1
|
Stukan I, Żuk A, Pukacka K, Mierzejewska J, Pawłowski J, Kowalski B, Dąbkowska M. Wolf in Sheep's Clothing: Taming Cancer's Resistance with Human Serum Albumin? Int J Nanomedicine 2025; 20:3493-3525. [PMID: 40125439 PMCID: PMC11930253 DOI: 10.2147/ijn.s500997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
Human serum albumin (HSA) has emerged as a promising carrier for nanodrug delivery, offering unique structural properties that can be engineered to overcome key challenges in cancer treatment, especially resistance to chemotherapy. This review focuses on the cellular uptake of albumin-based nanoparticles and the modifications that enhance their ability to bypass resistance mechanisms, particularly multidrug resistance type 1 (MDR1), by improving targeting to cancer cells. In our unique approach, we integrate the chemical properties of albumin, its interactions with cancer cells, and surface modifications of albumin-based delivery systems that enable to bypass resistance mechanisms, particularly those related to MDR1, and precisely target receptors on cancer cells to improve treatment efficacy. We discuss that while well-established albumin receptors such as gp60 and gp18/30 are crucial for cellular uptake and transcytosis, their biology remains underexplored, limiting their translational potential. Additionally, we explore the potential of emerging targets, such as cluster of differentiation 44 (CD44), cluster of differentiation (CD36) and transferrin receptor TfR1, as well as the advantages of using dimeric forms of albumin (dHSA) to further enhance delivery to resistant cancer cells. Drawing from clinical examples, including the success of albumin-bound paclitaxel (Abraxane) and new formulations like Pazenir and Fyarro (for Sirolimus), we identify gaps in current knowledge and propose strategies to optimize albumin-based systems. In conclusion, albumin-based nanoparticles, when tailored with appropriate modifications, have the potential to bypass multidrug resistance and improve the targeting of cancer cells. By enhancing albumin's ability to efficiently deliver therapeutic agents, these carriers represent a promising approach to addressing one of oncology's most persistent challenges, with substantial potential to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Iga Stukan
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Anna Żuk
- Independent Laboratory of Community Pharmacy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Kamila Pukacka
- Department of Pharmaceutical Technology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Julia Mierzejewska
- Independent Laboratory of Pharmacokinetic and Clinical Pharmacy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Jakub Pawłowski
- Independent Laboratory of Pharmacokinetic and Clinical Pharmacy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Bogusław Kowalski
- Independent Laboratory of Pharmacokinetic and Clinical Pharmacy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Maria Dąbkowska
- Independent Laboratory of Pharmacokinetic and Clinical Pharmacy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| |
Collapse
|
2
|
Mikhailova DM, Skverchinskaya E, Sudnitsyna J, Butov KR, Koltsova EM, Mindukshev IV, Gambaryan S. Hematin- and Hemin-Induced Spherization and Hemolysis of Human Erythrocytes Are Independent of Extracellular Calcium Concentration. Cells 2024; 13:554. [PMID: 38534398 PMCID: PMC10969559 DOI: 10.3390/cells13060554] [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: 02/12/2024] [Revised: 03/10/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
Pathologies such as malaria, hemorrhagic stroke, sickle cell disease, and thalassemia are characterized by the release of hemoglobin degradation products from damaged RBCs. Hematin (liganded with OH-) and hemin (liganded with Cl-)-are the oxidized forms of heme with toxic properties due to their hydrophobicity and the presence of redox-active Fe3. In the present study, using the original LaSca-TM laser particle analyzer, flow cytometry, and confocal microscopy, we showed that both hematin and hemin induce dose-dependent RBC spherization and hemolysis with ghost formation. Hematin and hemin at nanomolar concentrations increased [Ca2+]i in RBC; however, spherization and hemolysis occurred in the presence and absence of calcium, indicating that both processes are independent of [Ca2+]i. Both compounds triggered acute phosphatidylserine exposure on the membrane surface, reversible after 60 min of incubation. A comparison of hematin and hemin effects on RBCs revealed that hematin is a more reactive toxic metabolite than hemin towards human RBCs. The toxic effects of heme derivatives were reduced and even reversed in the presence of albumin, indicating the presence in RBCs of the own recovery system against the toxic effects of heme derivatives.
Collapse
Affiliation(s)
- Diana M. Mikhailova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Ave., 194223 Saint Petersburg, Russia; (D.M.M.); (E.S.); (J.S.); (I.V.M.)
