominent role in the overall toxicity of free Hb or HBOCs. HBOCs display a diversity of physicochemical properties, including molecular size/cross-linking characteristics leading to differences in oxygen affinity, allosteric, redox properties, and even oxidative inactivation by protein/heme clearing mechanisms. These diverse characteristics can therefore be manipulated independently, leaving open the possibility of engineering a safe and effective HBOC. To date, several antioxidative strategies have been proposed to counteract the redox side reactions of current generation HBOCs.
Collapse
|
19
|
Seno S, Wang J, Cao S, Saraswati M, Park S, Simoni J, Ma L, Soltys B, Hsia CJC, Koehler RC, Robertson CL. Resuscitation with macromolecular superoxide dismutase/catalase mimetic polynitroxylated PEGylated hemoglobin offers neuroprotection in guinea pigs after traumatic brain injury combined with hemorrhage shock. BMC Neurosci 2020; 21:22. [PMID: 32404052 PMCID: PMC7222507 DOI: 10.1186/s12868-020-00571-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 05/09/2020] [Indexed: 12/21/2022] Open
Abstract
Background Polynitroxylated PEGylated hemoglobin (PNPH, aka SanFlow) possesses superoxide dismutase/catalase mimetic activities that may directly protect the brain from oxidative stress. Stabilization of PNPH with bound carbon monoxide prevents methemoglobin formation during storage and permits it to serve as a carbon monoxide donor. We determined whether small volume transfusion of hyperoncotic PNPH is neuroprotective in a polytrauma model of traumatic brain injury (TBI) plus hemorrhagic shock. Guinea pigs were used because, like humans, they do not synthesize their own ascorbic acid, which is important in reducing methemoglobin. Results TBI was produced by controlled cortical impact and was followed by 20 mL/kg hemorrhage to a mean arterial pressure (MAP) of 40 mmHg. At 90 min, animals were resuscitated with 20 mL/kg lactated Ringer’s solution or 10 mL/kg PNPH. Resuscitation with PNPH significantly augmented the early recovery of MAP after hemorrhagic shock by 10–18 mmHg; whole blood methemoglobin was only 1% higher and carboxyhemoglobin was 2% higher. At 9 days of recovery, unbiased stereology analysis revealed that, compared to animals resuscitated with lactated Ringer’s solution, those treated with PNPH had significantly more viable neurons in the hippocampus CA1 + 2 region (59 ± 10% versus 87 ± 18% of sham and naïve mean value) and in the dentate gyrus (70 ± 21% versus 96 ± 24%; n = 12 per group). Conclusion PNPH may serve as a small-volume resuscitation fluid for polytrauma involving TBI and hemorrhagic shock. The neuroprotection afforded by PNPH seen in other species was sustained in a species without endogenous ascorbic acid synthesis, thereby supporting potential translatability for human use.
Collapse
Affiliation(s)
- Soichiro Seno
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, 600 North Wolfe Street, Blalock 1404, Baltimore, MD, USA.,Division of Traumatology, Research Institute, National Defense Medical College, Saitama, Japan
| | - Jun Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, 600 North Wolfe Street, Blalock 1404, Baltimore, MD, USA
| | - Suyi Cao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, 600 North Wolfe Street, Blalock 1404, Baltimore, MD, USA
| | - Manda Saraswati
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, 600 North Wolfe Street, Blalock 1404, Baltimore, MD, USA
| | - Sharon Park
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, 600 North Wolfe Street, Blalock 1404, Baltimore, MD, USA
| | - Jan Simoni
- AntiRadical Therapeutics, Sioux Falls, SD, USA
| | - Li Ma
- Department of Physics, Georgia Southern University, Statesboro, GA, USA
| | | | | | - Raymond C Koehler
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, 600 North Wolfe Street, Blalock 1404, Baltimore, MD, USA.
| | - Courtney L Robertson
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, 600 North Wolfe Street, Blalock 1404, Baltimore, MD, USA
| |
Collapse
|
20
|
Huang B, Chen S, Pei W, Xu Y, Jiang Z, Niu C, Wang L. Oxygen-Sufficient Nanoplatform for Chemo-Sonodynamic Therapy of Hypoxic Tumors. Front Chem 2020; 8:358. [PMID: 32411675 PMCID: PMC7199163 DOI: 10.3389/fchem.2020.00358] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022] Open
Abstract
Modulation of hypoxia is an essential factor for enhancing the effects of antitumor therapies, especially sonodynamic therapy and chemotherapy. To improve the efficacy of combination therapy by reversing the hypoxic tumor microenvironment, we developed shell-core structured PPID-NPs, which were designed with a polymer shell onto the sonosensitizer and a chemotherapeutic drug were loaded and a perfluorocarbon core loaded with oxygen. The perfluorocarbon core provides sufficient oxygen not only for causing the sonosensitizer to produce more singlet oxygen to induce cell apoptosis but also for reducing drug resistance to enhance therapeutic efficacy. Furthermore, the release of chemotherapeutic drugs at the tumor site can be controlled. Thus, PPID-NPs can efficiently inhibit the growth of breast cancer by synergistic therapy under ultrasound exposure. We believe that our oxygen-sufficient nanoplatform could be an ideal therapeutic system for hypoxic tumors.
Collapse
Affiliation(s)
- Biying Huang
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Sijie Chen
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wenjing Pei
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yan Xu
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zichao Jiang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Chengcheng Niu
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Long Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
21
|
Williams AT, Muller CR, Eaker AM, Belcher DA, Bolden-Rush C, Palmer AF, Cabrales P. Polymerized Hemoglobin With Increased Molecular Size Reduces Toxicity in Healthy Guinea Pigs. ACS APPLIED BIO MATERIALS 2020; 3:2976-2985. [DOI: 10.1021/acsabm.0c00039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Alexander T. Williams
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Cynthia R. Muller
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Allyn M. Eaker
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Donald A. Belcher
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Crystal Bolden-Rush
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Andre F. Palmer
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Pedro Cabrales
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
22
|
Jungner Å, Vallius Kvist S, Romantsik O, Bruschettini M, Ekström C, Bendix I, Herz J, Felderhoff-Mueser U, Bibic A, In Apos T Zandt R, Gram M, Ley D. White Matter Brain Development after Exposure to Circulating Cell-Free Hemoglobin and Hyperoxia in a Rat Pup Model. Dev Neurosci 2020; 41:234-246. [PMID: 31991415 DOI: 10.1159/000505206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 12/03/2019] [Indexed: 12/19/2022] Open
Abstract
Neonates born with critical congenital heart defects are at risk of diffuse white matter injuries and neurodevelopmental impairments. This study aimed to determine the impact of circulating cell-free hemoglobin and hyperoxia, both present during cardiopulmonary bypass circulation, on white matter brain development. Postnatal day 6 rat pups were injected intraperitoneally with cell-free Hb or vehicle and exposed to hyperoxia (fiO2 = 0.8) or normoxia (fiO2 = 0.21) for 24 h. We evaluated apoptosis, myelination, and oligodendrocyte maturation with immunohistochemistry, gene and protein analyses, and in vivo diffusion tensor magnetic resonance imaging (MRI). Consistent with previous studies, we found an increase in apoptosis of oligodendrocytes as determined by TUNEL+ staining in Olig2+ cells in white matter, cortex, thalamus, and hippocampus following exposure to hyperoxia with no additional effect of cell-free Hb. A transient increase in the mRNA expression of intercellular adhesion molecule 1 at 6 h was observed following combined exposure to cell-free Hb and hyperoxia. No indications of oligodendrocyte maturational delay or hypomyelination were observed after either insult, delivered separately or combined, as determined by immunohistochemistry, Western blot, and diffusion tensor MRI. In our model, exposure to circulatory cell-free Hb, with or without concomitant hyperoxia, did not significantly alter brain white matter development.
