1
|
Ala C, Joshi RP, Gupta P, Goswami SG, Ramalingam S, Kondapalli Venkata Gowri CS, Sankaranarayanan M. A critical review of therapeutic interventions in sickle cell disease: Progress and challenges. Arch Pharm (Weinheim) 2024; 357:e2400381. [PMID: 39031925 DOI: 10.1002/ardp.202400381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/22/2024]
Abstract
Sickle cell disease (SCD) is an autosomal recessive genetic disorder that occurs due to the point mutation in the β-globin gene, which results in the formation of sickle hemoglobin (HbS) in the red blood cells (RBCs). When HbS is exposed to an oxygen-depleted environment, it polymerizes, resulting in hemolysis, vaso-occlusion pain, and impaired blood flow. Still, there is no affordable cure for this inherited disease. Approved medications held promise but were met with challenges due to limited patient tolerance and undesired side effects, thereby inhibiting their ability to enhance the quality of life across various individuals with SCD. Progress has been made in understanding the pathophysiology of SCD during the past few decades, leading to the discovery of novel targets and therapies. However, there is a compelling need for research to discover medications with improved efficacy and reduced side effects. Also, more clinical investigations on various drug combinations with different mechanisms of action are needed. This review comprehensively presents therapeutic approaches for SCD, including those currently available or under investigation. It covers fundamental aspects of the disease, such as epidemiology and pathophysiology, and provides detailed discussions on various disease-modifying agents. Additionally, expert insights are offered on the future development of pharmacotherapy for SCD.
Collapse
Affiliation(s)
- Chandu Ala
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Medicinal Chemistry Research Laboratory, Pilani Campus, Pilani, Rajasthan, India
| | - Renuka Parshuram Joshi
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Medicinal Chemistry Research Laboratory, Pilani Campus, Pilani, Rajasthan, India
| | - Pragya Gupta
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | | | | | | | - Murugesan Sankaranarayanan
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Medicinal Chemistry Research Laboratory, Pilani Campus, Pilani, Rajasthan, India
| |
Collapse
|
2
|
Chonat S, Stowell SR. COping with acute sickle cell hemolysis. Blood 2024; 143:2452-2453. [PMID: 38869921 DOI: 10.1182/blood.2024024710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
|
3
|
Nguyen KA, Matte A, Foresti R, Federti E, Kiger L, Lefebvre C, Hocini H, Pelinski Y, Kitagishi H, Bencheikh L, Pirenne F, de Franceschi L, Motterlini R, Bartolucci P. An oral carbon monoxide-releasing molecule protects against acute hyperhemolysis in sickle cell disease. Blood 2024; 143:2544-2558. [PMID: 38518106 DOI: 10.1182/blood.2023023165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/06/2024] [Accepted: 03/17/2024] [Indexed: 03/24/2024] Open
Abstract
ABSTRACT Acute hyperhemolysis is a severe life-threatening complication in patients with sickle cell disease (SCD) that may occur during delayed hemolytic transfusion reaction (DHTR), or vaso-occlusive crises associated with multiorgan failure. Here, we developed in vitro and in vivo animal models to mimic endothelial damage during the early phase of hyperhemolysis in SCD. We then used the carbon monoxide (CO)-releasing molecule CORM-401 and examined its effects against endothelial activation, damage, and inflammation inflicted by hemolysates containing red blood cell membrane-derived particles. The in vitro results revealed that CORM-401: (1) prevented the upregulation of relevant proinflammatory and proadhesion markers controlled by the NF-κB enhancer of activated B cells, and (2) abolished the expression of the nuclear factor erythroid-2-related factor 2 (Nrf2) that regulates the inducible antioxidant cell machinery. We also show in SCD mice that CORM-401 protects against hemolysate-induced acute damage of target organs such as the lung, liver, and kidney through modulation of NF-κB proinflammatory and Nrf2 antioxidant pathways. Our data demonstrate the efficacy of CORM-401 as a novel therapeutic agent to counteract hemolysate-induced organ damage during hyperhemolysis in SCD. This approach might be considered as possible preventive treatment in high-risk situations such as patients with SCD with history of DHTR.
Collapse
Affiliation(s)
- Kim Anh Nguyen
- Etablissement Français du Sang, Île-de-France Mondor, Créteil, France
- Laboraroire d'Excellence, INSERM U955, Mondor Biomedical Research Institute, Team Pirenne, Créteil, France
| | - Alessandro Matte
- Department of Medicine, University of Verona and AOUI Verona, Verona, Italy
| | - Roberta Foresti
- Université Paris-Est Créteil, INSERM, Mondor Biomedical Research Institute, Créteil, France
| | - Enrica Federti
- Department of Medicine, University of Verona and AOUI Verona, Verona, Italy
| | - Laurent Kiger
- Laboraroire d'Excellence, INSERM U955, Mondor Biomedical Research Institute, Team Pirenne, Créteil, France
| | - Cécile Lefebvre
- Université Paris-Est Créteil, INSERM, Mondor Biomedical Research Institute, Genomic Platform, Créteil, France
| | - Hakim Hocini
- Université Paris-Est Créteil, INSERM, Mondor Biomedical Research Institute, Genomic Platform, Créteil, France
| | - Yanis Pelinski
- Centre de référence des Syndromes Drépanocytaires Majeurs, Hôpital Henri-Mondor, Assistance Publique Hôpitaux de Paris, Créteil, France
| | - Hiroaki Kitagishi
- Department of Molecular Chemistry and Biochemistry, Faculty of Science and Engineering, Doshisha University, Kyotanabe, Japan
| | - Laura Bencheikh
- Laboraroire d'Excellence, INSERM U955, Mondor Biomedical Research Institute, Team Pirenne, Créteil, France
| | - France Pirenne
- Etablissement Français du Sang, Île-de-France Mondor, Créteil, France
- Laboraroire d'Excellence, INSERM U955, Mondor Biomedical Research Institute, Team Pirenne, Créteil, France
- Université Paris-Est Créteil, Créteil, France
| | | | - Roberto Motterlini
- Université Paris-Est Créteil, INSERM, Mondor Biomedical Research Institute, Créteil, France
| | - Pablo Bartolucci
- Laboraroire d'Excellence, INSERM U955, Mondor Biomedical Research Institute, Team Pirenne, Créteil, France
- Centre de référence des Syndromes Drépanocytaires Majeurs, Hôpital Henri-Mondor, Assistance Publique Hôpitaux de Paris, Créteil, France
- Université Paris-Est Créteil, Créteil, France
| |
Collapse
|
4
|
Alves de Souza RW, Voltarelli V, Gallo D, Shankar S, Tift MS, Young M, Gomperts E, Gomperts A, Otterbein LE. Beneficial Effects of Oral Carbon Monoxide on Doxorubicin-Induced Cardiotoxicity. J Am Heart Assoc 2024; 13:e032067. [PMID: 38700010 PMCID: PMC11179858 DOI: 10.1161/jaha.123.032067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/21/2023] [Indexed: 05/05/2024]
Abstract
BACKGROUND Doxorubicin and other anthracyclines are crucial cancer treatment drugs. However, they are associated with significant cardiotoxicity, severely affecting patient care and limiting dosage and usage. Previous studies have shown that low carbon monoxide (CO) concentrations protect against doxorubicin toxicity. However, traditional methods of CO delivery pose complex challenges for daily administration, such as dosing and toxicity. To address these challenges, we developed a novel oral liquid drug product containing CO (HBI-002) that can be easily self-administered by patients with cancer undergoing doxorubicin treatment, resulting in CO being delivered through the upper gastrointestinal tract. METHODS AND RESULTS HBI-002 was tested in a murine model of doxorubicin cardiotoxicity in the presence and absence of lung or breast cancer. The mice received HBI-002 twice daily before doxorubicin administration and experienced increased carboxyhemoglobin levels from a baseline of ≈1% to 7%. Heart tissue from mice treated with HBI-002 had a 6.3-fold increase in CO concentrations and higher expression of the cytoprotective enzyme heme oxygenase-1 compared with placebo control. In both acute and chronic doxorubicin toxicity scenarios, HBI-002 protected the heart from cardiotoxic effects, including limiting tissue damage and cardiac dysfunction and improving survival. In addition, HBI-002 did not compromise the efficacy of doxorubicin in reducing tumor volume, but rather enhanced the sensitivity of breast 4T1 cancer cells to doxorubicin while simultaneously protecting cardiac function. CONCLUSIONS These findings strongly support using HBI-002 as a cardioprotective agent that maintains the therapeutic benefits of doxorubicin cancer treatment while mitigating cardiac damage.
Collapse
Affiliation(s)
| | - Vanessa Voltarelli
- Department of SurgeryBeth Israel Deaconess Medical Center, Harvard Medical SchoolBostonMAUSA
| | - David Gallo
- Department of SurgeryBeth Israel Deaconess Medical Center, Harvard Medical SchoolBostonMAUSA
| | - Sidharth Shankar
- Department of SurgeryBeth Israel Deaconess Medical Center, Harvard Medical SchoolBostonMAUSA
| | - Michael S. Tift
- Department of Biology and Marine BiologyUniversity of North Carolina WilmingtonWilmingtonNCUSA
| | - Mark Young
- Hillhurst Biopharmaceuticals, lncMontroseCAUSA
| | | | | | - Leo E. Otterbein
- Department of SurgeryBeth Israel Deaconess Medical Center, Harvard Medical SchoolBostonMAUSA
| |
Collapse
|
5
|
Yeudall S, Upchurch CM, Leitinger N. The clinical relevance of heme detoxification by the macrophage heme oxygenase system. Front Immunol 2024; 15:1379967. [PMID: 38585264 PMCID: PMC10995405 DOI: 10.3389/fimmu.2024.1379967] [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: 01/31/2024] [Accepted: 03/11/2024] [Indexed: 04/09/2024] Open
Abstract
Heme degradation by the heme oxygenase (HMOX) family of enzymes is critical for maintaining homeostasis and limiting heme-induced tissue damage. Macrophages express HMOX1 and 2 and are critical sites of heme degradation in healthy and diseased states. Here we review the functions of the macrophage heme oxygenase system and its clinical relevance in discrete groups of pathologies where heme has been demonstrated to play a driving role. HMOX1 function in macrophages is essential for limiting oxidative tissue damage in both acute and chronic hemolytic disorders. By degrading pro-inflammatory heme and releasing anti-inflammatory molecules such as carbon monoxide, HMOX1 fine-tunes the acute inflammatory response with consequences for disorders of hyperinflammation such as sepsis. We then discuss divergent beneficial and pathological roles for HMOX1 in disorders such as atherosclerosis and metabolic syndrome, where activation of the HMOX system sits at the crossroads of chronic low-grade inflammation and oxidative stress. Finally, we highlight the emerging role for HMOX1 in regulating macrophage cell death via the iron- and oxidation-dependent form of cell death, ferroptosis. In summary, the importance of heme clearance by macrophages is an active area of investigation with relevance for therapeutic intervention in a diverse array of human diseases.