- Department of Cytology and Histology, Saint Petersburg State University, 7/9 Universitetskaya Emb., 199034 Saint Petersburg, Russia
| | - Elisaveta Skverchinskaya
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Ave., 194223 Saint Petersburg, Russia; (D.M.M.); (E.S.); (J.S.); (I.V.M.)
| | - Julia Sudnitsyna
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Ave., 194223 Saint Petersburg, Russia; (D.M.M.); (E.S.); (J.S.); (I.V.M.)
| | - Kirill R. Butov
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia;
| | - Ekaterina M. Koltsova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia;
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya st., 109029 Moscow, Russia
| | - Igor V. Mindukshev
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Ave., 194223 Saint Petersburg, Russia; (D.M.M.); (E.S.); (J.S.); (I.V.M.)
| | - Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Ave., 194223 Saint Petersburg, Russia; (D.M.M.); (E.S.); (J.S.); (I.V.M.)
| |
Collapse
|
3
|
Voltarelli VA, Alves de Souza RW, Miyauchi K, Hauser CJ, Otterbein LE. Heme: The Lord of the Iron Ring. Antioxidants (Basel) 2023; 12:antiox12051074. [PMID: 37237940 DOI: 10.3390/antiox12051074] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Heme is an iron-protoporphyrin complex with an essential physiologic function for all cells, especially for those in which heme is a key prosthetic group of proteins such as hemoglobin, myoglobin, and cytochromes of the mitochondria. However, it is also known that heme can participate in pro-oxidant and pro-inflammatory responses, leading to cytotoxicity in various tissues and organs such as the kidney, brain, heart, liver, and in immune cells. Indeed, heme, released as a result of tissue damage, can stimulate local and remote inflammatory reactions. These can initiate innate immune responses that, if left uncontrolled, can compound primary injuries and promote organ failure. In contrast, a cadre of heme receptors are arrayed on the plasma membrane that is designed either for heme import into the cell, or for the purpose of activating specific signaling pathways. Thus, free heme can serve either as a deleterious molecule, or one that can traffic and initiate highly specific cellular responses that are teleologically important for survival. Herein, we review heme metabolism and signaling pathways, including heme synthesis, degradation, and scavenging. We will focus on trauma and inflammatory diseases, including traumatic brain injury, trauma-related sepsis, cancer, and cardiovascular diseases where current work suggests that heme may be most important.
Collapse
Affiliation(s)
- Vanessa Azevedo Voltarelli
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Rodrigo W Alves de Souza
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Kenji Miyauchi
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Carl J Hauser
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Leo Edmond Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
4
|
Wang Y, Zha W, Wang J, Dong S, Liu C, Jiang Y, Li X. Local delivery of artesunate dimer liposomes incorporated injectable hydrogel for H 2O 2 and pH-independent chemodynamic therapy. Int J Pharm 2023; 636:122822. [PMID: 36914018 DOI: 10.1016/j.ijpharm.2023.122822] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/20/2023] [Accepted: 03/08/2023] [Indexed: 03/13/2023]
Abstract
Chemodynamic therapy (CDT) has emerged as a powerful tumor treatment option by inducing the imbalance of redox homeostasis in cancer cells. Nevertheless, the therapeutic outcomes were greatly limited because of insufficient endogenous H2O2 and upregulated cellular antioxidant defense in the tumor microenvironment (TME). Herein, a liposome-incorporated in-situ alginate hydrogel locoregional treatment strategy was developed, which involves using hemin-loaded artesunate dimer liposomes (HAD-LP) as redox-triggered self-amplified C-center free radical nanogenerator to enhance CDT. First, HAD-LP based on artesunate dimer glycerophosphocholine (ART-GPC) was prepared by a thin film method. Their spherical structure was manifested by dynamic light scattering (DLS) and transmission electron microscope (TEM). The generation of C-center free radicals from HAD-LP was carefully evaluated by using methylene blue (MB) degradation method. The results suggested that the hemin was reduced to heme under the action of glutathione (GSH), which could catalyze the breakage of endoperoxide of ART-GPC derived dihydroartemisinin (DHA) to generate toxic C-centered free radicals in a H2O2 and pH-independent manner. Moreover, the change of intracellular GSH and free radical level was monitored through ultraviolet spectroscopy and confocal laser scanning microscope (CLSM). It was revealed that the hemin reduction induced GSH depletion and elevated free radical level, disrupting cellular redox homeostasis. After co-incubation with MDA-MB-231 or 4 T1 cells, HAD-LP was found to be highly cytotoxic. In order to prolong retention and improve antitumor efficacy, HAD-LP was mixed with alginate and injected intratumorally into 4 T1 tumor bearing mice. The injected HAD-LP and alginate mixture formed in-situ hydrogel and achieved best antitumor effect with the growth inhibition rate of 72.6%. Together, the hemin-loaded artesunate dimer liposome-incorporated alginate hydrogel possessed effective antitumor activity through redox-triggered C-center free radical generation induced apoptosis in a H2O2 and pH-independent manner, which might be a promising candidate in the application of chemodynamic anti-tumor therapy.