Collapse
Affiliation(s)
- Åsa Jungner
- Department of Clinical Sciences Lund, Pediatric Surgery and Neonatal Care, Skane University Hospital, Lund University, Lund, Sweden,
| | - Suvi Vallius Kvist
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Lund, Sweden
| | - Olga Romantsik
- Department of Clinical Sciences Lund, Pediatric Surgery and Neonatal Care, Skane University Hospital, Lund University, Lund, Sweden
| | - Matteo Bruschettini
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Lund, Sweden
| | - Claes Ekström
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Lund, Sweden
| | - Ivo Bendix
- Department of Pediatrics I, Neonatology and Experimental Perinatal Neuroscience, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Josephine Herz
- Department of Pediatrics I, Neonatology and Experimental Perinatal Neuroscience, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ursula Felderhoff-Mueser
- Department of Pediatrics I, Neonatology and Experimental Perinatal Neuroscience, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Adnan Bibic
- Lund University Bioimaging Center, Lund University, Lund, Sweden
| | | | - Magnus Gram
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Lund, Sweden
| | - David Ley
- Department of Clinical Sciences Lund, Pediatric Surgery and Neonatal Care, Skane University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
23
|
Baek JH, Yalamanoglu A, Brown RP, Saylor DM, Malinauskas RA, Buehler PW. Renal Toxicodynamic Effects of Extracellular Hemoglobin After Acute Exposure. Toxicol Sci 2019; 166:180-191. [PMID: 30085279 DOI: 10.1093/toxsci/kfy193] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Plasma hemoglobin (Hb) is elevated in some hematologic disease states, during exposures to certain toxicants, and with the use of some medical devices. Exposure to free Hb can precipitate oxidative reactions within tissues and alter the normal physiological function of critical organ systems. As kidney structures can be highly sensitive to Hb exposures, we evaluated the acute dose dependent renal toxicologic response to purified Hb isolated from RBCs. Male Hartley guinea pigs (n = 5 per group) were dosed with 0.9% saline (2 ml), 15, 75, 150, or 300 mg of purified Hb, infused over a 2-h period. The primary endpoints of this study were to define toxicokinetic parameters after increasing doses of purified Hb, identify clinically recognized and experimental markers of acute kidney injury (AKI), and determine relevant toxicological parameters and potential causes of renal toxicity in this model. Experimental findings demonstrated a dose dependent increase in Cmax after a 2-h infusion, which correlated with an elevation in serum creatinine, renal Kim-1 mRNA expression and increased urinary Kim-1. Renal NGAL mRNA expression and urinary NGAL excretion were also increased in several groups, but these parameters did not correlate with exposure. Iron increased in the renal cortex as Hb exposure increased and its deposition colocalized with 4-hydroxy-nonenal and 8-Oxo-2-deoxyguanosine immune reactivity, suggesting oxidative stressors may contribute to AKI in animals exposed to Hb. The results presented here suggest that Cmax may effectively predict the risk of AKI in normal healthy animals exposed to Hb.
Collapse
Affiliation(s)
| | | | - Ronald P Brown
- Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Maryland 20993
| | - David M Saylor
- Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Maryland 20993
| | - Richard A Malinauskas
- Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Maryland 20993
| | | |
Collapse
|
24
|
Edmondson M, Jana S, Meng F, Strader MB, Baek JH, Gao Y, Buehler PW, Alayash AI. Redox states of hemoglobin determine left ventricle pressure recovery and activity of mitochondrial complex IV in hypoxic rat hearts. Free Radic Biol Med 2019; 141:348-361. [PMID: 31302228 DOI: 10.1016/j.freeradbiomed.2019.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/03/2019] [Accepted: 07/10/2019] [Indexed: 11/30/2022]
Abstract
Cardiovascular effects were reported to occur in humans and in animal models during transfusion with hemoglobin (Hb)-based oxygen therapeutics. The effects of Hb's iron redox states on cardiac parameters during hypoxia/reoxygenation are however poorly defined. We hypothesize that acute exposures to ferric Hb during hypoxia leads to cardiomyocyte injury and an impaired left ventricular response accompanied by cardiac mitochondrial bioenergetic dysfunction. Recovery of left ventricular functions in an isolated rat heart Langendorff perfusion system was observed following perfusion with ferrous but not with ferric Hb. Ferric Hb induced the development of heart lesions, and impairment of the respiratory chain complex activity. Under normoxia, a sharp decline in cardiac parameters was observed following co-perfusion of low (20 μM) and high (100 μM) ascorbic acid (Asc) with ferrous Hb. This trend continued with ferric Hb co-perfusion, but only at the higher concentration of Asc. These observations suggest that perfusion of the hypoxic heart with ferric Hb increases oxidative stress thereby resulting in cardiac dysfunction. Intervention with Asc to reduce ferric Hb may offer a strategy to control Hb toxicity; however, timing of administration, and dosage of Asc may require individual optimization to target specific redox forms of Hb.
Collapse
Affiliation(s)
- Makhosazane Edmondson
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Maryland, USA
| | - Sirsendu Jana
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Maryland, USA
| | - Fantao Meng
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Maryland, USA
| | - Michael Brad Strader
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Maryland, USA
| | - Jin Hyen Baek
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Maryland, USA
| | - Yamei Gao
- Laboratory of Pediatric and Respiratory Viral Disease, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Silver Spring, MD, USA
| | - Paul W Buehler
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Maryland, USA
| | - Abdu I Alayash
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Maryland, USA.
| |
Collapse
|
25
|
Cooper CE, Silkstone GGA, Simons M, Rajagopal B, Syrett N, Shaik T, Gretton S, Welbourn E, Bülow L, Eriksson NL, Ronda L, Mozzarelli A, Eke A, Mathe D, Reeder BJ. Engineering tyrosine residues into hemoglobin enhances heme reduction, decreases oxidative stress and increases vascular retention of a hemoglobin based blood substitute. Free Radic Biol Med 2019; 134:106-118. [PMID: 30594736 PMCID: PMC6597946 DOI: 10.1016/j.freeradbiomed.2018.12.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 11/22/2018] [Accepted: 12/24/2018] [Indexed: 01/29/2023]
Abstract
Hemoglobin (Hb)-based oxygen carriers (HBOC) are modified extracellular proteins, designed to replace or augment the oxygen-carrying capacity of erythrocytes. However, clinical results have generally been disappointing due to adverse side effects, in part linked to the intrinsic oxidative toxicity of Hb. Previously a redox-active tyrosine residue was engineered into the Hb β subunit (βF41Y) to facilitate electron transfer between endogenous antioxidants such as ascorbate and the oxidative ferryl heme species, converting the highly oxidizing ferryl species into the less reactive ferric (met) form. We inserted different single tyrosine mutations into the α and β subunits of Hb to determine if this effect of βF41Y was unique. Every mutation that was inserted within electron transfer range of the protein surface and the heme increased the rate of ferryl reduction. However, surprisingly, three of the mutations (βT84Y, αL91Y and βF85Y) also increased the rate of ascorbate reduction of ferric(met) Hb to ferrous(oxy) Hb. The rate enhancement was most evident at ascorbate concentrations equivalent to that found in plasma (< 100 μM), suggesting that it might be of benefit in decreasing oxidative stress in vivo. The most promising mutant (βT84Y) was stable with no increase in autoxidation or heme loss. A decrease in membrane damage following Hb addition to HEK cells correlated with the ability of βT84Y to maintain the protein in its oxygenated form. When PEGylated and injected into mice, βT84Y was shown to have an increased vascular half time compared to wild type PEGylated Hb. βT84Y represents a new class of mutations with the ability to enhance reduction of both ferryl and ferric Hb, and thus has potential to decrease adverse side effects as one component of a final HBOC product.