Collapse
Affiliation(s)
- Scott Yeudall
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States
- Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Clint M. Upchurch
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States
- Robert M Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
6
|
Beckman JD, Sparkenbaugh EM. The invisible string of coagulation, complement, iron, and inflammation in sickle cell disease. Curr Opin Hematol 2023; 30:153-158. [PMID: 37462409 PMCID: PMC10529498 DOI: 10.1097/moh.0000000000000773] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
PURPOSE OF REVIEW This review provides an update on recent advances in mechanistic studies of thromboinflammatory mechanisms that contribute to the disease pathology in sickle cell disease (SCD). There is a focus on novel pathways, clinical relevance, and translational potential of these findings. We hope to encourage more advances in this area to reduce organ damage in young patients prior to gene therapy, and to serve the aging SCD patient population. RECENT FINDINGS Novel insights into the roles of neutrophils, the ADAMTS-13/VWF axis, oxidative stress, and the intrinsic coagulation cascade, as well as relevant clinical trials, are discussed. SUMMARY Several studies implicate dysregulation of the ADAMTS-13/VWF axis as playing a major role in vaso-occlusive events (VOE) in SCD. Another highlight is reducing iron overload, which has beneficial effects on erythrocyte and neutrophil function that reduce VOE and inflammation. Multiple studies suggest that targeting HO-1/ROS in erythrocytes, platelets, and endothelium can attenuate disease pathology. New insights into coagulation activation identify intrinsic coagulation factor XII as a central regulator of many thromboinflammatory pathologies in SCD. The complement cascade and modulators of neutrophil function and release of neutrophil extracellular traps are also discussed.
Collapse
Affiliation(s)
- Joan D Beckman
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Erica M Sparkenbaugh
- Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Blood Research Center, Chapel Hill, North Carolina, USA
| |
Collapse
|
7
|
Chauhan W, Zennadi R. Keap1-Nrf2 Heterodimer: A Therapeutic Target to Ameliorate Sickle Cell Disease. Antioxidants (Basel) 2023; 12:antiox12030740. [PMID: 36978988 PMCID: PMC10045360 DOI: 10.3390/antiox12030740] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/04/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Sickle cell disease (SCD) is a monogenic inheritable disease characterized by severe anemia, increased hemolysis, and recurrent, painful vaso-occlusive crises due to the polymerization of hemoglobin S (HbS)-generated oxidative stress. Up until now, only four drugs are approved for SCD in the US. However, each of these drugs affects only a limited array of SCD pathologies. Importantly, curative therapies, such as gene therapy, or hematopoietic stem cell transplantation are not available for every patient because of their high costs, availability of donor matching, and their serious adverse effects. Therefore, there is an unmet medical need for novel therapeutic strategies that target broader SCD sequelae. SCD phenotypic severity can be alleviated by increasing fetal hemoglobin (HbF) expression. This results in the inhibition of HbS polymerization and thus sickling, and a reduction in oxidative stress. The efficacy of HbF is due to its ability to dilute HbS levels below the threshold required for polymerization and to influence HbS polymer stability in RBCs. Nuclear factor-E2-related factor 2 (Nrf2)/Kelch-like ECH-associated protein-1 (Keap1)-complex signaling is one of the most important cytoprotective signaling controlling oxidative stress. Nrf2 is present in most organs and, after dissociation from Keap1, it accumulates in the cytoplasm, then translocates to the nucleus where it binds to the antioxidant response element (ARE) sequences and increases the expression of various cytoprotective antioxidant genes. Keeping this in mind, various researchers have proposed a role of multiple agents, more importantly tert-Butylhydroquinone (tBHQ), curcumin, etc., (having electrophilic properties) in inhibiting keap1 activity, so that Nrf2 can translocate to the nucleus to activate the gamma globin gene, thus maintaining alpha-hemoglobin-stabilizing protein (AHSP) and HbF levels. This leads to reduced oxidative stress, consequently minimizing SCD-associated complications. In this review, we will discuss the role of the Keap-1–Nrf2 complex in hemoglobinopathies, especially in SCD, and how this complex might represent a better target for more effective treatment options.
Collapse
|
8
|
Sun R, Tian X, Li Y, Zhao Y, Wang Z, Hu Y, Zhang L, Wang Y, Gao D, Zheng S, Yao J. The m6A reader YTHDF3-mediated PRDX3 translation alleviates liver fibrosis. Redox Biol 2022; 54:102378. [PMID: 35779442 PMCID: PMC9287738 DOI: 10.1016/j.redox.2022.102378] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/15/2022] [Accepted: 06/19/2022] [Indexed: 01/07/2023] Open
Affiliation(s)
- Ruimin Sun
- Department of Pharmacology, Dalian Medical University, Dalian, China; Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xinyao Tian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Hepatobiliary and Pancreatic Surgery, Department of Liver Transplantation, Shulan (Hangzhou) Hospital, Hangzhou, China
| | - Yang Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Zhao
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Zhecheng Wang
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Yan Hu
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Lijun Zhang
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Yue Wang
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Dongyan Gao
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Hepatobiliary and Pancreatic Surgery, Department of Liver Transplantation, Shulan (Hangzhou) Hospital, Hangzhou, China.
| | - Jihong Yao
- Department of Pharmacology, Dalian Medical University, Dalian, China.
| |
Collapse
|
9
|
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
|
10
|
Chen H, Qin J, Shi H, Li Q, Zhou S, Chen L. Rhoifolin ameliorates osteoarthritis via the Nrf2/NF-κB axis: in vitro and in vivo experiments. Osteoarthritis Cartilage 2022; 30:735-745. [PMID: 35139424 DOI: 10.1016/j.joca.2022.01.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 12/22/2021] [Accepted: 01/21/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is an age-related degenerative disease accompanied by an increasing number of senescent cells and chronic low-grade inflammation. Rhoifolin (ROF) showed considerable inhibition to inflammation, but its role in chondrocyte senescence and OA progress has not been fully characterized. We aimed to evaluate the protective effects of ROF on OA through a series of in vitro and in vivo experiments. METHODS The role of ROF in the expression of senescence-associated secretory phenotype (SASP) factors was investigated using RT-qPCR, western blotting, and ELISA. Chondrocyte senescence was assessed by SA-β-gal staining. We applied molecular docking to screen candidate proteins regulated by ROF. Meanwhile, SASP factors and cellular senescence were further assessed after the transfection of Nrf2 siRNA. In the anterior cruciate ligament transection (ACLT) rat model, X-ray, hematoxylin-eosin (HE), and Masson's staining were performed to evaluate the therapeutic effects of ROF on OA. RESULTS We found that ROF inhibited SASP factors expression and senescence phenotype in IL-1β-treated chondrocytes. Furthermore, ROF suppressed IL-1β-induced activation of the NF-κB pathway cascades. Also, molecular docking and knock-down studies demonstrated that ROF might bind to Nrf2 to suppress the NF-κB pathway. In vivo, ROF ameliorated the OA process in the ACLT rat model. CONCLUSIONS ROF inhibits SASP factors expression and senescence phenotype in chondrocytes and ameliorates the progression of OA via the Nrf2/NF-κB axis, which supports ROF as a potential therapeutic agent for the treatment of OA.
Collapse
Affiliation(s)
- H Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - J Qin
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - H Shi
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Q Li
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - S Zhou
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - L Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
11
|
Faggiano S, Ronda L, Bruno S, Abbruzzetti S, Viappiani C, Bettati S, Mozzarelli A. From hemoglobin allostery to hemoglobin-based oxygen carriers. Mol Aspects Med 2021; 84:101050. [PMID: 34776270 DOI: 10.1016/j.mam.2021.101050] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/05/2021] [Accepted: 11/05/2021] [Indexed: 12/18/2022]
Abstract
Hemoglobin (Hb) plays its vital role through structural and functional properties evolutionarily optimized to work within red blood cells, i.e., the tetrameric assembly, well-defined oxygen affinity, positive cooperativity, and heterotropic allosteric regulation by protons, chloride and 2,3-diphosphoglycerate. Outside red blood cells, the Hb tetramer dissociates into dimers, which exhibit high oxygen affinity and neither cooperativity nor allosteric regulation. They are prone to extravasate, thus scavenging endothelial NO and causing hypertension, and cause nephrotoxicity. In addition, they are more prone to autoxidation, generating radicals. The need to overcome the adverse effects associated with cell-free Hb has always been a major hurdle in the development of substitutes of allogeneic blood transfusions for all clinical situations where blood is unavailable or cannot be used due to, for example, religious objections. This class of therapeutics, indicated as hemoglobin-based oxygen carriers (HBOCs), is formed by genetically and/or chemically modified Hbs. Many efforts were devoted to the exploitation of the wealth of biochemical and biophysical information available on Hb structure, function, and dynamics to design safe HBOCs, overcoming the negative effects of free plasma Hb. Unfortunately, so far, no HBOC has been approved by FDA and EMA, except for compassionate use. However, the unmet clinical needs that triggered intensive investigations more than fifty years ago are still awaiting an answer. Recently, HBOCs "repositioning" has led to their successful application in organ perfusion fluids.