Collapse
Affiliation(s)
- Yang Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 214122, PR China
| | - Wenhui Zha
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 214122, PR China
| | - Ji Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 214122, PR China
| | - Shuo Dong
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 214122, PR China
| | - Chao Liu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 214122, PR China
| | - Yuhao Jiang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 214122, PR China
| | - Xinsong Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 214122, PR China.
| |
Collapse
|
5
|
Song D, Yeh CT, Wang J, Guo F. Perspectives on the mechanism of pyroptosis after intracerebral hemorrhage. Front Immunol 2022; 13:989503. [PMID: 36131917 PMCID: PMC9484305 DOI: 10.3389/fimmu.2022.989503] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/17/2022] [Indexed: 12/18/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a highly harmful neurological disorder with high rates of mortality, disability, and recurrence. However, effective therapies are not currently available. Secondary immune injury and cell death are the leading causes of brain injury and a poor prognosis. Pyroptosis is a recently discovered form of programmed cell death that differs from apoptosis and necrosis and is mediated by gasdermin proteins. Pyroptosis is caused by multiple pathways that eventually form pores in the cell membrane, facilitating the release of inflammatory substances and causing the cell to rupture and die. Pyroptosis occurs in neurons, glial cells, and endothelial cells after ICH. Furthermore, pyroptosis causes cell death and releases inflammatory factors such as interleukin (IL)-1β and IL-18, leading to a secondary immune-inflammatory response and further brain damage. The NOD-like receptor protein 3 (NLRP3)/caspase-1/gasdermin D (GSDMD) pathway plays the most critical role in pyroptosis after ICH. Pyroptosis can be inhibited by directly targeting NLRP3 or its upstream molecules, or directly interfering with caspase-1 expression and GSDMD formation, thus significantly improving the prognosis of ICH. The present review discusses key pathological pathways and regulatory mechanisms of pyroptosis after ICH and suggests possible intervention strategies to mitigate pyroptosis and brain dysfunction after ICH.
Collapse
Affiliation(s)
- Dengpan Song
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chi-Tai Yeh
- Department of Medical Research and Education, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- *Correspondence: Fuyou Guo, ; Jian Wang, ; Chi-Tai Yeh,
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Fuyou Guo, ; Jian Wang, ; Chi-Tai Yeh,
| | - Fuyou Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Fuyou Guo, ; Jian Wang, ; Chi-Tai Yeh,
| |
Collapse
|
6
|
Alsharabasy AM, Glynn S, Farràs P, Pandit A. Protein nitration induced by Hemin/NO: A complementary mechanism through the catalytic functions of hemin and NO-scavenging. Nitric Oxide 2022; 124:49-67. [PMID: 35513288 DOI: 10.1016/j.niox.2022.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/07/2022] [Accepted: 04/29/2022] [Indexed: 12/13/2022]
Abstract
Hemin and heme-peroxidases have been considered essential catalysts for the nitrite/hydrogen peroxide (H2O2)-mediated protein nitration in vitro, understood as one of the main pathways for protein modification in biological systems. However, the role of nitric oxide (●NO) in the heme/hemin-induced protein nitration has not been studied in-depth. This is despite its reductive nitrosylating effects following binding to hemin and the possible involvement of the reactive nitrogen species in the nitration of various functional proteins. Here, the ●NO-binding affinity of hemin has been studied along with the influence of ●NO on the internalization of hemin into MDA-MB-231 cells and the accompanying changes in the profile of intracellular nitrated proteins. Moreover, to further understand the mechanism involved, bovine serum albumin (BSA) nitration was studied after treatment with hemin and ●NO, with an investigation of the effects of pH of the reaction medium, generation of H2O2, and the oxidation of the tyrosine residues as the primary sites for the nitration. We demonstrated that hemin nitrosylation enhanced its cellular uptake and induced the one-electron oxidation and nitration of different intracellular proteins along with its ●NO-scavenging efficiency. Moreover, the hemin/NO-mediated BSA nitration was proved to be dependent on the concentration of ●NO and the pH of the reaction medium, with a vital role being played by the scavenging effects of protein for the free hemin molecules. Collectively, our results reaffirm the involvement of hemin and ●NO in the nitration mechanism, where the nitrosylation products can induce protein nitration while promoting the effects of the components of the nitrite/H2O2-mediated pathway.
Collapse
Affiliation(s)
- Amir M Alsharabasy
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, Ireland
| | - Sharon Glynn
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, Ireland; Discipline of Pathology, Lambe Institute for Translational Medicine, School of Medicine, National University of Ireland Galway, Ireland
| | - Pau Farràs
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, Ireland; School of Chemistry, Ryan Institute, National University of Ireland Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, Ireland.