Collapse
Affiliation(s)
- Chris E Cooper
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, United Kingdom.
| | - Gary G A Silkstone
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, United Kingdom
| | - Michelle Simons
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, United Kingdom
| | - Badri Rajagopal
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, United Kingdom
| | - Natalie Syrett
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, United Kingdom
| | - Thoufieq Shaik
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, United Kingdom
| | - Svetlana Gretton
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, United Kingdom
| | - Elizabeth Welbourn
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, United Kingdom
| | - Leif Bülow
- Pure and Applied Biochemistry, Department of Chemistry, Lund University, Box 124, 221 00 Lund, Sweden
| | - Nélida Leiva Eriksson
- Pure and Applied Biochemistry, Department of Chemistry, Lund University, Box 124, 221 00 Lund, Sweden
| | - Luca Ronda
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Andrea Mozzarelli
- Department of Food and Drug, University of Parma, Parma, Italy; Institute of Biophysics, National Research Council (CNR), Pisa, Italy
| | - Andras Eke
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Domokos Mathe
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Brandon J Reeder
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, Essex CO4 3SQ, United Kingdom.
| |
Collapse
|
26
|
Wang A, Singh S, Yu B, Bloch DB, Zapol WM, Kluger R. Cross-linked hemoglobin bis-tetramers from bioorthogonal coupling do not induce vasoconstriction in the circulation. Transfusion 2018; 59:359-370. [PMID: 30444016 DOI: 10.1111/trf.15003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/07/2018] [Accepted: 09/08/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Hemoglobin-based oxygen carriers (HBOCs) are potential alternatives to red blood cells in transfusions. Clinical trials using early versions of HBOCs noted adverse effects that appeared to result from removal of the vasodilator nitric oxide (NO). Previous reports suggest that size-enlarged HBOCs may avoid NO-rich regions along the vasculature and therefore not cause vasoconstriction and hypertension. STUDY DESIGN AND METHODS Hemoglobin (Hb) bis-tetramers (bis-tetramers of hemoglobin that are prepared using CuAAC chemistry [BT-Hb] and bis-tetramers of hemoglobin that are specifically acetylated and prepared using CuAAC chemistry [BT-acHb]) can be reliably produced by a bio-orthogonal cyclo-addition approach. We considered that an HBOC derived from chemical coupling of two Hbs would be sufficiently large to avoid NO scavenging and related side effects. The ability of intravenously infused BT-Hb and BT-acHb to remain in the circulation without causing hypertension were determined in wild-type (WT) and diabetic (db/db) mouse models. RESULTS In WT mice, the coupled oxygen-carrying proteins retained their function over several hours after administration. No significant changes in systolic blood pressure from baseline were observed after intravenous infusion of BT-Hb or BT-acHb in awake WT and db/db mice. In contrast, infusion of native Hb or cross-linked Hb tetramers in both animal models induced systemic hypertension. CONCLUSION The results of this study indicate that bis-tetrameric HBOCs derived from the bio-orthogonal cyclo-addition process are likely to overcome clinical issues that arise from NO scavenging by Hb derivatives.
Collapse
Affiliation(s)
- Aizhou Wang
- Davenport Chemistry Research Laboratories, Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Serena Singh
- Davenport Chemistry Research Laboratories, Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Binglan Yu
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Donald B Bloch
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Division of Rheumatology, Allergy and Clinical Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Warren M Zapol
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ronald Kluger
- Davenport Chemistry Research Laboratories, Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Alomari E, Ronda L, Bruno S, Paredi G, Marchetti M, Bettati S, Olivari D, Fumagalli F, Novelli D, Ristagno G, Latini R, Cooper CE, Reeder BJ, Mozzarelli A. High- and low-affinity PEGylated hemoglobin-based oxygen carriers: Differential oxidative stress in a Guinea pig transfusion model. Free Radic Biol Med 2018; 124:299-310. [PMID: 29920341 PMCID: PMC6191936 DOI: 10.1016/j.freeradbiomed.2018.06.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/11/2018] [Accepted: 06/15/2018] [Indexed: 12/26/2022]
Abstract
Hemoglobin-based oxygen carriers (HBOCs) are an investigational replacement for blood transfusions and are known to cause oxidative damage to tissues. To investigate the correlation between their oxygen binding properties and these detrimental effects, we investigated two PEGylated HBOCs endowed with different oxygen binding properties - but otherwise chemically identical - in a Guinea pig transfusion model. Plasma samples were analyzed for biochemical markers of inflammation, tissue damage and organ dysfunction; proteins and lipids of heart and kidney extracts were analyzed for markers of oxidative damage. Overall, both HBOCs produced higher oxidative stress in comparison to an auto-transfusion control group. Particularly, tissue 4-hydroxynonenal adducts, tissue malondialdehyde adducts and plasma 8-oxo-2'-deoxyguanosine exhibited significantly higher levels in comparison with the control group. For malondialdehyde adducts, a higher level in the renal tissue was observed for animals treated with the high-affinity HBOC, hinting at a correlation between the HBOCs oxygen binding properties and the oxidative stress they produce. Moreover, we found that the high-affinity HBOC produced greater tissue oxygenation in comparison with the low affinity one, possibly correlating with the higher oxidative stress it induced.
Collapse
Affiliation(s)
- Esra'a Alomari
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Luca Ronda
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Biopharmanet-TEC, University of Parma, Parma, Italy
| | - Stefano Bruno
- Department of Food and Drug, University of Parma, Parma, Italy; Biopharmanet-TEC, University of Parma, Parma, Italy.