Collapse
Affiliation(s)
- Serena Faggiano
- Department of Food and Drug, University of Parma, Parma, Italy; Institute of Biophysics, National Research Council, Pisa, Italy
| | - Luca Ronda
- Institute of Biophysics, National Research Council, Pisa, Italy; Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Stefano Bruno
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Stefania Abbruzzetti
- Department of Mathematical, Physical and Computer Sciences, University of Parma, Parma, Italy
| | - Cristiano Viappiani
- Department of Mathematical, Physical and Computer Sciences, University of Parma, Parma, Italy
| | - Stefano Bettati
- Institute of Biophysics, National Research Council, Pisa, Italy; Department of Medicine and Surgery, University of Parma, Parma, Italy; National Institute of Biostructures and Biosystems, Rome, Italy
| | - Andrea Mozzarelli
- Department of Food and Drug, University of Parma, Parma, Italy; Institute of Biophysics, National Research Council, Pisa, Italy.
| |
Collapse
|
12
|
Carbon Monoxide-Saturated Polymerized Placenta Hemoglobin Optimizes Mitochondrial Function and Protects Heart Against Ischemia-Reperfusion Injury. J Cardiovasc Pharmacol 2021; 77:814-821. [PMID: 34001725 DOI: 10.1097/fjc.0000000000001022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/05/2021] [Indexed: 02/05/2023]
Abstract
ABSTRACT Ischemia-reperfusion (I-R) injury is detrimental to cardiovascular system. This study was designed to investigate whether carbon monoxide-saturated polymerized human placenta hemoglobin (CO-PolyPHb) attenuates cardiac I-R injury and to elucidate the underlying mechanism(s). Sixty male adult Sprague-Dawley rats were randomly divided into 6 groups: saline + sham group, PolyPHb + sham group, CO-PolyPHb + sham group, saline + I-R group, PolyPHb + I-R group, and CO-PolyPHb + I-R group. Rats were pretreated with injection of PolyPHb, CO-PolyPHb (0.5 g Hb/kg/d), or an equivalent volume of saline via caudal vein for 3 days. After pretreatment, hearts were isolated Langendorff perfused and subjected to 30-minute no-flow ischemia and 120-minute reperfusion. As compared with the saline + I-R group, pretreatment with CO-PolyPHb greatly improved the recovery of cardiac function, reduced infarct size, and suppressed the release of cardiac enzyme. Importantly, CO-PolyPHb showed more prominent cardioprotective effect than PolyPHb, exhibiting a promising therapeutic potential in cardiac I-R injury. Further study demonstrated that CO-PolyPHb activated molecular signaling toward mitophagy and significantly elevated the mitochondrial respiratory function in the heart. In addition, CO-PolyPHb upregulated the phosphorylation of the proteins in insulin signaling pathway and increased the glucose uptake rate in cardiomyocytes. Pharmacological inhibition of this pathway by wortmannin abrogated the anti-I-R effect of CO-PolyPHb. In conclusion, using an isolated rat heart model, we have demonstrated that pretreatment with CO-PolyPHb provided protective effect against cardiac I-R injury, and this protection was mediated by the improvement of mitochondrial function and activation of insulin signaling pathway in the heart.
Collapse
|
13
|
Nader E, Conran N, Romana M, Connes P. Vasculopathy in Sickle Cell Disease: From Red Blood Cell Sickling to Vascular Dysfunction. Compr Physiol 2021; 11:1785-1803. [PMID: 33792905 DOI: 10.1002/cphy.c200024] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Sickle cell disease (SCD) is a hereditary disorder that leads to the production of an abnormal hemoglobin, hemoglobin S (HbS). HbS polymerizes in deoxygenated conditions, which can prompt red blood cell (RBC) sickling and leaves the RBCs more rigid, fragile, and prone to hemolysis. SCD patients suffer from a plethora of complications, ranging from acute complications, such as characteristic, frequent, and debilitating vaso-occlusive episodes to chronic organ damage. While RBC sickling is the primary event at the origin of vaso-occlusive processes, other factors that can further increase RBC transit times in the microcirculation may also be required to precipitate vaso-occlusive processes. The adhesion of RBC and leukocytes to activated endothelium and the formation of heterocellular aggregates, as well as increased blood viscosity, are among the mechanisms involved in slowing the progress of RBCs in deoxygenated vascular areas, favoring RBC sickling and promoting vascular occlusion. Chronic inflammatory processes and oxidative stress, which are perpetuated by hemolytic events and ischemia-reperfusion injury, result in this pan cellular activation and some acute events, such as stroke and acute chest syndrome, as well as chronic end-organ damage. Furthermore, impaired vasodilation and vasomotor hyperresponsiveness in SCD also contribute to vaso-occlusive processes. Treating SCD as a vascular disease in addition to its hematological perspective, the present article looks at the interplay between abnormal RBC physiology/integrity, vascular dysfunction and clinical severity in SCD, and discusses existing therapies and novel drugs in development that may ameliorate vascular complications in the disease. © 2021 American Physiological Society. Compr Physiol 11:1785-1803, 2021.
Collapse
Affiliation(s)
- Elie Nader
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team Vascular Biology and Red Blood Cell, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
| | - Nicola Conran
- Hematology Center, University of Campinas - UNICAMP, Cidade Universitária, Campinas-SP, Brazil
| | - Marc Romana
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France.,Université des Antilles, UMR_S1134, BIGR, Pointe-à-Pitre, France.,Université de Paris, UMR_S1134, BIGR, INSERM, Paris, France
| | - Philippe Connes
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team Vascular Biology and Red Blood Cell, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
| |
Collapse
|
14
|
Macrophage metabolic adaptation to heme detoxification involves CO-dependent activation of the pentose phosphate pathway. Blood 2021; 136:1535-1548. [PMID: 32556090 DOI: 10.1182/blood.2020004964] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/08/2020] [Indexed: 02/07/2023] Open
Abstract
Heme is an essential cofactor for numerous cellular functions, but release of free heme during hemolysis results in oxidative tissue damage, vascular dysfunction, and inflammation. Macrophages play a key protective role in heme clearance; however, the mechanisms that regulate metabolic adaptations that are required for effective heme degradation remain unclear. Here we demonstrate that heme loading drives a unique bioenergetic switch in macrophages, which involves a metabolic shift from oxidative phosphorylation toward glucose consumption. Metabolomic and transcriptional analysis of heme-loaded macrophages revealed that glucose is funneled into the pentose phosphate pathway (PPP), which is indispensable for efficient heme detoxification and is required to maintain redox homeostasis. We demonstrate that the metabolic shift to the PPP is controlled by heme oxygenase-dependent generation of carbon monoxide (CO). Finally, we show that PPP upregulation occurs in vivo in organ systems central to heme clearance and that PPP activity correlates with heme levels in mouse sickle cell disease (SCD). Together, our findings demonstrate that metabolic adaptation to heme detoxification in macrophages requires a shift to the PPP that is induced by heme-derived CO, suggesting pharmacologic targeting of macrophage metabolism as a novel therapeutic strategy to improve heme clearance in patients with hemolytic disorders.
Collapse
|
15
|
Vinchi F, Sparla R, Passos ST, Sharma R, Vance SZ, Zreid HS, Juaidi H, Manwani D, Yazdanbakhsh K, Nandi V, Silva AMN, Agarvas AR, Fibach E, Belcher JD, Vercellotti GM, Ghoti H, Muckenthaler MU. Vasculo-toxic and pro-inflammatory action of unbound haemoglobin, haem and iron in transfusion-dependent patients with haemolytic anaemias. Br J Haematol 2021; 193:637-658. [PMID: 33723861 PMCID: PMC8252605 DOI: 10.1111/bjh.17361] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Abstract
Increasing evidence suggests that free haem and iron exert vasculo‐toxic and pro‐inflammatory effects by activating endothelial and immune cells. In the present retrospective study, we compared serum samples from transfusion‐dependent patients with β‐thalassaemia major and intermedia, hereditary spherocytosis and sickle cell disease (SCD). Haemolysis, transfusions and ineffective erythropoiesis contribute to haem and iron overload in haemolytic patients. In all cohorts we observed increased systemic haem and iron levels associated with scavenger depletion and toxic ‘free’ species formation. Endothelial dysfunction, oxidative stress and inflammation markers were significantly increased compared to healthy donors. In multivariable logistic regression analysis, oxidative stress markers remained significantly associated with both haem‐ and iron‐related parameters, while soluble vascular cell adhesion molecule 1 (sVCAM‐1), soluble endothelial selectin (sE‐selectin) and tumour necrosis factor α (TNFα) showed the strongest association with haem‐related parameters and soluble intercellular adhesion molecule 1 (sICAM‐1), sVCAM‐1, interleukin 6 (IL‐6) and vascular endothelial growth factor (VEGF) with iron‐related parameters. While hereditary spherocytosis was associated with the highest IL‐6 and TNFα levels, β‐thalassaemia major showed limited inflammation compared to SCD. The sVCAM1 increase was significantly lower in patients with SCD receiving exchange compared to simple transfusions. The present results support the involvement of free haem/iron species in the pathogenesis of vascular dysfunction and sterile inflammation in haemolytic diseases, irrespective of the underlying haemolytic mechanism, and highlight the potential therapeutic benefit of iron/haem scavenging therapies in these conditions.
Collapse
Affiliation(s)
- Francesca Vinchi
- Iron Research Program, New York Blood Center, New York, NY, USA.,Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA.,Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory (EMBL), Heidelberg University, Heidelberg, Germany
| | - Richard Sparla
- Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, University of Heidelberg, Heidelberg, Germany
| | - Sara T Passos
- Iron Research Program, New York Blood Center, New York, NY, USA
| | - Richa Sharma
- Iron Research Program, New York Blood Center, New York, NY, USA
| | - S Zebulon Vance
- Iron Research Program, New York Blood Center, New York, NY, USA
| | - Hala S Zreid
- Department of Internal Medicine, Al Shifa Hospital, Gaza, Palestine
| | - Hesham Juaidi
- Department of Internal Medicine, Al Shifa Hospital, Gaza, Palestine
| | - Deepa Manwani
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA.,Pediatric Hematology, The Children's Hospital at Montefiore, New York, NY, USA
| | | | - Vijay Nandi
- Laboratory of Data Analytic Services, New York Blood Center, New York, NY, USA
| | - André M N Silva
- REQUIMTE-LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências, University of Porto, Porto, Portugal
| | - Anand R Agarvas
- Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, University of Heidelberg, Heidelberg, Germany
| | - Eitan Fibach
- Department of Hematology, The Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - John D Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
| | - Gregory M Vercellotti
- Department of Medicine, Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
| | - Husam Ghoti
- European Center for Cancer and Cell Therapy (ECCT), Nicosia, Cyprus
| | - Martina U Muckenthaler
- Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory (EMBL), Heidelberg University, Heidelberg, Germany.,Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, University of Heidelberg, Heidelberg, Germany.,German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg, Germany.,Translational Lung Research Center (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
16
|
Coll-Satue C, Bishnoi S, Chen J, Hosta-Rigau L. Stepping stones to the future of haemoglobin-based blood products: clinical, preclinical and innovative examples. Biomater Sci 2021; 9:1135-1152. [DOI: 10.1039/d0bm01767a] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Critical overview of the different oxygen therapeutics developed so far to be used when donor blood is not available.