| |
Collapse
|
7
|
Khramtsov P, Bochkova M, Timganova V, Kiselkov D, Zamorina S, Rayev M. Albumin Nanoparticles Loaded with Hemin as Peroxidase Mimics for Immunoassay**. ChemistrySelect 2022. [DOI: 10.1002/slct.202103892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Pavel Khramtsov
- Lab of Ecological immunology Institute of Ecology and Genetics of Microorganisms UB RAS 614081 13 Golev str. Perm Russia
- Department of Biology Perm State University 614068 15 Bukirev str. Perm Russia
| | - Maria Bochkova
- Lab of Ecological immunology Institute of Ecology and Genetics of Microorganisms UB RAS 614081 13 Golev str. Perm Russia
- Department of Biology Perm State University 614068 15 Bukirev str. Perm Russia
| | - Valeria Timganova
- Lab of Ecological immunology Institute of Ecology and Genetics of Microorganisms UB RAS 614081 13 Golev str. Perm Russia
| | - Dmitriy Kiselkov
- Lab of Structural Chemical Modification of Polymers Institute of Technical Chemistry UB RAS 614013 3 Academician Korolev str. Perm Russia
| | - Svetlana Zamorina
- Lab of Ecological immunology Institute of Ecology and Genetics of Microorganisms UB RAS 614081 13 Golev str. Perm Russia
- Department of Biology Perm State University 614068 15 Bukirev str. Perm Russia
| | - Mikhail Rayev
- Lab of Ecological immunology Institute of Ecology and Genetics of Microorganisms UB RAS 614081 13 Golev str. Perm Russia
- Department of Biology Perm State University 614068 15 Bukirev str. Perm Russia
| |
Collapse
|
8
|
Serum Albumin: A Multifaced Enzyme. Int J Mol Sci 2021; 22:ijms221810086. [PMID: 34576249 PMCID: PMC8466385 DOI: 10.3390/ijms221810086] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023] Open
Abstract
Human serum albumin (HSA) is the most abundant protein in plasma, contributing actively to oncotic pressure maintenance and fluid distribution between body compartments. HSA acts as the main carrier of fatty acids, recognizes metal ions, affects pharmacokinetics of many drugs, provides the metabolic modification of some ligands, renders potential toxins harmless, accounts for most of the anti-oxidant capacity of human plasma, and displays esterase, enolase, glucuronidase, and peroxidase (pseudo)-enzymatic activities. HSA-based catalysis is physiologically relevant, affecting the metabolism of endogenous and exogenous compounds including proteins, lipids, cholesterol, reactive oxygen species (ROS), and drugs. Catalytic properties of HSA are modulated by allosteric effectors, competitive inhibitors, chemical modifications, pathological conditions, and aging. HSA displays anti-oxidant properties and is critical for plasma detoxification from toxic agents and for pro-drugs activation. The enzymatic properties of HSA can be also exploited by chemical industries as a scaffold to produce libraries of catalysts with improved proficiency and stereoselectivity for water decontamination from poisonous agents and environmental contaminants, in the so called “green chemistry” field. Here, an overview of the intrinsic and metal dependent (pseudo-)enzymatic properties of HSA is reported to highlight the roles played by this multifaced protein.
Collapse
|
9
|
Silva RCMC, Tan L, Rodrigues DA, Prestes EB, Gomes CP, Gama AM, Oliveira PLD, Paiva CN, Manoury B, Bozza MT. Chloroquine inhibits pro-inflammatory effects of heme on macrophages and invivo. Free Radic Biol Med 2021; 173:104-116. [PMID: 34303829 DOI: 10.1016/j.freeradbiomed.2021.07.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Chloroquine has been used successfully to treat Malaria, including by chloroquine-resistant Plasmodium sp., indicating that it has effects on disease itself. Since heme has inflammatory effects and contributes to the pathogenesis of hemolytic diseases, we hypothesize that the anti-inflammatory effect of chloroquine is partially due to its inhibitory effect on heme-induced macrophage activation and on inflammatory tissue damage. METHODS Bone marrow derived macrophages (BMDMs) were incubated with chloroquine before stimulation with heme, in different conditions, to evaluate cytokines secretion, ROS production, mitogen activated protein kinases (MAPK) or spleen tyrosine kinase (Syk) activation, alone or combined with LPS. The effects of chloroquine upon heme inflammation were also evaluated in vivo, through simultaneous i.p. injection of LPS and heme, intratracheal instillation of Poly-IC followed by heme injection, and in a rhabdomyolysis model. RESULTS Chloroquine inhibited TNF secretion, mitochondrial ROS production, MAPK, and Syk activation induced by heme. Inhibition of TNF production could be mimicked by zinc ionophore quercetin, but not by primaquine, a chloroquine analog with low affinity for heme. IL-6 and IL-1β secretions induced by heme in the presence of PRRs agonists were inhibited by chloroquine, but not by calcium chelator BAPTA or inhibitor of endosomal acidification concamycin B. Chloroquine also protected mice from heme inflammatory effects in vivo, inhibiting lethal synergism with PRR agonists, lung pathology caused by heme injection after intratracheal instillation of Poly-IC, and delaying death after rhabdomyolisis. CONCLUSION Our data indicate that chloroquine might be used as a supportive therapy to control heme-induced deleterious inflammation in different hemolytic diseases.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia. Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil; Institut Necker Enfants Malades. INSERM U1151-CNRS UMR8253, Paris, France; Laboratório Intermediário de Imunoreceptores e Sinalização Celular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| | - Luis Tan
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia. Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| | - Danielle Aparecida Rodrigues
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia. Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| | - Elisa Beatriz Prestes
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia. Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil; Institut Necker Enfants Malades. INSERM U1151-CNRS UMR8253, Paris, France
| | - Caroline Pereira Gomes
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia. Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| | - Andreza Moreira Gama
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia. Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| | - Pedro Lagerblad de Oliveira
- Laboratório de Bioquímica de Insetos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Brazil
| | - Claudia Neto Paiva
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia. Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| | - Benedicte Manoury
- Institut Necker Enfants Malades. INSERM U1151-CNRS UMR8253, Paris, France
| | - Marcelo Torres Bozza
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia. Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil.