| | - Gianluca Paredi
- Department of Food and Drug, University of Parma, Parma, Italy; Interdepartmental Center SITEIA.PARMA, University of Parma, Parma 43121, Italy
| | - Marialaura Marchetti
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Biopharmanet-TEC, University of Parma, Parma, Italy
| | - Stefano Bettati
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Biopharmanet-TEC, University of Parma, Parma, Italy; Istituto Nazionale Biostrutture e Biosistemi, Rome, Italy
| | - Davide Olivari
- Istituto di Ricerche Farmacologiche 'Mario Negri', Milan, Italy
| | | | - Deborah Novelli
- Istituto di Ricerche Farmacologiche 'Mario Negri', Milan, Italy
| | | | - Roberto Latini
- Istituto di Ricerche Farmacologiche 'Mario Negri', Milan, Italy
| | - Chris E Cooper
- School of Biological Sciences, University of Essex, Colchester, United Kingdom
| | - Brandon J Reeder
- School of Biological Sciences, University of Essex, Colchester, United Kingdom
| | - Andrea Mozzarelli
- Department of Food and Drug, University of Parma, Parma, Italy; Biopharmanet-TEC, University of Parma, Parma, Italy; Istituto Nazionale Biostrutture e Biosistemi, Rome, Italy; Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Pisa, Italy
| |
Collapse
|
28
|
Zhang X, Williams MC, Rentsendorj O, D’Agnillo F. Reversible renal glomerular dysfunction in guinea pigs exposed to glutaraldehyde-polymerized cell-free hemoglobin. Toxicology 2018; 402-403:37-49. [DOI: 10.1016/j.tox.2018.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/13/2018] [Accepted: 04/18/2018] [Indexed: 01/17/2023]
|
29
|
Meng F, Kassa T, Jana S, Wood F, Zhang X, Jia Y, D’Agnillo F, Alayash AI. Comprehensive Biochemical and Biophysical Characterization of Hemoglobin-Based Oxygen Carrier Therapeutics: All HBOCs Are Not Created Equally. Bioconjug Chem 2018; 29:1560-1575. [DOI: 10.1021/acs.bioconjchem.8b00093] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Fantao Meng
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Tigist Kassa
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Sirsendu Jana
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Francine Wood
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Xiaoyuan Zhang
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Yiping Jia
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Felice D’Agnillo
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Abdu I. Alayash
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993, United States
| |
Collapse
|
30
|
Baek JH, Yalamanoglu A, Moon SE, Gao Y, Buehler PW. Evaluation of renal oxygen homeostasis in a preclinical animal model to elucidate difference in blood quality after transfusion. Transfusion 2018; 58:1474-1485. [DOI: 10.1111/trf.14560] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 01/03/2018] [Accepted: 01/19/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Jin Hyen Baek
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices
| | - Ayla Yalamanoglu
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices
| | - So-Eun Moon
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices
| | - Yamei Gao
- Division of Viral Products; Center of Biologics Evaluation and Research (CBER), FDA; Silver Spring Maryland
| | - Paul W. Buehler
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices
| |
Collapse
|
31
|
Movahed M, Roudkenar MH, Bahadori M, Mohammadipour M, Jalili MA, Amiri F. Establishment of Stable CHO Cell Line Expressing Recombinant Human Haptoglobin: Toward New Haptoglobin-Based Therapeutics. IRANIAN JOURNAL OF SCIENCE AND TECHNOLOGY TRANSACTION A-SCIENCE 2017. [DOI: 10.1007/s40995-017-0381-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
32
|
Jungner Å, Vallius S, Bruschettini M, Romantsik O, Gram M, Ley D. Cardiopulmonary bypass in the newborn: effects of circulatory cell-free hemoglobin and hyperoxia evaluated in a novel rat pup model. Intensive Care Med Exp 2017; 5:45. [PMID: 28980221 PMCID: PMC5628085 DOI: 10.1186/s40635-017-0153-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/21/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Infants with congenital heart defects (CHD) are at risk for white matter brain injury. This novel rat pup model characterizes the systemic effects of intravasal cell-free hemoglobin and hyperoxia, hypothesizing that immature endogenous scavenging systems relate to increased vulnerability to conditions present during cardiopulmonary bypass (CPB). METHODS Plasma pharmacokinetics of cell-free human hemoglobin (Hb) was determined after intraperitoneal (i.p.) administration in postnatal day 6 (P6) rat pups. Cell-free hemoglobin degradation, scavenger- and oxidative stress responses in altered oxygen environments were evaluated in P6 rat pups exposed to i.p. cell-free Hb or vehicle and subjected to hyperoxia or normoxia for 24 h. Plasma and liver were analyzed for free heme, haptoglobin, hemopexin, heme-oxygenase 1, and 8-OHdG at 3-120 h post-injection. Baseline scavenging properties were evaluated in P0-P12 rat pups. RESULTS Cell-free Hb displayed peak plasma concentrations of 3.6 ± 0.5 mg/mL (mean ± SD) at 3 h post-administration. Animals exposed to cell-free Hb demonstrated a 30-fold increase in plasma haptoglobin and a decrease in plasma hemopexin to 1/6 of concentrations observed in pups exposed to vehicle. Exposure to cell-free Hb and hyperoxia mediated increased plasma concentrations of free heme (72.7 ± 19.5 μM, mean ± SD) compared to exposure to cell-free Hb and normoxia (49.3 ± 13.1 μM) at 3 h, and an elevated hepatic mRNA expression of heme-oxygenase 1. mRNA expression of haptoglobin and hemopexin was increased in animals exposed to hemoglobin with a mitigated response in pups exposed to hemoglobin and hyperoxia. Animals exposed to hyperoxia displayed an increase in hepatic transcription of scavenger proteins at 24 h. Combined exposure to cell-free Hb and hyperoxia mediated an increased DNA-oxidation at 6 h, whereas all insults conveyed a decrease in DNA-oxidation at 120 h. CONCLUSIONS In this study, we present a novel rat pup model with scavenging characteristics and brain maturation similar to newborns with CHD. We have confirmed a distinct scavenger response after exposure to systemic cell-free hemoglobin. We have indications of an accelerated metabolism of cell-free Hb and of an altered transcription of scavenger proteins in a hyperoxic environment. We believe that this model will prove valuable in future delineation of inflammatory and oxidative end-organ damage following CPB.
Collapse
Affiliation(s)
- Åsa Jungner
- Department of Clinical Sciences Lund, Pediatrics, Skane University Hospital, Lund University, Lund, Sweden
- Department of Clinical Sciences Lund, Pediatric Surgery and Neonatal Care, Skane University Hospital, Lund University, Lund, Sweden
- Pediatric Intensive Care Unit (BIVA), Skane University Hospital, Lund, Sweden
| | - Suvi Vallius
- Department of Clinical Sciences Lund, Pediatrics, Skane University Hospital, Lund University, Lund, Sweden
| | - Matteo Bruschettini
- Department of Clinical Sciences Lund, Pediatrics, Skane University Hospital, Lund University, Lund, Sweden
- Department of Clinical Sciences Lund, Pediatric Surgery and Neonatal Care, Skane University Hospital, Lund University, Lund, Sweden
| | - Olga Romantsik
- Department of Clinical Sciences Lund, Pediatrics, Skane University Hospital, Lund University, Lund, Sweden
- Department of Clinical Sciences Lund, Pediatric Surgery and Neonatal Care, Skane University Hospital, Lund University, Lund, Sweden
| | - Magnus Gram
- Department of Clinical Sciences Lund, Pediatrics, Skane University Hospital, Lund University, Lund, Sweden
- Department of Clinical Sciences Lund, Infection Medicine, Lund University, Lund, Sweden
| | - David Ley
- Department of Clinical Sciences Lund, Pediatrics, Skane University Hospital, Lund University, Lund, Sweden
- Department of Clinical Sciences Lund, Pediatric Surgery and Neonatal Care, Skane University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
33
|
Alayash AI. Oxidative pathways in the sickle cell and beyond. Blood Cells Mol Dis 2017; 70:78-86. [PMID: 28554826 DOI: 10.1016/j.bcmd.2017.05.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 02/07/2023]
Abstract
Polymerization of deoxy sickle cell hemoglobin (HbS) is well recognized as the primary event that triggers the classic cycles of sickling/unsickling of patients red blood cells (RBCs). RBCs are also subjected to continuous endogenous and exogenous oxidative onslaughts resulting in hemolytic rate increases which contribute to the evolution of vasculopathies associated with this disease. Compared to steady-state conditions, the occurrences of vaso-occlusive crises increase the levels of both RBC-derived microparticles as well as extracellular Hb in circulation. Common byproduct resulting from free Hb oxidation and from Hb-laden microparticles is heme (now recognized as damage associated molecular pattern (DAMP) molecule) which has been shown to initiate inflammatory responses. This review provides new insights into the interplay between microparticles, free Hb and heme focusing on Hb's pseudoperoxidative activity that drives RBC's cytosolic, membrane changes as well as oxidative toxicity towards the vascular system. Emerging antioxidative strategies that include the use of protein and heme scavengers in controlling Hb oxidative pathways are discussed.