Collapse
Affiliation(s)
- Clara Coll-Satue
- Department of Health Technology
- Centre for Nanomedicine and Theranostics
- DTU Health Tech
- Technical University of Denmark
- 2800 Lyngby
| | - Shahana Bishnoi
- Department of Health Technology
- Centre for Nanomedicine and Theranostics
- DTU Health Tech
- Technical University of Denmark
- 2800 Lyngby
| | - Jiantao Chen
- Department of Health Technology
- Centre for Nanomedicine and Theranostics
- DTU Health Tech
- Technical University of Denmark
- 2800 Lyngby
| | - Leticia Hosta-Rigau
- Department of Health Technology
- Centre for Nanomedicine and Theranostics
- DTU Health Tech
- Technical University of Denmark
- 2800 Lyngby
| |
Collapse
|
17
|
Gáll T, Pethő D, Nagy A, Balla G, Balla J. Therapeutic Potential of Carbon Monoxide (CO) and Hydrogen Sulfide (H 2S) in Hemolytic and Hemorrhagic Vascular Disorders-Interaction between the Heme Oxygenase and H 2S-Producing Systems. Int J Mol Sci 2020; 22:ijms22010047. [PMID: 33374506 PMCID: PMC7793096 DOI: 10.3390/ijms22010047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 02/07/2023] Open
Abstract
Over the past decades, substantial work has established that hemoglobin oxidation and heme release play a pivotal role in hemolytic/hemorrhagic disorders. Recent reports have shown that oxidized hemoglobins, globin-derived peptides, and heme trigger diverse biological responses, such as toll-like receptor 4 activation with inflammatory response, reprogramming of cellular metabolism, differentiation, stress, and even death. Here, we discuss these cellular responses with particular focus on their mechanisms that are linked to the pathological consequences of hemorrhage and hemolysis. In recent years, endogenous gasotransmitters, such as carbon monoxide (CO) and hydrogen sulfide (H2S), have gained a lot of interest in connection with various human pathologies. Thus, many CO and H2S-releasing molecules have been developed and applied in various human disorders, including hemolytic and hemorrhagic diseases. Here, we discuss our current understanding of oxidized hemoglobin and heme-induced cell and tissue damage with particular focus on inflammation, cellular metabolism and differentiation, and endoplasmic reticulum stress in hemolytic/hemorrhagic human diseases, and the potential beneficial role of CO and H2S in these pathologies. More detailed mechanistic insights into the complex pathology of hemolytic/hemorrhagic diseases through heme oxygenase-1/CO as well as H2S pathways would reveal new therapeutic approaches that can be exploited for clinical benefit.
Collapse
Affiliation(s)
- Tamás Gáll
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.G.); (D.P.); (A.N.)
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4032 Debrecen, Hungary;
| | - Dávid Pethő
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.G.); (D.P.); (A.N.)
- Faculty of Medicine, University of Debrecen, Kálmán Laki Doctoral School, 4032 Debrecen, Hungary
| | - Annamária Nagy
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.G.); (D.P.); (A.N.)
- Faculty of Medicine, University of Debrecen, Kálmán Laki Doctoral School, 4032 Debrecen, Hungary
| | - György Balla
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4032 Debrecen, Hungary;
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - József Balla
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.G.); (D.P.); (A.N.)
- Correspondence: ; Tel.: +36-52-255-500/55004
| |
Collapse
|
18
|
Hebbel RP, Belcher JD, Vercellotti GM. The multifaceted role of ischemia/reperfusion in sickle cell anemia. J Clin Invest 2020; 130:1062-1072. [PMID: 32118586 DOI: 10.1172/jci133639] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sickle cell anemia is a unique disease dominated by hemolytic anemia and vaso-occlusive events. The latter trigger a version of ischemia/reperfusion (I/R) pathobiology that is singular in its origin, cyclicity, complexity, instability, perpetuity, and breadth of clinical consequences. Specific clinical features are probably attributable to local I/R injury (e.g., stroke syndromes) or remote organ injury (e.g., acute chest syndrome) or the systematization of inflammation (e.g., multifocal arteriopathy). Indeed, by fashioning an underlying template of endothelial dysfunction and vulnerability, the robust inflammatory systematization no doubt contributes to all sickle pathology. In this Review, we highlight I/R-targeting therapeutics shown to improve microvascular blood flow in sickle transgenic mice undergoing I/R, and we suggest how such insights might be translated into human therapeutic strategies.
Collapse
|
19
|
Taguchi K, Maruyama T, Otagiri M. Use of Hemoglobin for Delivering Exogenous Carbon Monoxide in Medicinal Applications. Curr Med Chem 2020; 27:2949-2963. [PMID: 30421669 DOI: 10.2174/0929867325666181113122340] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 09/25/2018] [Accepted: 11/09/2018] [Indexed: 01/02/2023]
Abstract
Carbon Monoxide (CO), at low concentrations, can have a variety of positive effects on the body including anti-apoptosis, anti-inflammatory, anti-oxidative and anti-proliferative effects. Although CO has great potential for use as a potent medical bioactive gas, for it to exist in the body in stable form, it must be associated with a carrier. Hemoglobin (Hb) represents a promising material for use as a CO carrier because most of the total CO in the body is stored associated with Hb in red blood cells (RBC). Attempts have been made to develop an Hb-based CO carrying system using RBC and Hb-based artificial oxygen carriers. Some of these have been reported to be safe and to have therapeutic value as a CO donor in preclinical and clinical studies. In the present review, we overview the potential of RBC and Hb-based artificial oxygen carriers as CO carriers based on the currently available literature evidence for their use in pharmaceutical therapy against intractable disorders.
Collapse
Affiliation(s)
- Kazuaki Taguchi
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan.,DDS Research Institute, Sojo University, Kumamoto, Japan
| |
Collapse
|
20
|
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
|
21
|
Abstract
Sickle cell disease (SCD) afflicts millions of people worldwide but is referred to as an orphan disease in the United States. Over the past several decades, there has been an increasing understanding of the pathophysiology of SCD and its complications. While most individuals with SCD in resource-rich countries survive into adulthood, the life expectancy of patients with SCD remains substantially shorter than for the general African-American population. SCD can be cured using hematopoietic stem cell transplantation and possibly gene therapy, but these treatment approaches are not available to most patients, the majority of whom reside in low- and middle-income countries. Until relatively recently, only one drug, hydroxyurea, was approved by the US Food and Drug Administration to ameliorate disease severity. Multiple other drugs (L-glutamine, crizanlizumab, and voxelotor) have recently been approved for the treatment of SCD, with several others at various stages of clinical testing. The availability of multiple agents to treat SCD raises questions related to the choice of appropriate drug therapy, combination of multiple agents, and affordability of recently approved products. The enthusiasm for new drug development provides opportunities to involve patients in low- and middle-income nations in the testing of potentially disease-modifying therapies and has the potential to contribute to capacity building in these environments. Demonstration that these agents, alone or in combination, can prevent or decrease end-organ damage would provide additional evidence for the role of drug therapies in improving outcomes in SCD.
Collapse
Affiliation(s)
- Parul Rai
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kenneth I. Ataga
- Center for Sickle Cell Disease, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
22
|
Khan F, Singh K, Friedman MT. Artificial Blood: The History and Current Perspectives of Blood Substitutes. Discoveries (Craiova) 2020; 8:e104. [PMID: 32309621 PMCID: PMC7086064 DOI: 10.15190/d.2020.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/07/2020] [Accepted: 03/07/2020] [Indexed: 01/09/2023] Open
Abstract
Blood transfusions are one of the most common procedures performed in hospitalized patients. Yet, despite all of the measures taken to ensure the safety of the blood supply, there are known risks associated with transfusions, including infectious and noninfectious complications. Meanwhile, issues with blood product availability, the need for compatibility testing, and the storage and transport requirements of blood products, have presented challenges for the administration of blood transfusions. Additionally, there are individuals who do not accept blood transfusions (e.g., Jehovah's Witnesses). Therefore, there is a need to develop alternative agents that can reliably and safely replace blood. However, although there have been many attempts to develop blood substitutes over the years, there are currently no such products available that have been approved by the United States Food and Drug Administration (FDA). However, a more-recently developed hemoglobin-based oxygen carrier has shown promise in early clinical trials and has achieved the status of "Orphan Drug" under the FDA.
Collapse
Affiliation(s)
- Fahad Khan
- Mount Sinai Health System, Department of Pathology and Laboratory Medicine, Icahn School of Medicine, New York, NY, USA
| | - Kunwar Singh
- Mount Sinai Health System, Department of Pathology and Laboratory Medicine, Icahn School of Medicine, New York, NY, USA
| | - Mark T. Friedman
- Mount Sinai Health System, Department of Pathology and Laboratory Medicine, Icahn School of Medicine, New York, NY, USA
| |
Collapse
|
23
|
Advances in the molecular mechanisms of NLRP3 inflammasome activators and inactivators. Biochem Pharmacol 2020; 175:113863. [PMID: 32081791 DOI: 10.1016/j.bcp.2020.113863] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/13/2020] [Indexed: 12/15/2022]
Abstract
NLRP3 inflammasome is an intracellular protein complex that initiates cellular injury via assembly of NLRP3, ASC and caspase-1 in response to microbial infection and sterile stressors. The importance of NLRP3 inflammasome in immunity and human diseases has been well documented. Up to now, targeted inhibition of the assembly of NLRP3 inflammasome complex and of its activation was thought to be therapeutic strategy for associated diseases. Recent studies show that a host of molecules such as NIMA-related kinase 7 (Nek7) and DEAD-box helicase 3 X-linked (DDX3X) and a large number of biological mediators including cytokines, microRNAs, nitric oxide, carbon monoxide, nuclear factor erythroid-2 related factor 2 (Nrf2) and cellular autophagy participate in the activation and inactivation of NLRP3 inflammasome. This review summarizes current understanding of the molecular basis of NLRP3 inflammasome activation and inactivation. This knowledge may lead to development of new therapies directed at NLRP3 inflammasome related diseases.
Collapse
|
24
|
Ballas SK. The Evolving Pharmacotherapeutic Landscape for the Treatment of Sickle Cell Disease. Mediterr J Hematol Infect Dis 2020; 12:e2020010. [PMID: 31934320 PMCID: PMC6951351 DOI: 10.4084/mjhid.2020.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 12/17/2019] [Indexed: 01/02/2023] Open
Abstract
Sickle cell disease (SCD) is an extremely heterogeneous disease that has been associated with global morbidity and early mortality. More effective and inexpensive therapies are needed. During the last five years, the landscape of the pharmacotherapy of SCD has changed dramatically. Currently, 54 drugs have been used or under consideration to use for the treatment of SCD. These fall into 3 categories: the first category includes the four drugs (Hydroxyurea, L-Glutamine, Crizanlizumab tmca and Voxelotor) that have been approved by the United States Food and Drug Administration (FDA) based on successful clinical trials. The second category includes 22 drugs that failed, discontinued or terminated for now and the third category includes 28 drugs that are actively being considered for the treatment of SCD. Crizanlizumab and Voxelotor are included in the first and third categories because they have been used in more than one trial. New therapies targeting multiple pathways in the complex pathophysiology of SCD have been achieved or are under continued investigation. The emerging trend seems to be the use of multimodal drugs (i.e. drugs that have different mechanisms of action) to treat SCD similar to the use of multiple chemotherapeutic agents to treat cancer.