| |
Collapse
|
10
|
Neuroprotective Therapies for Spontaneous Intracerebral Hemorrhage. Neurocrit Care 2021; 35:862-886. [PMID: 34341912 DOI: 10.1007/s12028-021-01311-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 06/25/2021] [Indexed: 12/15/2022]
Abstract
Patients who survive the initial ictus of spontaneous intracerebral hemorrhage (ICH) remain vulnerable to subsequent injury of the perilesional parenchyma by molecular and cellular responses to the hematoma. Secondary brain injury after ICH, which contributes to long-term functional impairment and mortality, has emerged as an attractive therapeutic target. This review summarizes preclinical and clinical evidence for neuroprotective therapies targeting secondary injury pathways following ICH. A focus on therapies with pleiotropic antiinflammatory effects that target thrombin-mediated chemotaxis and inflammatory cell migration has led to studies investigating statins, anticholinergics, sphingosine-1-phosphate receptor modulators, peroxisome proliferator activated receptor gamma agonists, and magnesium. Attempts to modulate ICH-induced blood-brain barrier breakdown and perihematomal edema formation has prompted studies of nonsteroidal antiinflammatory agents, matrix metalloproteinase inhibitors, and complement inhibitors. Iron chelators, such as deferoxamine and albumin, have been used to reduce the free radical injury that ensues from erythrocyte lysis. Stem cell transplantation has been assessed for its potential to enhance subacute neurogenesis and functional recovery. Despite promising preclinical results of numerous agents, their outcomes have not yet translated into positive clinical trials in patients with ICH. Further studies are necessary to improve our understanding of the molecular events that promote damage and inflammation of the perihematomal parenchyma after ICH. Elucidating the temporal and pathophysiologic features of this secondary brain injury could enhance the clinical efficacy of neuroprotective therapies for ICH.
Collapse
|
11
|
Normant V, Brault A, Avino M, Mourer T, Vahsen T, Beaudoin J, Labbé S. Hemeprotein Tpx1 interacts with cell-surface heme transporter Str3 in Schizosaccharomyces pombe. Mol Microbiol 2021; 115:699-722. [PMID: 33140466 DOI: 10.1111/mmi.14638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 11/30/2022]
Abstract
Str3 is a transmembrane protein that mediates low-affinity heme uptake in Schizosaccharomyces pombe. Under iron-limiting conditions, Str3 remains at the cell surface in the presence of increasing hemin concentrations. Using a proximity-dependent biotinylation approach coupled to mass spectrometry and coimmunoprecipitation assays, we report that the peroxiredoxin Tpx1 is a binding partner of Str3. Under microaerobic conditions, cells deficient in heme biosynthesis and lacking the heme receptor Shu1 exhibit poor hemin-dependent growth in the absence of Tpx1. Analysis of membrane protein preparations from iron-starved hem1Δ shu1Δ str3Δ tpx1Δ cells coexpressing Str3-GFP and TAP-Tpx1 showed that TAP-Tpx1 is enriched in membrane protein fractions in response to hemin. Bimolecular fluorescence complementation assays brought additional evidence that an interaction between Tpx1 and Str3 occurs at the plasma membrane. Results showed that Tpx1 exhibits an equilibrium constant value of 0.26 μM for hemin. The association of Tpx1 with hemin protects hemin from degradation by H2 O2 . The peroxidase activity of hemin is lowered when it is bound to Tpx1. Taken together, these results revealed that Tpx1 is a novel interacting partner of Str3. Our data are the first example of an interaction between a cytoplasmic heme-binding protein and a cell-surface heme transporter.