Collapse
Affiliation(s)
- Abdu I Alayash
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD, USA.
| |
Collapse
|
34
|
Alayash AI. Hemoglobin-Based Blood Substitutes and the Treatment of Sickle Cell Disease: More Harm than Help? Biomolecules 2017; 7:biom7010002. [PMID: 28054978 PMCID: PMC5372714 DOI: 10.3390/biom7010002] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 12/20/2016] [Accepted: 12/26/2016] [Indexed: 01/17/2023] Open
Abstract
Intense efforts have been made by both industry and academia over the last three decades to produce viable hemoglobin (Hb)-based oxygen carriers (HBOCs), also known as “blood substitutes”. Human trials conducted so far by several manufactures in a variety of clinical indications, including trauma, and elective surgeries have failed and no product has gained the Food and Drug Administration approval for human use. Safety concerns due to frequent incidences of hemodynamic, cardiac events, and even death led to the termination of some of these trials. Several second generation HBOC products that have been chemically and/or genetically modified (or in some cases ligated with carbon monoxide (CO)) found a new clinical application in conditions as complex as sickle cell disease (SCD). By virtue of higher oxygen affinity (P50) (R-state), and smaller size, HBOCs may be able to reach the microvasculature unload of oxygen to reverse the cycles of sickling/unsickling of the deoxy-sickle cell Hb (HbS) (T-state), thus preventing vaso-occlusion, a central event in SCD pathophysiology. However, biochemically, it is thought that outside the red blood cell (due to frequent hemolysis), free HbS or infused HBOCs are capable of interfering with a number of oxidative and signaling pathways and may, thus, negate any benefit that HBOCs may provide. This review discusses the advantages and disadvantages of using HBOCs in SCD.
Collapse
Affiliation(s)
- Abdu I Alayash
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20993, USA.
| |
Collapse
|
35
|
Kassa T, Jana S, Meng F, Alayash AI. Differential heme release from various hemoglobin redox states and the upregulation of cellular heme oxygenase-1. FEBS Open Bio 2016; 6:876-84. [PMID: 27642551 PMCID: PMC5011486 DOI: 10.1002/2211-5463.12103] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/05/2016] [Accepted: 07/11/2016] [Indexed: 01/24/2023] Open
Abstract
Despite advances in our understanding of the oxidative pathways mediated by free hemoglobin (Hb), the precise contribution of its highly reactive redox forms to tissue and organ toxicities remains ambiguous. Heme, a key degradation byproduct of Hb oxidation, has recently been recognized as a damage-associated molecular pattern (DAMP) molecule, able to trigger inflammatory responses. Equally damaging is the interaction of the highly redox active forms of Hb with other biological molecules. We determined the kinetics of heme loss from individual Hb redox states-ferrous (Fe(2+)), ferric (Fe(3+)), and ferryl (Fe(4+))-using two different heme receptor proteins: hemopexin (Hxp), a naturally occurring heme scavenger in plasma, and a double mutant (H64Y/V86F), apomyoglobin (ApoMb), which avidly binds heme released from Hb. We show for the first time that ferric Hb (Fe(3+)) loses heme at rates substantially higher than that of ferryl Hb (Fe(4+)). This was also supported by a higher expression of heme oxygenase-1 (HO-1) when ferric Hb was added to cultured lung alveolar epithelial cells (E10). The reported cytotoxicity of Hb may therefore be attributed to a combination of accelerated heme loss from the ferric form and protein radical formation associated with ferryl Hb. Targeted therapeutic interventions can therefore be designed to curb specific oxidative pathways of Hb in hemolytic anemias and when Hb is used as an oxygen-carrying therapeutic.
Collapse
Affiliation(s)
- Tigist Kassa
- Laboratory of Biochemistry and Vascular Biology Center for Biologics Evaluation and Research Food and Drug Administration Silver Spring MD USA
| | - Sirsendu Jana
- Laboratory of Biochemistry and Vascular Biology Center for Biologics Evaluation and Research Food and Drug Administration Silver Spring MD USA
| | - Fantao Meng
- Laboratory of Biochemistry and Vascular Biology Center for Biologics Evaluation and Research Food and Drug Administration Silver Spring MD USA
| | - Abdu I Alayash
- Laboratory of Biochemistry and Vascular Biology Center for Biologics Evaluation and Research Food and Drug Administration Silver Spring MD USA
| |
Collapse
|
36
|
Clafshenkel WP, Murata H, Andersen J, Creeger Y, Koepsel RR, Russell AJ. The Effect of Covalently-Attached ATRP-Synthesized Polymers on Membrane Stability and Cytoprotection in Human Erythrocytes. PLoS One 2016; 11:e0157641. [PMID: 27331401 PMCID: PMC4917246 DOI: 10.1371/journal.pone.0157641] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 06/02/2016] [Indexed: 12/28/2022] Open
Abstract
Erythrocytes have been described as advantageous drug delivery vehicles. In order to ensure an adequate circulation half-life, erythrocytes may benefit from protective enhancements that maintain membrane integrity and neutralize oxidative damage of membrane proteins that otherwise facilitate their premature clearance from circulation. Surface modification of erythrocytes using rationally designed polymers, synthesized via atom-transfer radical polymerization (ATRP), may further expand the field of membrane-engineered red blood cells. This study describes the fate of ATRP-synthesized polymers that were covalently attached to human erythrocytes as well as the effect of membrane engineering on cell stability under physiological and oxidative conditions in vitro. The biocompatible, membrane-reactive polymers were homogenously retained on the periphery of modified erythrocytes for at least 24 hours. Membrane engineering stabilized the erythrocyte membrane and effectively neutralized oxidative species, even in the absence of free-radical scavenger-containing polymers. The targeted functionalization of Band 3 protein by NHS-pDMAA-Cy3 polymers stabilized its monomeric form preventing aggregation in the presence of the crosslinking reagent, bis(sulfosuccinimidyl)suberate (BS3). A free radical scavenging polymer, NHS-pDMAA-TEMPO˙, provided additional protection of surface modified erythrocytes in an in vitro model of oxidative stress. Preserving or augmenting cytoprotective mechanisms that extend circulation half-life is an important consideration for the use of red blood cells for drug delivery in various pathologies, as they are likely to encounter areas of imbalanced oxidative stress as they circuit the vascular system.