Collapse
Affiliation(s)
- Samir K Ballas
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
25
|
Adebiyi MG, Zhao Z, Ye Y, Manalo J, Hong Y, Lee CC, Xian W, McKeon F, Culp-Hill R, D' Alessandro A, Kellems RE, Yoo SH, Han L, Xia Y. Circadian period 2: a missing beneficial factor in sickle cell disease by lowering pulmonary inflammation, iron overload, and mortality. FASEB J 2019; 33:10528-10537. [PMID: 31260634 DOI: 10.1096/fj.201900246rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The circadian clock is important for cellular and organ function. However, its function in sickle cell disease (SCD), a life-threatening hemolytic disorder, remains unknown. Here, we performed an unbiased microarray screen, which revealed significantly altered expression of circadian rhythmic genes, inflammatory response genes, and iron metabolic genes in SCD Berkeley transgenic mouse lungs compared with controls. Given the vital role of period 2 (Per2) in the core clock and the unrecognized role of Per2 in SCD, we transplanted the bone marrow (BM) of SCD mice to Per2Luciferase mice, which revealed that Per2 expression was up-regulated in SCD mouse lung. Next, we transplanted the BM of SCD mice to period 1 (Per1)/Per2 double deficient [Per1/Per2 double knockout (dKO)] and wild-type mice, respectively. We discovered that Per1/Per2 dKO mice transplanted with SCD BM (SCD → Per1/Per2 dKO) displayed severe irradiation sensitivity and were more susceptible to an early death. Although we observed an increase of peripheral inflammatory cells, we did not detect differences in erythrocyte sickling. However, there was further lung damage due to elevated pulmonary congestion, inflammatory cell infiltration, iron overload, and secretion of IL-6 in lavage fluid. Overall, we demonstrate that Per1/Per2 is beneficial to counteract elevated systemic inflammation, lung tissue inflammation, and iron overload in SCD.-Adebiyi, M. G., Zhao, Z., Ye, Y., Manalo, J., Hong, Y., Lee, C. C., Xian, W., McKeon, F., Culp-Hill, R., D' Alessandro, A., Kellems, R. E., Yoo, S.-H., Han, L., Xia, Y. Circadian period 2: a missing beneficial factor in sickle cell disease by lowering pulmonary inflammation, iron overload, and mortality.
Collapse
Affiliation(s)
- Morayo G Adebiyi
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Zhaoyang Zhao
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Youqiong Ye
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Jeanne Manalo
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Yue Hong
- Department of Biology and Biochemistry, The University of Houston, Houston, Texas, USA
| | - Cheng Chi Lee
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Wa Xian
- The Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Frank McKeon
- Department of Biology and Biochemistry, The University of Houston, Houston, Texas, USA
| | - Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Angelo D' Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rodney E Kellems
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Leng Han
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA.,The Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, USA.,The Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| |
Collapse
|
26
|
Ferenz KB, Steinbicker AU. Artificial Oxygen Carriers-Past, Present, and Future-a Review of the Most Innovative and Clinically Relevant Concepts. J Pharmacol Exp Ther 2019; 369:300-310. [PMID: 30837280 DOI: 10.1124/jpet.118.254664] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 02/12/2019] [Indexed: 12/31/2022] Open
Abstract
Blood transfusions are a daily practice in hospitals. Since these products are limited in availability and have various, harmful side effects, researchers have pursued the goal to develop artificial blood components for about 40 years. Development of oxygen therapeutics and stem cells are more recent goals. Medline (https://www.ncbi.nlm.nih.gov/pubmed/?holding=ideudelib), ClinicalTrials.gov (https://clinicaltrials.gov), EU Clinical Trials Register (https://www.clinicaltrialsregister.eu), and Australian New Zealand Clinical Trials Registry (http://www.anzctr.org.au) were searched up to July 2018 using search terms related to artificial blood products in order to identify new and ongoing research over the last 5 years. However, for products that are already well known and important to or relevant in gaining a better understanding of this field of research, the reader is punctually referred to some important articles published over 5 years ago. This review includes not only clinically relevant substances such as heme-oxygenating carriers, perfluorocarbon-based oxygen carriers, stem cells, and organ conservation, but also includes interesting preclinically advanced compounds depicting the pipeline of potential new products. In- depth insights into specific benefits and limitations of each substance, including the biochemical and physiologic background are included. "Fancy" ideas such as iron-based substances, O2 microbubbles, cyclodextranes, or lugworms are also elucidated. To conclude, this systematic up-to-date review includes all actual achievements and ongoing clinical trials in the field of artificial blood products to pursue the dream of artificial oxygen carrier supply. Research is on the right track, but the task is demanding and challenging.
Collapse
Affiliation(s)
- Katja B Ferenz
- Institute of Physiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.B.F.); and Department of Anesthesiology, Intensive Care and Pain Medicine, Westphalian Wilhelminian University Muenster, University Hospital Muenster, Muenster, Germany (A.U.S.)
| | - Andrea U Steinbicker
- Institute of Physiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.B.F.); and Department of Anesthesiology, Intensive Care and Pain Medicine, Westphalian Wilhelminian University Muenster, University Hospital Muenster, Muenster, Germany (A.U.S.)
| |
Collapse
|
27
|
Vercellotti GM, Dalmasso AP, Schaid TR, Nguyen J, Chen C, Ericson ME, Abdulla F, Killeen T, Lindorfer MA, Taylor RP, Belcher JD. Critical role of C5a in sickle cell disease. Am J Hematol 2019; 94:327-337. [PMID: 30569594 DOI: 10.1002/ajh.25384] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/16/2018] [Accepted: 12/10/2018] [Indexed: 12/31/2022]
Abstract
Innate immune complement activation may contribute to sickle cell disease (SCD) pathogenesis. Ischemia-reperfusion physiology is a key component of the inflammatory and vaso-occlusive milieu in SCD and is associated with complement activation. C5a is an anaphylatoxin, a potent pro-inflammatory mediator that can activate leukocytes, platelets, and endothelial cells, all of which play a role in vaso-occlusion. We hypothesize that hypoxia-reoxygenation (H/R) in SCD mice activates complement, promoting inflammation and vaso-occlusion. At baseline and after H/R, sickle Townes-SS mice had increased C3 activation fragments and C5b-9 deposition in kidneys, livers and lungs and alternative pathway Bb fragments in plasma compared to control AA-mice. Activated complement promoted vaso-occlusion (microvascular stasis) in SS-mice; infusion of zymosan-activated, but not heat-inactivated serum, induced substantial vaso-occlusion in the skin venules of SS-mice. Infusion of recombinant C5a induced stasis in SS, but not AA-mice that was blocked by anti-C5a receptor (C5aR) IgG. C5a-mediated stasis was accompanied by inflammatory responses in SS-mice including NF-κB activation and increased expression of TLR4 and adhesion molecules VCAM-1, ICAM-1, and E-selectin in the liver. Anti-C5aR IgG blocked these inflammatory responses. Also, C5a rapidly up-regulated Weibel-Palade body P-selectin and von Willebrand factor on the surface of human umbilical vein endothelial cells in vitro and on vascular endothelium in vivo. In SS-mice, a blocking antibody to P-selectin inhibited C5a-induced stasis. Similarly, an antibody to C5 that blocks murine C5 cleavage or an antibody that blocks C5aR inhibited H/R-induced stasis in SS-mice. These results suggest that inhibition of C5a may be beneficial in SCD.
Collapse
Affiliation(s)
- Gregory M. Vercellotti
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | | | - Terry R. Schaid
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Julia Nguyen
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Chunsheng Chen
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Marna E. Ericson
- Department of Dermatology; University of Minnesota; Minneapolis Minnesota
| | - Fuad Abdulla
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Trevor Killeen
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Margaret A. Lindorfer
- Department of Biochemistry and Molecular Genetics; University of Virginia School of Medicine; Charlottesville Virginia
| | - Ronald P. Taylor
- Department of Biochemistry and Molecular Genetics; University of Virginia School of Medicine; Charlottesville Virginia
| | - John D. Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| |
Collapse
|
28
|
Telen MJ, Malik P, Vercellotti GM. Therapeutic strategies for sickle cell disease: towards a multi-agent approach. Nat Rev Drug Discov 2019; 18:139-158. [PMID: 30514970 PMCID: PMC6645400 DOI: 10.1038/s41573-018-0003-2] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
For over 100 years, clinicians and scientists have been unravelling the consequences of the A to T substitution in the β-globin gene that produces haemoglobin S, which leads to the systemic manifestations of sickle cell disease (SCD), including vaso-occlusion, anaemia, haemolysis, organ injury and pain. However, despite growing understanding of the mechanisms of haemoglobin S polymerization and its effects on red blood cells, only two therapies for SCD - hydroxyurea and L-glutamine - are approved by the US Food and Drug Administration. Moreover, these treatment options do not fully address the manifestations of SCD, which arise from a complex network of interdependent pathophysiological processes. In this article, we review efforts to develop new drugs targeting these processes, including agents that reactivate fetal haemoglobin, anti-sickling agents, anti-adhesion agents, modulators of ischaemia-reperfusion and oxidative stress, agents that counteract free haemoglobin and haem, anti-inflammatory agents, anti-thrombotic agents and anti-platelet agents. We also discuss gene therapy, which holds promise of a cure, although its widespread application is currently limited by technical challenges and the expense of treatment. We thus propose that developing systems-oriented multi-agent strategies on the basis of SCD pathophysiology is needed to improve the quality of life and survival of people with SCD.
Collapse
Affiliation(s)
- Marilyn J Telen
- Division of Hematology, Department of Medicine and Duke Comprehensive Sickle Cell Center, Duke University, Durham, NC, USA.
| | - Punam Malik
- Division of Experimental Hematology and Cancer Biology and the Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Gregory M Vercellotti
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
29
|
Meng F, Kaul D, Thangaswamy S, Bhutoria S, Gerfen G, Branch C, Intaglietta M, Acharya SA. Semisynthetic supra plasma expanders: a new class of therapeutics to improve microcircualtion in sickle cell anaemia. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:73-82. [DOI: 10.1080/21691401.2018.1543194] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Fantao Meng
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MA, USA
| | - Dhananjaya Kaul
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Savita Bhutoria
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Gary Gerfen
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Craig Branch
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Radiology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Marcos Intaglietta
- Department of Bioengineering, University of California, San Diego, CA, USA
| | - Seetharama A. Acharya
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
30
|
New Therapeutic Options for the Treatment of Sickle Cell Disease. Mediterr J Hematol Infect Dis 2019; 11:e2019002. [PMID: 30671208 PMCID: PMC6328043 DOI: 10.4084/mjhid.2019.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/11/2018] [Indexed: 02/08/2023] Open
Abstract
Sickle cell disease (SCD; ORPHA232; OMIM # 603903) is a chronic and invalidating disorder distributed worldwide, with high morbidity and mortality. Given the disease complexity and the multiplicity of pathophysiological targets, development of new therapeutic options is critical, despite the positive effects of hydroxyurea (HU), for many years the only approved drug for SCD. New therapeutic strategies might be divided into (1) pathophysiology-related novel therapies and (2) innovations in curative therapeutic options such as hematopoietic stem cell transplantation and gene therapy. The pathophysiology related novel therapies are: a) Agents which reduce sickling or prevent sickle red cell dehydration; b) Agents targeting SCD vasculopathy and sickle cell-endothelial adhesive events; c) Anti-oxidant agents. This review highlights new therapeutic strategies in SCD and discusses future developments, research implications, and possible innovative clinical trials.