Collapse
Affiliation(s)
- Vincent Normant
- Département de Biochimie et de Génomique Fonctionnelle, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Ariane Brault
- Département de Biochimie et de Génomique Fonctionnelle, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mariano Avino
- Département de Biochimie et de Génomique Fonctionnelle, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Thierry Mourer
- Département de Biochimie et de Génomique Fonctionnelle, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Tobias Vahsen
- Département de Biochimie et de Génomique Fonctionnelle, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jude Beaudoin
- Département de Biochimie et de Génomique Fonctionnelle, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Simon Labbé
- Département de Biochimie et de Génomique Fonctionnelle, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
12
|
Alvarado G, Tóth A, Csősz É, Kalló G, Dankó K, Csernátony Z, Smith A, Gram M, Akerström B, Édes I, Balla G, Papp Z, Balla J. Heme-Induced Oxidation of Cysteine Groups of Myofilament Proteins Leads to Contractile Dysfunction of Permeabilized Human Skeletal Muscle Fibres. Int J Mol Sci 2020; 21:ijms21218172. [PMID: 33142923 PMCID: PMC7663642 DOI: 10.3390/ijms21218172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/21/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023] Open
Abstract
Heme released from red blood cells targets a number of cell components including the cytoskeleton. The purpose of the present study was to determine the impact of free heme (20–300 µM) on human skeletal muscle fibres made available during orthopedic surgery. Isometric force production and oxidative protein modifications were monitored in permeabilized skeletal muscle fibre segments. A single heme exposure (20 µM) to muscle fibres decreased Ca2+-activated maximal (active) force (Fo) by about 50% and evoked an approximately 3-fold increase in Ca2+-independent (passive) force (Fpassive). Oxidation of sulfhydryl (SH) groups was detected in structural proteins (e.g., nebulin, α-actinin, meromyosin 2) and in contractile proteins (e.g., myosin heavy chain and myosin-binding protein C) as well as in titin in the presence of 300 µM heme. This SH oxidation was not reversed by dithiothreitol (50 mM). Sulfenic acid (SOH) formation was also detected in the structural proteins (nebulin, α-actinin, meromyosin). Heme effects on SH oxidation and SOH formation were prevented by hemopexin (Hpx) and α1-microglobulin (A1M). These data suggest that free heme has a significant impact on human skeletal muscle fibres, whereby oxidative alterations in structural and contractile proteins limit contractile function. This may explain and or contribute to the weakness and increase of skeletal muscle stiffness in chronic heart failure, rhabdomyolysis, and other hemolytic diseases. Therefore, therapeutic use of Hpx and A1M supplementation might be effective in preventing heme-induced skeletal muscle alterations.
Collapse
Affiliation(s)
- Gerardo Alvarado
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, H-4032 Debrecen, Hungary; (G.A.); (A.T.)
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Attila Tóth
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, H-4032 Debrecen, Hungary; (G.A.); (A.T.)
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Éva Csősz
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.C.); (G.K.)
| | - Gergő Kalló
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (É.C.); (G.K.)
| | - Katalin Dankó
- Department of Rheumatology, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Zoltán Csernátony
- Department of Orthopedics, Faculty of Medicine, University of Debrecen, H-4012 Debrecen, Hungary;
| | - Ann Smith
- Department of Cell and Molecular Biology and Biochemistry, School of Biological and Chemical Sciences, University of Missouri-Kansas City, Missouri, MO 64110, USA;
| | - Magnus Gram
- Department of Clinical Sciences Lund, Pediatrics, Lund University, 22184 Lund, Sweden;
| | - Bo Akerström
- Department of Clinical Sciences Lund, Infection Medicine, Lund University, 22184 Lund, Sweden;
| | - István Édes
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
| | - György Balla
- Institute of Pediatrics, Faculty of Medicine, University of Debrecen, H-4012 Debrecen, Hungary;
| | - Zoltán Papp
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, H-4032 Debrecen, Hungary; (G.A.); (A.T.)
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
- Correspondence: (Z.P.); (J.B.); Tel./Fax: +36-(52)-411717 (Z.P.); +36-(52)-413653 (J.B.)
| | - József Balla
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, H-4032 Debrecen, Hungary; (G.A.); (A.T.)
- Department of Nephrology, Institute of Medicine, Faculty of Medicine, University of Debrecen, H-4012 Debrecen, Hungary
- Correspondence: (Z.P.); (J.B.); Tel./Fax: +36-(52)-411717 (Z.P.); +36-(52)-413653 (J.B.)
| |
Collapse
|
13
|
Jennifer B, Berg V, Modak M, Puck A, Seyerl-Jiresch M, Künig S, Zlabinger GJ, Steinberger P, Chou J, Geha RS, Öhler L, Yachie A, Choe H, Kraller M, Stockinger H, Stöckl J. Transferrin receptor 1 is a cellular receptor for human heme-albumin. Commun Biol 2020; 3:621. [PMID: 33110194 PMCID: PMC7591885 DOI: 10.1038/s42003-020-01294-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Iron is essential for living cells. Uptake of iron-loaded transferrin by the transferrin receptor 1 (CD71, TFR) is a major but not sufficient mechanism and an alternative iron-loaded ligand for CD71 has been assumed. Here, we demonstrate that CD71 utilizes heme-albumin as cargo to transport iron into human cells. Binding and endocytosis of heme-albumin via CD71 was sufficient to promote proliferation of various cell types in the absence of transferrin. Growth and differentiation of cells induced by heme-albumin was dependent on heme-oxygenase 1 (HO-1) function and was accompanied with an increase of the intracellular labile iron pool (LIP). Import of heme-albumin via CD71 was further found to contribute to the efficacy of albumin-based drugs such as the chemotherapeutic Abraxane. Thus, heme-albumin/CD71 interaction is a novel route to transport nutrients or drugs into cells and adds to the emerging function of CD71 as a scavenger receptor. Brell, Berg et al find that iron enters cells not only through iron-transferrin uptake by the transferrin receptor (CD71) but also through uptake of heme-albumin by this receptor and that heme-albumin stimulates proliferation in a manner dependent on heme oxygenase 1. This study presents a new route for iron uptake in mammalian cells.