Collapse
Affiliation(s)
- William P. Clafshenkel
- The Institute for Complex Engineered Systems, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Hironobu Murata
- The Institute for Complex Engineered Systems, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Jill Andersen
- The Institute for Complex Engineered Systems, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Yehuda Creeger
- Molecular Biosensor and Imaging Center, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Richard R. Koepsel
- The Institute for Complex Engineered Systems, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Alan J. Russell
- The Institute for Complex Engineered Systems, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
37
|
Rentsendorj O, Zhang X, Williams MC, Buehler PW, D’Agnillo F. Transcriptional Suppression of Renal Antioxidant Enzyme Systems in Guinea Pigs Exposed to Polymerized Cell-Free Hemoglobin. TOXICS 2016; 4. [PMID: 27471729 PMCID: PMC4961095 DOI: 10.3390/toxics4010006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Hemoglobin-based oxygen carriers (HBOCs) are being developed as oxygen and plasma volume-expanding therapeutics though their potential to promote oxidative tissue injury has raised safety concerns. Using a guinea pig exchange transfusion model, we examined the effects of polymerized bovine hemoglobin (HbG) on the transcriptional regulation, activity, and expression of the renal antioxidant enzymes; superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx). HbG infusion downregulated the mRNA levels for genes encoding SOD isoforms 1-3, GPx1, GPx3, GPx4, and CAT. This transcriptional suppression correlated with decreased enzymatic activities for SOD, CAT, and GPx. Immunostaining revealed decreased protein expression of SOD1, CAT, and GPx1 primarily in renal cortical tubules. DNA methylation analyses identified CpG hypermethylation in the gene promoters for SOD1-3, GPx1, GPx3, and GPx4, suggesting an epigenetic-based mechanism underlying the observed gene repression. HbG also induced oxidative stress as evidenced by increased renal lipid peroxidation end-products and 4-HNE immunostaining, which could be the result of the depleted antioxidant defenses and/or serve as a trigger for increased DNA methylation. Together, these findings provide evidence that the renal exposure to HbG suppresses the function of major antioxidant defense systems which may have relevant implications for understanding the safety of hemoglobin-based products.
Collapse
|
38
|
Estep TN. Pharmacokinetics and mechanisms of plasma removal of hemoglobin-based oxygen carriers. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 43:203-15. [PMID: 26024447 DOI: 10.3109/21691401.2015.1047501] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The circulatory persistence, distribution, and metabolism of hemoglobin-based oxygen carriers (HBOCs) is a major determinant of their safety and efficacy. In this communication, published data on the pharmacokinetics and routes of plasma elimination of HBOCs are summarized and evaluated. The circulating half-life of HBOCs is dose-dependent in both animals and humans. Half-life also increases with molecular weight in animals, at least up to the MDa range. The functional half-life of HBOCs is diminished by as much as 40% due to oxidation of the heme group relative to the overall rate of removal of hemoglobin (Hb) from plasma. Kidney excretion of HBOCs is greatly diminished compared to that of unmodified Hb, but the liver remains a primary site of catabolism. Both hepatocytes and Kupffer cells have been implicated in receptor-mediated HBOC uptake. Removal also occurs in the spleen and/or bone marrow and probably at dispersed sites in the endothelium as well. HBOCs extravasate into the lymph at a rate inversely proportional to their molecular weight and are taken up by monocyte/macrophage CD163 receptors, both as free Hb and in complexes with haptoglobin (Hp). The interactions with both Hp and the CD163 receptor are altered by Hb modification. However, monocyte/macrophage uptake may not be a quantitatively important route for the removal of clinically relevant doses of HBOCs. The relative contributions of different removal pathways have yet to be comprehensively determined, particularly in humans.
Collapse
|
39
|
Baek JH, Zhang X, Williams MC, Hicks W, Buehler PW, D'Agnillo F. Sodium nitrite potentiates renal oxidative stress and injury in hemoglobin exposed guinea pigs. Toxicology 2015; 333:89-99. [PMID: 25891524 DOI: 10.1016/j.tox.2015.04.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 03/17/2015] [Accepted: 04/14/2015] [Indexed: 12/18/2022]
Abstract
Methemoglobin-forming drugs, such as sodium nitrite (NaNO2), may exacerbate oxidative toxicity under certain chronic or acute hemolytic settings. In this study, we evaluated markers of renal oxidative stress and injury in guinea pigs exposed to extracellular hemoglobin (Hb) followed by NaNO2 at doses sufficient to simulate clinically relevant acute methemoglobinemia. NaNO2 induced rapid and extensive oxidation of plasma Hb in this model. This was accompanied by increased renal expression of the oxidative response effectors nuclear factor erythroid 2-derived-factor 2 (Nrf-2) and heme oxygenase-1 (HO-1), elevated non-heme iron deposition, lipid peroxidation, interstitial inflammatory cell activation, increased expression of tubular injury markers kidney injury-1 marker (KIM-1) and liver-fatty acid binding protein (L-FABP), podocyte injury, and cell death. Importantly, these indicators of renal oxidative stress and injury were minimal or absent following infusion of Hb or NaNO2 alone. Together, these results suggest that the exposure to NaNO2 in settings associated with increased extracellular Hb may potentiate acute renal toxicity via processes that are independent of NaNO2 induced erythrocyte methemoglobinemia.
Collapse
Affiliation(s)
- Jin Hyen Baek
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Xiaoyuan Zhang
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Matthew C Williams
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Wayne Hicks
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Paul W Buehler
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Felice D'Agnillo
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA.
| |
Collapse
|
40
|
Schaer DJ, Vinchi F, Ingoglia G, Tolosano E, Buehler PW. Haptoglobin, hemopexin, and related defense pathways-basic science, clinical perspectives, and drug development. Front Physiol 2014; 5:415. [PMID: 25389409 PMCID: PMC4211382 DOI: 10.3389/fphys.2014.00415] [Citation(s) in RCA: 201] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 10/08/2014] [Indexed: 12/13/2022] Open
Abstract
Hemolysis, which occurs in many disease states, can trigger a diverse pathophysiologic cascade that is related to the specific biochemical activities of free Hb and its porphyrin component heme. Normal erythropoiesis and concomitant removal of senescent red blood cells (RBC) from the circulation occurs at rates of approximately 2 × 106 RBCs/second. Within this physiologic range of RBC turnover, a small fraction of hemoglobin (Hb) is released into plasma as free extracellular Hb. In humans, there is an efficient multicomponent system of Hb sequestration, oxidative neutralization and clearance. Haptoglobin (Hp) is the primary Hb-binding protein in human plasma, which attenuates the adverse biochemical and physiologic effects of extracellular Hb. The cellular receptor target of Hp is the monocyte/macrophage scavenger receptor, CD163. Following Hb-Hp binding to CD163, cellular internalization of the complex leads to globin and heme metabolism, which is followed by adaptive changes in antioxidant and iron metabolism pathways and macrophage phenotype polarization. When Hb is released from RBCs within the physiologic range of Hp, the potential deleterious effects of Hb are prevented. However, during hyper-hemolytic conditions or with chronic hemolysis, Hp is depleted and Hb readily distributes to tissues where it might be exposed to oxidative conditions. In such conditions, heme can be released from ferric Hb. The free heme can then accelerate tissue damage by promoting peroxidative reactions and activation of inflammatory cascades. Hemopexin (Hx) is another plasma glycoprotein able to bind heme with high affinity. Hx sequesters heme in an inert, non-toxic form and transports it to the liver for catabolism and excretion. In the present review we discuss the components of physiologic Hb/heme detoxification and their potential therapeutic application in a wide range of hemolytic conditions.