Collapse
|
31
|
Sundd P, Gladwin MT, Novelli EM. Pathophysiology of Sickle Cell Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2018; 14:263-292. [PMID: 30332562 DOI: 10.1146/annurev-pathmechdis-012418-012838] [Citation(s) in RCA: 339] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Since the discovery of sickle cell disease (SCD) in 1910, enormous strides have been made in the elucidation of the pathogenesis of its protean complications, which has inspired recent advances in targeted molecular therapies. In SCD, a single amino acid substitution in the β-globin chain leads to polymerization of mutant hemoglobin S, impairing erythrocyte rheology and survival. Clinically, erythrocyte abnormalities in SCD manifest in hemolytic anemia and cycles of microvascular vaso-occlusion leading to end-organ ischemia-reperfusion injury and infarction. Vaso-occlusive events and intravascular hemolysis promote inflammation and redox instability that lead to progressive small- and large-vessel vasculopathy. Based on current evidence, the pathobiology of SCD is considered to be a vicious cycle of four major processes, all the subject of active study and novel therapeutic targeting: ( a) hemoglobin S polymerization, ( b) impaired biorheology and increased adhesion-mediated vaso-occlusion, ( c) hemolysis-mediated endothelial dysfunction, and ( d) concerted activation of sterile inflammation (Toll-like receptor 4- and inflammasome-dependent innate immune pathways). These molecular, cellular, and biophysical processes synergize to promote acute and chronic pain and end-organ injury and failure in SCD. This review provides an exhaustive overview of the current understanding of the molecular pathophysiology of SCD, how this pathophysiology contributes to complications of the central nervous and cardiopulmonary systems, and how this knowledge is being harnessed to develop current and potential therapies.
Collapse
Affiliation(s)
- Prithu Sundd
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA; .,Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Sickle Cell Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | - Mark T Gladwin
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA; .,Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Sickle Cell Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | - Enrico M Novelli
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Sickle Cell Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| |
Collapse
|
32
|
Oral carbon monoxide therapy in murine sickle cell disease: Beneficial effects on vaso-occlusion, inflammation and anemia. PLoS One 2018; 13:e0205194. [PMID: 30308028 PMCID: PMC6181332 DOI: 10.1371/journal.pone.0205194] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/20/2018] [Indexed: 01/06/2023] Open
Abstract
Carbon monoxide (CO) at low, non-toxic concentrations has been previously demonstrated to exert anti-inflammatory protection in murine models of sickle cell disease (SCD). However CO delivery by inhalation, CO-hemoglobin infusion or CO-releasing molecules presents problems for daily CO administration. Oral administration of a CO-saturated liquid avoids many of these issues and potentially provides a platform for self-administration to SCD patients. To test if orally-delivered CO could modulate SCD vaso-occlusion and inflammation, a liquid CO formulation (HBI-002) was administered by gavage (10 ml/kg) once-daily to NY1DD and Townes-SS transgenic mouse models of SCD. Baseline CO-hemoglobin (CO-Hb) levels were 1.6% and 1.8% in NY1DD and Townes-SS sickle mice and 0.6% in Townes-AS control mice. CO-Hb levels reached 5.4%, 4.7% and 3.0% within 5 minutes in NY1DD, SS and AS mice respectively after gavage with HBI-002. After ten treatments, each once-daily, hemoglobin levels rose from 5.3g/dL in vehicle-treated Townes-SS mice to 6.3g/dL in HBI-002-treated. Similarly, red blood cell (RBC) counts rose from 2.36 x 106/μL in vehicle-treated SS mice to 2.89 x 106/μL in HBI-002-treated mice. In concordance with these findings, hematocrits rose from 26.3% in vehicle-treated mice to 30.0% in HBI-002-treated mice. Reticulocyte counts were not significantly different between vehicle and HBI-002-treated SS mice implying less hemolysis and not an increase in RBC production. White blood cell counts decreased from 29.1 x 103/μL in vehicle-treated versus 20.3 x 103/μL in HBI-002-treated SS mice. Townes-SS mice treated with HBI-002 had markedly increased Nrf2 and HO-1 expression and decreased NF-κB activation compared to vehicle-treated mice. These anti-inflammatory effects were examined for the ability of HBI-002 (administered orally once-daily for up to 5 days) to inhibit vaso-occlusion induced by hypoxia-reoxygenation. In NY1DD and Townes-SS sickle mice, HBI-002 decreased microvascular stasis in a duration-dependent manner. Collectively, these findings support HBI-002 as a useful anti-inflammatory agent to treat SCD and warrants further development as a therapeutic.
Collapse
|
33
|
Ben Hamda C, Sangeda R, Mwita L, Meintjes A, Nkya S, Panji S, Mulder N, Guizani-Tabbane L, Benkahla A, Makani J, Ghedira K. A common molecular signature of patients with sickle cell disease revealed by microarray meta-analysis and a genome-wide association study. PLoS One 2018; 13:e0199461. [PMID: 29979707 PMCID: PMC6034806 DOI: 10.1371/journal.pone.0199461] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 06/07/2018] [Indexed: 12/16/2022] Open
Abstract
A chronic inflammatory state to a large extent explains sickle cell disease (SCD) pathophysiology. Nonetheless, the principal dysregulated factors affecting this major pathway and their mechanisms of action still have to be fully identified and elucidated. Integrating gene expression and genome-wide association study (GWAS) data analysis represents a novel approach to refining the identification of key mediators and functions in complex diseases. Here, we performed gene expression meta-analysis of five independent publicly available microarray datasets related to homozygous SS patients with SCD to identify a consensus SCD transcriptomic profile. The meta-analysis conducted using the MetaDE R package based on combining p values (maxP approach) identified 335 differentially expressed genes (DEGs; 224 upregulated and 111 downregulated). Functional gene set enrichment revealed the importance of several metabolic pathways, of innate immune responses, erythrocyte development, and hemostasis pathways. Advanced analyses of GWAS data generated within the framework of this study by means of the atSNP R package and SIFT tool identified 60 regulatory single-nucleotide polymorphisms (rSNPs) occurring in the promoter of 20 DEGs and a deleterious SNP, affecting CAMKK2 protein function. This novel database of candidate genes, transcription factors, and rSNPs associated with SCD provides new markers that may help to identify new therapeutic targets.
Collapse
Affiliation(s)
- Cherif Ben Hamda
- Laboratory of Bioinformatics, Biomathematics and Biostatistics, Institute Pasteur of Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
- Faculty of Science of Bizerte, Jarzouna, University of Carthage, Tunisia
- * E-mail: (KG); (CBH)
| | - Raphael Sangeda
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Liberata Mwita
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | | | - Siana Nkya
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Sumir Panji
- University of Cape Town, Cape Town, South Africa
| | | | - Lamia Guizani-Tabbane
- University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institute Pasteur of Tunis, Tunis, Tunisia
| | - Alia Benkahla
- Laboratory of Bioinformatics, Biomathematics and Biostatistics, Institute Pasteur of Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
| | - Julie Makani
- Faculty of Science of Bizerte, Jarzouna, University of Carthage, Tunisia
| | - Kais Ghedira
- Laboratory of Bioinformatics, Biomathematics and Biostatistics, Institute Pasteur of Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
- * E-mail: (KG); (CBH)
| | | |
Collapse
|
34
|
Comparison of the oxidative reactivity of recombinant fetal and adult human hemoglobin: implications for the design of hemoglobin-based oxygen carriers. Biosci Rep 2018; 38:BSR20180370. [PMID: 29802155 PMCID: PMC6028758 DOI: 10.1042/bsr20180370] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 12/21/2022] Open
Abstract
Hemoglobin (Hb)-based oxygen carriers (HBOCs) have been engineered to replace or augment the oxygen carrying capacity of erythrocytes. However, clinical results have generally been disappointing, in part due to the intrinsic oxidative toxicity of Hb. The most common HBOC starting material is adult human or bovine Hb. However, it has been suggested that fetal Hb may offer advantages due to decreased oxidative reactivity. Large-scale manufacturing of HBOC will likely and ultimately require recombinant sources of human proteins. We, therefore, directly compared the functional properties and oxidative reactivity of recombinant fetal (rHbF) and recombinant adult (rHbA) Hb. rHbA and rHbF produced similar yields of purified functional protein. No differences were seen in the two proteins in: autoxidation rate; the rate of hydrogen peroxide reaction; NO scavenging dioxygenase activity; and the NO producing nitrite reductase activity. The rHbF protein was: less damaged by low levels of hydrogen peroxide; less damaging when added to human umbilical vein endothelial cells (HUVEC) in the ferric form; and had a slower rate of intrinsic heme loss. The rHbA protein was: more readily reducible by plasma antioxidants such as ascorbate in both the reactive ferryl and ferric states; less readily damaged by lipid peroxides; and less damaging to phosphatidylcholine liposomes. In conclusion in terms of oxidative reactivity, there are advantages and disadvantages to the use of rHbA or rHbF as the basis for an effective HBOC.
Collapse
|
35
|
Abstract
The primary β-globin gene mutation that causes sickle cell disease (SCD) has significant pathophysiological consequences that result in hemolytic events and the induction of the inflammatory processes that ultimately lead to vaso-occlusion. In addition to their role in the initiation of the acute painful vaso-occlusive episodes that are characteristic of SCD, inflammatory processes are also key components of many of the complications of the disease including autosplenectomy, acute chest syndrome, pulmonary hypertension, leg ulcers, nephropathy and stroke. We, herein, discuss the events that trigger inflammation in the disease, as well as the mechanisms, inflammatory molecules and cells that propagate these inflammatory processes. Given the central role that inflammation plays in SCD pathophysiology, many of the therapeutic approaches currently under pre-clinical and clinical development for the treatment of SCD endeavor to counter aspects or specific molecules of these inflammatory processes and it is possible that, in the future, we will see anti-inflammatory drugs being used either together with, or in place of, hydroxyurea in those SCD patients for whom hematopoietic stem cell transplants and evolving gene therapies are not a viable option.