Collapse
Affiliation(s)
- Brell Jennifer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Verena Berg
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Madhura Modak
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Alexander Puck
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Maria Seyerl-Jiresch
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Sarojinidevi Künig
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Gerhard J Zlabinger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Peter Steinberger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Janet Chou
- Division of Immunology, Boston Children´s Hospital, Boston, MA, 02115, USA
| | - Raif S Geha
- Division of Immunology, Boston Children´s Hospital, Boston, MA, 02115, USA
| | - Leopold Öhler
- Department of Internal Medicine, St. Josef Hospital, 1130, Vienna, Austria
| | - Akihiro Yachie
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical, and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Hyeryun Choe
- Department of Immunology and Microbiology, The Scripps Research Institute, Florida, CA, 92037, USA
| | - Markus Kraller
- Institute of Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Hannes Stockinger
- Institute of Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Johannes Stöckl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
14
|
Englert FA, Seidel RA, Galler K, Gouveia Z, Soares MP, Neugebauer U, Clemens MG, Sponholz C, Heinemann SH, Pohnert G, Bauer M, Weis S. Labile heme impairs hepatic microcirculation and promotes hepatic injury. Arch Biochem Biophys 2019; 672:108075. [DOI: 10.1016/j.abb.2019.108075] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 08/04/2019] [Accepted: 08/10/2019] [Indexed: 12/13/2022]
|
15
|
Zhang P, Ma L, Yang Z, Li H, Gao Z. Study on the detoxification mechanisms to 5,10,15,20-tetrakis (4-sulfonatophenyl) porphyrinato iron(III) chloride (FeTPPS), an efficient pro-oxidant of heme water-soluble analogue. J Inorg Biochem 2018; 189:40-52. [DOI: 10.1016/j.jinorgbio.2018.08.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 08/19/2018] [Accepted: 08/30/2018] [Indexed: 11/30/2022]
|
16
|
Tyrosine residues of bovine serum albumin play an important role in protecting SH-SY5Y cells against heme/H2O2/NO2−-induced damage. Mol Cell Biochem 2018; 454:57-66. [DOI: 10.1007/s11010-018-3452-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 10/05/2018] [Indexed: 02/08/2023]
|
17
|
Vlasova II. Peroxidase Activity of Human Hemoproteins: Keeping the Fire under Control. Molecules 2018; 23:E2561. [PMID: 30297621 PMCID: PMC6222727 DOI: 10.3390/molecules23102561] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/28/2018] [Accepted: 10/01/2018] [Indexed: 12/21/2022] Open
Abstract
The heme in the active center of peroxidases reacts with hydrogen peroxide to form highly reactive intermediates, which then oxidize simple substances called peroxidase substrates. Human peroxidases can be divided into two groups: (1) True peroxidases are enzymes whose main function is to generate free radicals in the peroxidase cycle and (pseudo)hypohalous acids in the halogenation cycle. The major true peroxidases are myeloperoxidase, eosinophil peroxidase and lactoperoxidase. (2) Pseudo-peroxidases perform various important functions in the body, but under the influence of external conditions they can display peroxidase-like activity. As oxidative intermediates, these peroxidases produce not only active heme compounds, but also protein-based tyrosyl radicals. Hemoglobin, myoglobin, cytochrome c/cardiolipin complexes and cytoglobin are considered as pseudo-peroxidases. Рeroxidases play an important role in innate immunity and in a number of physiologically important processes like apoptosis and cell signaling. Unfavorable excessive peroxidase activity is implicated in oxidative damage of cells and tissues, thereby initiating the variety of human diseases. Hence, regulation of peroxidase activity is of considerable importance. Since peroxidases differ in structure, properties and location, the mechanisms controlling peroxidase activity and the biological effects of peroxidase products are specific for each hemoprotein. This review summarizes the knowledge about the properties, activities, regulations and biological effects of true and pseudo-peroxidases in order to better understand the mechanisms underlying beneficial and adverse effects of this class of enzymes.
Collapse
Affiliation(s)
- Irina I Vlasova
- Federal Research and Clinical Center of Physical-Chemical Medicine, Department of Biophysics, Malaya Pirogovskaya, 1a, Moscow 119435, Russia.