Collapse
Affiliation(s)
- Dominik J Schaer
- Division of Internal Medicine, University of Zurich Zurich, Switzerland
| | - Francesca Vinchi
- Department of Molecular Biotechnology and Health Sciences, University of Torino Torino, Italy
| | - Giada Ingoglia
- Department of Molecular Biotechnology and Health Sciences, University of Torino Torino, Italy
| | - Emanuela Tolosano
- Department of Molecular Biotechnology and Health Sciences, University of Torino Torino, Italy
| | - Paul W Buehler
- Division of Hematology, Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration Bethesda, MD, USA
| |
Collapse
|
41
|
Sakai H, Li B, Lim WL, Iga Y. Red blood cells donate electrons to methylene blue mediated chemical reduction of methemoglobin compartmentalized in liposomes in blood. Bioconjug Chem 2014; 25:1301-10. [PMID: 24877769 DOI: 10.1021/bc500153x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Electron-energy-rich coenzymes in cells, NADH and NADPH, are re-energized repeatedly through the Embden-Meyerhof and pentose-phosphate glycolytic pathways, respectively. This study demonstrates extraction of their electron energies in red blood cells (RBCs) for in vivo extracellular chemical reactions using an electron mediator shuttling across the biomembrane. Hemoglobin-vesicles (HbVs) are an artificial oxygen carrier encapsulating purified and concentrated Hb solution in liposomes. Because of the absence of a metHb-reducing enzymatic system in HbV, HbO2 gradually autoxidizes to form metHb. Wistar rats received HbV suspension (10 mL/kg body weight) intravenously. At the metHb level of around 50%, methylene blue [MB(+); 3,7-bis(dimethylamino)phenothiazinium chloride] was injected. The level of metHb quickly decreased to around 16% in 40 min, remaining for more than 5 h. In vitro mixing of HbV/MB(+) with RBCs recreated the in vivo metHb reduction, but not with plasma. NAD(P)H levels in RBCs decreased after metHb reduction. The addition of glucose facilitated metHb reduction. Liposome-encapsulated NAD(P)H, a model of RBC, reduced metHb in HbV in the presence of MB(+). These results indicate that (i) NAD(P)H in RBCs reacts with MB(+) to convert it to leukomethylene blue (MBH); (ii) MB(+) and MBH shuttle freely between RBC and HbV across the hydrophobic lipid membranes; and (iii) MBH is transferred into HbV and reduces metHb in HbV. Four other electron mediators with appropriate redox potentials appeared to be as effective as MB(+) was, indicating the possibility for further optimization of electron mediators. We established an indirect enzymatic metHb reducing system for HbV using unlimited endogenous electrons created in RBCs in combination with an effective electron mediator that prolongs the functional lifespan of HbV in blood circulation.
Collapse
Affiliation(s)
- Hiromi Sakai
- Department of Chemistry, Nara Medical University , 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | | | | | | |
Collapse
|
42
|
Alayash AI. Blood substitutes: why haven't we been more successful? Trends Biotechnol 2014; 32:177-85. [PMID: 24630491 PMCID: PMC4418436 DOI: 10.1016/j.tibtech.2014.02.006] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 02/07/2014] [Accepted: 02/10/2014] [Indexed: 02/07/2023]
Abstract
Persistent safety concerns have stalled the development of viable hemoglobin (Hb)-based oxygen carriers (HBOCs). HBOCs have several advantages over human blood, including availability, long-term storage, and lack of infectious risk. The basis of HBOC toxicity is poorly understood, however, several mechanisms have been suggested, including Hb extravasation across the blood vessel wall, scavenging of endothelial nitric oxide (NO), oversupply of oxygen, and heme-mediated oxidative side reactions. Although there are some in vitro and limited animal studies supporting these mechanisms, heme-mediated reactivity appears to provide an alternative path that can explain some of the observed pathophysiological changes. Moreover, recent mechanistic and animal studies support a role for globin and heme scavengers in controlling oxidative toxicity associated with Hb infusion.
Collapse
Affiliation(s)
- Abdu I Alayash
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland 20892, USA.
| |
Collapse
|
43
|
Vandegriff KD, Malavalli A, Lohman J, Young MA, Terraneo L, Virgili E, Bianciardi P, Caretti A, Samaja M. Impact of acellular hemoglobin-based oxygen carriers on brain apoptosis in rats. Transfusion 2014; 54:2045-54. [PMID: 24673504 DOI: 10.1111/trf.12643] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 12/23/2013] [Accepted: 12/26/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Extracellular hemoglobin (Hb)-based oxygen carriers (HBOCs) are under extensive consideration as oxygen therapeutics. Their effects on cellular mechanisms related to apoptosis are of particular interest, because the onset of proapoptotic pathways may give rise to tissue damage. STUDY DESIGN AND METHODS The objective was to assess whether the properties of the Hb that replaces blood during an isovolemic hemodilution would modulate apoptotic-response mechanisms in rat brain and whether such signaling favors cytoprotection or damage. We exposed rats to exchange transfusion (ET; 50% blood volume and isovolemic replacement with Hextend [negative colloid control], MP4OX [PEGylated HBOC with high oxygen affinity], and ααHb [αα-cross-linked HBOC with low oxygen affinity; n=4-6/group]). Sham rats acted as control. Animals were euthanized at 2, 6, and 12 hours after ET; brain tissue was harvested and processed for analysis. RESULTS In MP4OX animals, the number of neurons that overexpressed the hypoxia-inducible factor (HIF)-1α was higher than in ααHb, particularly at the early time points. In addition, MP4OX was associated with greater phosphorylation of protein kinase B (Akt), a well-known cytoprotective factor. Indeed, the degree of apoptosis, measured as terminal deoxynucleotidyl transferase-positive neurons and caspase-3 cleavage, ranked in order of MP4OX < Hextend < ααHb. CONCLUSION Even though both HBOCs showed increased levels of HIF-1α compared to shams or Hextend-treated animals, differences in signaling events resulted in very different outcomes for the two HBOCs. ααHb-treated brain tissue showed significant neuronal damage, measured as apoptosis. This was in stark contrast to the protection seen with MP4OX, apparently due to recruitment of Akt and neuronal specific HIF-1α pathways.
Collapse
|
44
|
Lisk C, Kominsky D, Ehrentraut S, Bonaventura J, Nuss R, Hassell K, Nozik-Grayck E, Irwin DC. Hemoglobin-induced endothelial cell permeability is controlled, in part, via a myeloid differentiation primary response gene-88-dependent signaling mechanism. Am J Respir Cell Mol Biol 2014; 49:619-26. [PMID: 23713977 DOI: 10.1165/rcmb.2012-0440oc] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The release of hemoglobin (Hb) with hemolysis causes vascular dysfunction. New evidence implicates Hb-induced NF-κB and hypoxia inducible factor (HIF) activation, which may be under the control of a Toll-like receptor (TLR)-signaling pathway. Nearly all TLR-signaling pathways activate the myeloid differentiation primary response gene-88 (MyD88) that regulates NF-κB. We hypothesized that the differing transition states of Hb influence endothelial cell permeability via NF-κB activation and HIF regulation through a MyD88-dependent pathway. In cultured human dermal microvascular endothelial cells (HMECs-1), we examined the effects of Hb in the ferrous (HbFe(2+)), ferric (HbFe(3+)), and ferryl (HbFe(4+)) transition states on NF-κB and HIF activity, HIF-1α and HIF-2α mRNA up-regulation, and monolayer permeability, in the presence or absence of TLR4, MyD88, NF-κB, or HIF inhibition, as well as superoxide dismutase (SOD) and catalase. Our data showed that cell-free Hb, in each transition state, induced NF-κB and HIF activity, up-regulated HIF-1α and HIF-2α mRNA, and increased HMEC-1 permeability. The blockade of either MyD88 or NF-κB, but not TLR4, attenuated Hb-induced HIF activity, the up-regulation HIF-1 and HIF-2α mRNA, and HMEC-1 permeability. The inhibition of HIF activity exerted less of an effect on Hb-induced monolayer permeability. Moreover, SOD and catalase attenuated NF-κB, HIF activity, and monolayer permeability. Our results demonstrate that Hb-induced NF-κB and HIF are regulated by two mechanisms, either MyD88 activation or Hb transition state-induced ROS formation, that influence HMEC-1 permeability.