Collapse
Affiliation(s)
- Nicola Conran
- Hematology Center, University of Campinas - UNICAMP, Cidade Universitária, Campinas-SP, Brazil
| | - John D Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
36
|
Danger signals from mitochondrial DAMPS in trauma and post-injury sepsis. Eur J Trauma Emerg Surg 2018; 44:317-324. [PMID: 29797026 DOI: 10.1007/s00068-018-0963-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 05/19/2018] [Indexed: 12/13/2022]
Abstract
In all multicellular organisms, immediate host responses to both sterile and infective threat are initiated by very primitive systems now grouped together under the general term 'danger responses'. Danger signals are generated when primitive 'pattern recognition receptors' (PRR) encounter activating 'alarmins'. These molecular species may be of pathogenic infective origin (pathogen-associated molecular patterns) or of sterile endogenous origin (danger-associated molecular patterns). There are many sterile and infective alarmins and there is considerable overlap in their ability to activate PRR, but in all cases the end result is inflammation. It is the overlap between sterile and infective signals acting via a relatively limited number of PRR that generally underlies the great clinical similarity we see between sterile and infective systemic inflammatory responses. Mitochondria (MT) are evolutionarily derived from bacteria, and thus they sit at the crossroads between sterile and infective danger signal pathways. Many of the molecular species in mitochondria are alarmins, and so the release of MT from injured cells results in a wide variety of inflammatory events. This paper discusses the known participation of MT in inflammation and reviews what is known about how the major.
Collapse
|
37
|
Abstract
Introduction Sickle cell disease (SCD) is an orphan disease in the United States, but is highly prevalent worldwide. Only two drugs, hydroxyurea and L-glutamine, are approved for this disease. With an improved understanding of the pathophysiology of SCD as well as the success of several recently approved drugs for other orphan diseases, there is an increased interest in the development of drugs for SCD. Areas covered This review summarizes published studies of drug therapies and ongoing trials of novel agents. Expert opinion The development of drugs with different mechanisms of action offers opportunities for combination and individualized therapy in SCD. In addition to acute pain crisis, the evaluation of other SCD-related complications, exercise capacity, patient reported outcomes and validated surrogate endpoints are necessary to advance drug development. It is important to involve sites in sub-Saharan Africa and India, which have the highest burden of SCD, in trials of novel therapies.
Collapse
Affiliation(s)
- Kenneth I Ataga
- Division of Hematology/Oncology, University of North Carolina, Chapel Hill, NC
| | - Payal C Desai
- Division of Hematology/Oncology, University of North Carolina, Chapel Hill, NC.,#Division of Hematology, The Ohio State University, Columbus, OH
| |
Collapse
|
38
|
HO-1 hi patrolling monocytes protect against vaso-occlusion in sickle cell disease. Blood 2018; 131:1600-1610. [PMID: 29437594 DOI: 10.1182/blood-2017-12-819870] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 01/29/2018] [Indexed: 12/18/2022] Open
Abstract
Patients with sickle cell disease (SCD) suffer from intravascular hemolysis associated with vascular injury and dysfunction in mouse models, and painful vaso-occlusive crisis (VOC) involving increased attachment of sickle erythrocytes and activated leukocytes to damaged vascular endothelium. Patrolling monocytes, which normally scavenge damaged cells and debris from the vasculature, express higher levels of anti-inflammatory heme oxygenase 1 (HO-1), a heme degrading enzyme. Here, we show that HO-1-expressing patrolling monocytes protect SCD vasculature from ongoing hemolytic insult and vaso-occlusion. We found that a mean 37% of patrolling monocytes from SCD patients express very high levels of HO-1 (HO-1hi) vs 6% in healthy controls and demonstrated that HO-1hi expression was dependent on uptake of heme-exposed endothelium. SCD patients with a recent VOC episode had lower numbers of HO-1hi patrolling monocytes. Heme-mediated vaso-occlusion by mouse SCD red blood cells was exacerbated in mice lacking patrolling monocytes, and reversed following transfer of patrolling monocytes. Altogether, these data indicate that SCD patrolling monocytes remove hemolysis-damaged endothelial cells, resulting in HO-1 upregulation and dampening of VOC, and that perturbation in patrolling monocyte numbers resulting in lower numbers of HO-1hi patrolling monocyte may predispose SCD patients to VOC. These data suggest that HO-1hi patrolling monocytes are key players in VOC pathophysiology and have potential as therapeutic targets for VOC.
Collapse
|
39
|
Gomperts E, Belcher JD, Otterbein LE, Coates TD, Wood J, Skolnick BE, Levy H, Vercellotti GM. The role of carbon monoxide and heme oxygenase in the prevention of sickle cell disease vaso-occlusive crises. Am J Hematol 2017; 92:569-582. [PMID: 28378932 PMCID: PMC5723421 DOI: 10.1002/ajh.24750] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/28/2017] [Accepted: 03/29/2017] [Indexed: 12/15/2022]
Abstract
Sickle Cell Disease (SCD) is a painful, lifelong hemoglobinopathy inherited as a missense point mutation in the hemoglobin (Hb) beta-globin gene. This disease has significant impact on quality of life and mortality, thus a substantial medical need exists to reduce the vaso-occlusive crises which underlie the pathophysiology of the disease. The concept that a gaseous molecule may exert biological function has been well known for over one hundred years. Carbon monoxide (CO), although studied in SCD for over 50 years, has recently emerged as a powerful cytoprotective biological response modifier capable of regulating a host of physiologic and therapeutic processes that, at low concentrations, exerts key physiological functions in various models of tissue inflammation and injury. CO is physiologically generated by the metabolism of heme by the heme oxygenase enzymes and is measurable in blood. A substantial amount of preclinical and clinical data with CO have been generated, which provide compelling support for CO as a potential therapeutic in a number of pathological conditions. Data underlying the therapeutic mechanisms of CO, including in SCD, have been generated by a plethora of in vitro and preclinical studies including multiple SCD mouse models. These data show CO to have key signaling impacts on a host of metallo-enzymes as well as key modulating genes that in sum, result in significant anti-inflammatory, anti-oxidant and anti-apoptotic effects as well as vasodilation and anti-adhesion of cells to the endothelium resulting in preservation of vascular flow. CO may also have a role as an anti-polymerization HbS agent. In addition, considerable scientific data in the non-SCD literature provide evidence for a beneficial impact of CO on cerebrovascular complications, suggesting that in SCD, CO could potentially limit these highly problematic neurologic outcomes. Research is needed and hopefully forthcoming, to carefully elucidate the safety and benefits of this potential therapy across the age spectrum of patients impacted by the host of pathophysiological complications of this devastating disease.
Collapse
Affiliation(s)
- Edward Gomperts
- Hillhurst Biopharmaceuticals, Inc, 2029 Verdugo Blvd., #125, Montrose, CA, 91020, USA
| | - John D Belcher
- University of Minnesota, 420 Delaware Street SE, MMC 480, Minneapolis, MN, 55455, USA
| | - Leo E Otterbein
- Harvard Medical School; Beth Israel Deaconess Medical Center, 3 Blackfan Circle Center for Life Sciences, #630, Boston, MA, 02115, USA
| | - Thomas D Coates
- Children's Hospital Los Angeles; University of Southern California, 4650 Sunset Boulevard MS #54 Los Angeles, CA, 90027, USA
| | - John Wood
- Children's Hospital Los Angeles; University of Southern California, 4650 Sunset Boulevard MS #54 Los Angeles, CA, 90027, USA
| | - Brett E Skolnick
- Hillhurst Biopharmaceuticals, Inc, 2029 Verdugo Blvd., #125, Montrose, CA, 91020, USA
| | - Howard Levy
- Hillhurst Biopharmaceuticals, Inc, 2029 Verdugo Blvd., #125, Montrose, CA, 91020, USA
| | - Gregory M Vercellotti
- University of Minnesota, 420 Delaware Street SE, MMC 480, Minneapolis, MN, 55455, USA
| |
Collapse
|
40
|
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
|
41
|
Belcher JD, Chen C, Nguyen J, Zhang P, Abdulla F, Nguyen P, Killeen T, Xu P, O'Sullivan G, Nath KA, Vercellotti GM. Control of Oxidative Stress and Inflammation in Sickle Cell Disease with the Nrf2 Activator Dimethyl Fumarate. Antioxid Redox Signal 2017; 26:748-762. [PMID: 26914345 PMCID: PMC5421647 DOI: 10.1089/ars.2015.6571] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIMS Heme derived from hemolysis is pro-oxidative and proinflammatory and promotes vaso-occlusion in murine models of sickle cell disease (SCD), suggesting that enhanced detoxification of heme may be beneficial. Nuclear factor erythroid-2-related factor-2 (Nrf2) transcription pathway is the principal cellular defense system responding to pro-oxidative and proinflammatory stress. Dimethyl fumarate (DMF), a drug approved for treatment of multiple sclerosis, provides neuroprotection by activating Nrf2-responsive genes. We hypothesized that induction of Nrf2 with DMF would be beneficial in murine SCD models. RESULTS DMF (30 mg/kg/day) or vehicle (0.08% methyl cellulose) was administered for 3-7 days to NY1DD and HbSS-Townes SCD mice. Vaso-occlusion, a hallmark of SCD, measured in sickle mice with dorsal skinfold chambers, was inhibited by DMF. The inhibitory effect of DMF was abrogated by the heme oxygenase-1 (HO-1) inhibitor tin protoporphyrin. DMF increased nuclear Nrf2 and cellular mRNA of Nrf2-responsive genes in livers and kidneys. DMF increased heme defenses, including HO-1, haptoglobin, hemopexin, and ferritin heavy chain, although plasma hemoglobin and heme levels were unchanged. DMF decreased markers of inflammation, including nuclear factor-kappa B phospho-p65, adhesion molecules, and toll-like receptor 4. DMF administered for 24 weeks to HbSS-Townes mice decreased hepatic necrosis, inflammatory cytokines, and irregularly shaped erythrocytes and increased hemoglobin F, but did not alter hematocrits, reticulocyte counts, lactate dehydrogenase, plasma heme, or spleen weights, indicating that the beneficial effects of DMF were not attributable to decreased hemolysis. INNOVATION These studies identify Nrf2 activation as a new therapeutic target for the treatment of SCD. CONCLUSION DMF activates Nrf2, enhances antioxidant defenses, and inhibits inflammation and vaso-occlusion in SCD mice. Antioxid. Redox Signal. 26, 748-762.
Collapse
Affiliation(s)
- John D Belcher
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Chunsheng Chen
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Julia Nguyen
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Ping Zhang
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Fuad Abdulla
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Phong Nguyen
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Trevor Killeen
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Pauline Xu
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Gerry O'Sullivan
- 2 Veterinary Population Medicine, University of Minnesota , St. Paul, Minnesota
| | - Karl A Nath
- 3 Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic/Foundation , Rochester, Minnesota
| | - Gregory M Vercellotti
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| |
Collapse
|
42
|
Comparison of the Pharmacokinetic Properties of Hemoglobin-Based Oxygen Carriers. J Funct Biomater 2017; 8:jfb8010011. [PMID: 28335469 PMCID: PMC5371884 DOI: 10.3390/jfb8010011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/15/2017] [Accepted: 03/15/2017] [Indexed: 12/23/2022] Open
Abstract
Hemoglobin (Hb) is an ideal material for use in the development of an oxygen carrier in view of its innate biological properties. However, the vascular retention of free Hb is too short to permit a full therapeutic effect because Hb is rapidly cleared from the kidney via glomerular filtration or from the liver via the haptogloblin-CD 163 pathway when free Hb is administered in the blood circulation. Attempts have been made to develop alternate acellular and cellular types of Hb based oxygen carriers (HBOCs), in which Hb is processed via various routes in order to regulate its pharmacokinetic properties. These HBOCs have been demonstrated to have superior pharmacokinetic properties including a longer half-life than the Hb molecule in preclinical and clinical trials. The present review summarizes and compares the pharmacokinetic properties of acellular and cellular type HBOCs that have been developed through different approaches, such as polymerization, PEGylation, cross-linking, and encapsulation.
Collapse
|
43
|
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
|
44
|
Oxygen Carriers: Are They Enough for Cellular Support? SPRINGER SERIES IN TRANSLATIONAL STROKE RESEARCH 2017. [DOI: 10.1007/978-3-319-45345-3_26] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
45
|
Wang Q, Hu L, Hu Y, Gong G, Tan H, Deng L, Sun X, Yi X, Sun Y, Wu W, Li T. Carbon Monoxide-Saturated Hemoglobin-Based Oxygen Carriers Attenuate High-Altitude-Induced Cardiac Injury by Amelioration of the Inflammation Response and Mitochondrial Oxidative Damage. Cardiology 2016; 136:180-191. [DOI: 10.1159/000448652] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/21/2016] [Indexed: 11/19/2022]
Abstract
Objective: To investigate the therapeutic effect of carbon monoxide (CO) on high-altitude hypoxia-induced cardiac damage. Methods: Forty male C57BL/6 mice were randomly divided into 4 groups. The mice were exposed to normoxia or simulated 5,500-meter high-altitude hypoxia in a hypobaric chamber for 7 days. During the first 3 days, the mice were pretreated with CO-saturated hemoglobin (Hb)-based oxygen carrier (CO-HBOC), oxygen-saturated hemoglobin-based oxygen carrier (O2-HBOC) at a dose of 0.3 g Hb/kg/day or an equivalent volume of saline. The in vivo left ventricle function, cardiac enzyme release, histopathological changes, apoptosis and inflammation were also measured. Results: High-altitude hypoxia induced significant cardiac damage, as demonstrated by impaired cardiac function and increased proapoptotic, proinflammatory and pro-oxidant markers. Pretreatment with CO-HBOC significantly improved cardiac performance, reduced cardiac enzyme release and limited myocardial apoptosis. The increased inflammatory response was also suppressed. In addition to the preserved mitochondrial structure, hypobaric hypoxia-induced mitochondrial oxidative damage was remarkably attenuated. Moreover, these antiapoptotic and antioxidative effects were accompanied by an upregulated phosphorylation of Akt, ERK and STAT3. Conclusion: This study demonstrated that CO-HBOC provides a promising protective effect on high-altitude hypoxia-induced myocardial injury, which is mediated by the inhibition of inflammation and mitochondrial oxidative damage.
Collapse
|
46
|
Shi PA, Choi E, Chintagari NR, Nguyen J, Guo X, Yazdanbakhsh K, Mohandas N, Alayash AI, Manci EA, Belcher JD, Vercellotti GM. Sustained treatment of sickle cell mice with haptoglobin increases HO-1 and H-ferritin expression and decreases iron deposition in the kidney without improvement in kidney function. Br J Haematol 2016; 175:714-723. [PMID: 27507623 DOI: 10.1111/bjh.14280] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/06/2016] [Indexed: 12/21/2022]
Abstract
There is growing evidence that extracellular haemoglobin and haem mediate inflammatory and oxidative damage in sickle cell disease. Haptoglobin (Hp), the scavenger for free haemoglobin, is depleted in most patients with sickle cell disease due to chronic haemolysis. Although single infusions of Hp can ameliorate vaso-occlusion in mouse models of sickle cell disease, prior studies have not examined the therapeutic benefits of more chronic Hp dosing on sickle cell disease manifestations. In the present study, we explored the effect of Hp treatment over a 3-month period in sickle mice at two dosing regimens: the first at a moderate dose of 200 mg/kg thrice weekly and the second at a higher dose of 400 mg/kg thrice weekly. We found that only the higher dosing regimen resulted in increased haem-oxygenase-1 and heavy chain ferritin (H-ferritin) expression and decreased iron deposition in the kidney. Despite the decreased kidney iron deposition following Hp treatment, there was no significant improvement in kidney function. However, there was a nearly significant trend towards decreased liver infarction.
Collapse
Affiliation(s)
- Patricia A Shi
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Erika Choi
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | | | - Julia Nguyen
- Department of Hematology, Oncology, and Transplantation, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Xinhua Guo
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Karina Yazdanbakhsh
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Narla Mohandas
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Abdu I Alayash
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Elizabeth A Manci
- Department of Pathology, University of South Alabama School of Medicine, Birmingham, AL, USA
| | - John D Belcher
- Department of Hematology, Oncology, and Transplantation, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Gregory M Vercellotti
- Department of Hematology, Oncology, and Transplantation, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
47
|
Abstract
Carbon monoxide (CO) has long been considered an environmental pollutant and a poison. Exogenous exposure to amounts of CO beyond the physiologic level of the body can result in a protective or adaptive response. However, as a gasotransmitter, endogenous CO is important for multiple physiologic functions. To date, at least seven distinct methods of delivering CO have been utilized in animal and clinical studies. In this mini-review, we summarize the exogenous CO delivery methods and compare their advantages and disadvantages.
Collapse
Affiliation(s)
- Hui-Jun Hu
- Department of Navy Aeromedicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China; Department of Hyperbaric Oxygen, Navy General Hospital, Beijing, China
| | - Qiang Sun
- Department of Hyperbaric Oxygen, Navy General Hospital, Beijing, China
| | - Zhou-Heng Ye
- Department of Navy Aeromedicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Xue-Jun Sun
- Department of Navy Aeromedicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| |
Collapse
|
48
|
Arnaud F, Higgins A, McCarron R, Moon-Massat PF. Determination of methemoglobin and hemoglobin levels in small volume samples. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:58-62. [DOI: 10.3109/21691401.2016.1138490] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Francoise Arnaud
- NeuroTrauma Department, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, MD, USA
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Adam Higgins
- NeuroTrauma Department, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, MD, USA
| | - Richard McCarron
- NeuroTrauma Department, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, MD, USA
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Paula F. Moon-Massat
- NeuroTrauma Department, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, MD, USA
| |
Collapse
|
49
|
Beyond hydroxyurea: new and old drugs in the pipeline for sickle cell disease. Blood 2016; 127:810-9. [PMID: 26758919 DOI: 10.1182/blood-2015-09-618553] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 10/21/2015] [Indexed: 01/09/2023] Open
Abstract
Despite Food and Drug Administration (FDA) approval of hydroxyurea to reduce the frequency of vaso-occlusive episodes, sickle cell disease (SCD) has continued to be treated primarily with analgesics for pain relief. However, elucidation of the multiple pathophysiologic mechanisms leading to vaso-occlusion and tissue injury in SCD has now resulted in a burgeoning effort to identify new treatment modalities to prevent or ameliorate the consequences of the disease. Development of new drugs as well as investigation of drugs previously used in other settings have targeted cell adhesion, inflammatory pathways, upregulation of hemoglobin F, hemoglobin polymerization and sickling, coagulation, and platelet activation. Although these efforts have not yet yielded drugs ready for FDA approval, several early studies have been extremely encouraging. Moreover, the marked increase in clinical pharmaceutical research addressing SCD and the new and old drugs in the pipeline make it reasonable to expect that we will soon have new treatments for SCD.
Collapse
|
50
|
Keipert PE. Clinical Evaluation of MP4CO: A Phase 1b Escalating-Dose, Safety and Tolerability Study in Stable Adult Patients with Sickle Cell Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 923:23-29. [PMID: 27526120 DOI: 10.1007/978-3-319-38810-6_3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
MP4CO, developed by Sangart Inc. (San Diego, CA), is a pegylated human hemoglobin-based carbon monoxide (CO) delivery agent and oxygen therapeutic that has shown potential to prevent and reverse red cell sickling. A double blind, comparator controlled, dose-escalation, Phase 1b study was conducted to assess the safety of MP4CO. Adult sickle cell patients with HbSS or S/β(0) Thal genotype who were not experiencing a painful crisis were randomized to receive either MP4CO or normal saline (NS) in a sequential series of six escalating dose cohorts (A-F). In each cohort, three patients received MP4CO (Treatment group) and one patient received NS (Controls). Single IV doses ranged from 15 mg/kg/dose (0.35 mL/kg infusion) to 172 mg/kg/dose (4 mL/kg infusion). Two cohorts received fractionated doses of 172 or 344 mg/kg (4-8 mL/kg, given as two IV infusions, 24 h apart). Overall, 16/24 patients (66.7 %) reported mild to moderate adverse events (AEs); with 13/18 (72 %) in MP4CO group vs. 3/6 (50 %) in NS Controls. No serious adverse events (SAEs) were experienced and no deaths occurred. Most common AEs (reported by >2 patients) included headaches (mild and transient), fatigue and rash at the application site of the Holter electrodes. No treatment-emergent abnormalities in clinical lab values were noted. Vital signs, ECG readings, and pulmonary pressures remained within normal limits. The maximum increase in blood CO-Hb level was ~2 %, which returned to pre-dosing levels within 8 h after dosing. The mean increase in free plasma Hb (an index of MP4CO dose) ranged from 0.20 to 0.35 g/dL in the two highest dose cohorts, with no significant change in total whole blood hemoglobin level. There was no symptomatic or clinical evidence of renal dysfunction in either group based on serum creatinine and urinary albumin results. Two patients had elevated renal biomarkers (β2M and NAG) at Hour 72, which normalized at follow-up visits. Both patients had documented intercurrent illnesses during the study. Further testing of stored urine samples were within normal limits, which suggested the changes were reflective of a generalized inflammatory state rather than direct tubular injury.
Collapse
Affiliation(s)
- Peter E Keipert
- KEIPERT Corp. Life Sciences Consulting, San Diego, CA, 92130, USA.
| | | |
Collapse
|