Collapse
Affiliation(s)
- Christina Lisk
- 1 Cardiovascular Pulmonary Research Group, Division of Cardiology, Department of Medicine
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Weiskopf RB, Silverman TA. Balancing potential risks and benefits of hemoglobin-based oxygen carriers. Transfusion 2013; 53:2327-33. [PMID: 23869543 DOI: 10.1111/trf.12339] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 05/20/2013] [Accepted: 05/21/2013] [Indexed: 12/23/2022]
Abstract
Hemoglobin-based oxygen carriers (HBOCs) are thought to have an adverse risk:benefit profile when compared to that of transfusing stored red blood cells (RBCs). However, there are clinical circumstances when RBC transfusion is not an option (e.g., patient refusal, unavailability owing to issues of compatibility or remote location). For these circumstances assessment of the risks of an HBOC should be compared to the risks of untransfused acute anemia. In this article we compare the risk of allowing a patient with severe anemia to have a further small decrease in hemoglobin (Hb) concentration to the risk of infusing an HBOC. We conclude that at Hb concentrations less than 6 g/dL, the risk of a further decrease in Hb concentration greatly exceeds the risk of HBOC infusion. Thus, we suggest that there may be a place for use of HBOCs when RBC transfusion is not an option.
Collapse
Affiliation(s)
- Richard B Weiskopf
- University of California, San Francisco, California; PAREXEL Consulting, Waltham, Massachusetts
| | | |
Collapse
|
46
|
Abstract
The development of oxygen (O2)-carrying blood substitutes has evolved from the goal of replicating blood O2 transport properties to that of preserving microvascular and organ function, reducing the inherent or potential toxicity of the material used to carry O2, and treating pathologies initiated by anemia and hypoxia. Furthermore, the emphasis has shifted from blood replacement fluid to "O2 therapeutics" that restore tissue oxygenation to specific tissues regions. This review covers the different alternatives, potential and limitations of hemoglobin-based O2 carriers (HBOCs) and perfluorocarbon-based O2 carriers (PFCOCs), with emphasis on the physiologic conditions disturbed in the situation that they will be used. It describes how concepts learned from plasma expanders without O2-carrying capacity can be applied to maintain O2 delivery and summarizes the microvascular responses due to HBOCs and PFCOCs. This review also presents alternative applications of HBOCs and PFCOCs namely: 1) How HBOC O2 affinity can be engineered to target O2 delivery to hypoxic tissues; and 2) How the high gas solubility of PFCOCs provides new opportunities for carrying, dissolving, and delivering gases with biological activity. It is concluded that the development of current blood substitutes has amplified their applications horizon by devising therapeutic functions for O2 carriers requiring limited O2 delivery capacity restoration. Conversely, full, blood-like O2-carrying capacity reestablishment awaits the control of O2 carrier toxicity.
Collapse
Affiliation(s)
- Pedro Cabrales
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093-0412, USA.
| | | |
Collapse
|
47
|
Abstract
SIGNIFICANCE There has been a striking advancement in our understanding of red cell substitutes over the past decade. Although regulatory oversight has influenced many aspects of product development in this period, those who have approached the demonstration of efficacy of red cell substitutes have failed to understand their implication at the level of the microcirculation, where blood interacts closely with tissue. RECENT ADVANCES The understanding of the adverse effects of acellular hemoglobin (Hb)-based oxygen carriers (HBOCs) has fortunately expanded from Hb-induced renal toxicity to a more complete list of biochemical mechanism. In addition, various unexpected adverse reactions were seen in early clinical studies. The effects of the presence of acellular Hb in plasma are relatively unique because of the convergence of mechanical and biochemical natures. CRITICAL ISSUES Controlling the variables using genetic engineering and chemical modification to change specific characteristics of the Hb molecule may allow for solving the complex multivariate problems of acellular Hb vasoactivity. HBOCs may never be rendered free of negative effects; however, quantifying the nature and extent of microvascular complications establishes a platform for designing new ameliorative therapies. FUTURE DIRECTIONS It is time to leave behind the study of vasoactivity and toxicity based on bench-top measurements of biochemical changes and those based solely on systemic parameters in vivo, and move to a more holistic analysis of the mechanisms creating the problems, complemented with meaningful studies of efficacy.
Collapse
Affiliation(s)
- Pedro Cabrales
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
48
|
Bonaventura C, Henkens R, Alayash AI, Banerjee S, Crumbliss AL. Molecular controls of the oxygenation and redox reactions of hemoglobin. Antioxid Redox Signal 2013; 18:2298-313. [PMID: 23198874 PMCID: PMC4047995 DOI: 10.1089/ars.2012.4947] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 11/12/2012] [Accepted: 12/01/2012] [Indexed: 01/04/2023]
Abstract
SIGNIFICANCE The broad classes of O(2)-binding proteins known as hemoglobins (Hbs) carry out oxygenation and redox functions that allow organisms with significantly different physiological demands to exist in a wide range of environments. This is aided by allosteric controls that modulate the protein's redox reactions as well as its O(2)-binding functions. RECENT ADVANCES The controls of Hb's redox reactions can differ appreciably from the molecular controls for Hb oxygenation and come into play in elegant mechanisms for dealing with nitrosative stress, in the malarial resistance conferred by sickle cell Hb, and in the as-yet unsuccessful designs for safe and effective blood substitutes. CRITICAL ISSUES An important basic principle in consideration of Hb's redox reactions is the distinction between kinetic and thermodynamic reaction control. Clarification of these modes of control is critical to gaining an increased understanding of Hb-mediated oxidative processes and oxidative toxicity in vivo. FUTURE DIRECTIONS This review addresses emerging concepts and some unresolved questions regarding the interplay between the oxygenation and oxidation reactions of structurally diverse Hbs, both within red blood cells and under acellular conditions. Developing methods that control Hb-mediated oxidative toxicity will be critical to the future development of Hb-based blood substitutes.
Collapse
Affiliation(s)
- Celia Bonaventura
- Nicholas School of the Environment, Duke University Marine Laboratory, Beaufort, NC 28516, USA.
| | | | | | | | | |
Collapse
|
49
|
Schaer DJ, Buehler PW. Cell-free hemoglobin and its scavenger proteins: new disease models leading the way to targeted therapies. Cold Spring Harb Perspect Med 2013; 3:cshperspect.a013433. [PMID: 23645855 DOI: 10.1101/cshperspect.a013433] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Hemoglobin (Hb) has multiple pathophysiologic effects when released into the intravascular space during hemolysis. The extracellular effects of Hb have resulted in novel models of toxicity, which help to explain endothelial dysfunction and cardiovascular complications that accompany genetic hemolytic anemias, malaria, blood transfusion, and atherosclerosis. The majority of models focus on nitric oxide (NO) depletion; however, in local tissue environments, Hb can also act as a pro-oxidant and inflammatory agent. This can alter cellular differentiation with the potential to deviate immune responses. The understanding of these mechanisms set in the context of natural scavenger and detoxification systems may accelerate the development of novel treatment strategies.
Collapse
Affiliation(s)
- Dominik J Schaer
- Division of Internal Medicine, University Hospital, Zurich CH-8091, Switzerland.
| | | |
Collapse
|
50
|
Hemolysis and free hemoglobin revisited: exploring hemoglobin and hemin scavengers as a novel class of therapeutic proteins. Blood 2012; 121:1276-84. [PMID: 23264591 DOI: 10.1182/blood-2012-11-451229] [Citation(s) in RCA: 585] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
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.
Collapse
|