- Institute for Regenerative Medicine, Laboratory of Navigational Redox Lipidomics, Sechenov University, 8-2 Trubetskaya St., Moscow 119991, Russia.
| |
Collapse
|
18
|
Ferreira CM, Stiebler R, Saraiva FM, Lechuga GC, Walter-Nuno AB, Bourguignon SC, Gonzalez MS, Azambuja P, Gandara ACP, Menna-Barreto RFS, Paiva-Silva GO, Paes MC, Oliveira MF. Heme crystallization in a Chagas disease vector acts as a redox-protective mechanism to allow insect reproduction and parasite infection. PLoS Negl Trop Dis 2018; 12:e0006661. [PMID: 30036366 PMCID: PMC6084092 DOI: 10.1371/journal.pntd.0006661] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/02/2018] [Accepted: 07/03/2018] [Indexed: 12/18/2022] Open
Abstract
Heme crystallization as hemozoin represents the dominant mechanism of heme disposal in blood feeding triatomine insect vectors of the Chagas disease. The absence of drugs or vaccine for the Chagas disease causative agent, the parasite Trypanosoma cruzi, makes the control of vector population the best available strategy to limit disease spread. Although heme and redox homeostasis regulation is critical for both triatomine insects and T. cruzi, the physiological relevance of hemozoin for these organisms remains unknown. Here, we demonstrate that selective blockage of heme crystallization in vivo by the antimalarial drug quinidine, caused systemic heme overload and redox imbalance in distinct insect tissues, assessed by spectrophotometry and fluorescence microscopy. Quinidine treatment activated compensatory defensive heme-scavenging mechanisms to cope with excessive heme, as revealed by biochemical hemolymph analyses, and fat body gene expression. Importantly, egg production, oviposition, and total T. cruzi parasite counts in R. prolixus were significantly reduced by quinidine treatment. These effects were reverted by oral supplementation with the major insect antioxidant urate. Altogether, these data underscore the importance of heme crystallization as the main redox regulator for triatomine vectors, indicating the dual role of hemozoin as a protective mechanism to allow insect fertility, and T. cruzi life-cycle. Thus, targeting heme crystallization in insect vectors represents an innovative way for Chagas disease control, by reducing simultaneously triatomine reproduction and T. cruzi transmission. Chagas disease is a fatal illness caused by Trypanosoma cruzi parasites, which are transmitted by blood sucking triatomine insect vectors. Although blood is a natural food source for these insects, its digestion releases toxic products, which poses a dietary challenge for both triatomine insects and trypanosomes. To overcome this, triatomines eliminate these toxic blood products by a unique process of heme crystallization into hemozoin that take place in their digestive tract. Here we describe that this detoxification process represents the major mechanism for redox balance control, and is necessary to allow triatomine insect reproduction, and Trypanosoma cruzi infection. Disruption of heme crystallization in triatomine insects thus represents a new venue for Chagas disease control, by targeting at the same time insect reproduction and parasite transmission.
Collapse
Affiliation(s)
- Caroline M. Ferreira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renata Stiebler
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Francis M. Saraiva
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Guilherme C. Lechuga
- Departamento de Biologia Celular e Molecular, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Ana Beatriz Walter-Nuno
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Saulo C. Bourguignon
- Departamento de Biologia Celular e Molecular, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Marcelo S. Gonzalez
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Biologia Geral, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Patrícia Azambuja
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Caroline P. Gandara
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Gabriela O. Paiva-Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia C. Paes
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcus F. Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
19
|
Liu Z, Chen C, Li X, Zhao C, Li Z, Liang W, Lin Y. Is cupping blister harmful?—A proteomical analysis of blister fluid induced by cupping therapy and scald. Complement Ther Med 2018; 36:25-29. [DOI: 10.1016/j.ctim.2017.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/02/2017] [Accepted: 11/01/2017] [Indexed: 11/16/2022] Open
|
20
|
Huang Y, Zhang P, Yang Z, Wang P, Li H, Gao Z. Interaction of glyceraldehyde-3-phosphate dehydrogenase and heme: The relevance of its biological function. Arch Biochem Biophys 2017; 619:54-61. [PMID: 28315300 DOI: 10.1016/j.abb.2017.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/11/2017] [Accepted: 03/11/2017] [Indexed: 10/20/2022]
Abstract
GAPDH was speculated to function as a transient trap to reduce the potential toxicity of free heme by a specific and reversible binding with heme. Up to now, there has been lack of studies focused on this effect. In this paper, the efficiency of GAPDH-heme complex on catalyzing protein carbonylation and nitration, the cross-linking of heme to protein formation, and cytotoxicity of GAPDH-heme were studied. It was found that the binding of GAPDH could inhibit H2O2-mediated degradation of heme. Peroxidase activity of GAPDH-heme complex was higher than that of free heme, but significantly lower than that of HSA-heme. Catalytic activity of heme corresponded complex toward tyrosine oxidation/nitration was decreased in the order of HSA-heme, heme and GAPDH-heme. GAPDH also inhibited heme-H2O2-NO2- induced protein carbonylation. No covalent bond was formed between heme and GAPDH after treated with H2O2. GAPDH was more effective than HSA on protecting cells against heme-NO2--H2O2 induced cytotoxicity. These results indicate that binding of GAPDH inhibits the activity of heme in catalyzing tyrosine nitration and protects the coexistent protein against oxidative damage, and the mechanism is different from that of HSA. This study may help clarifying the protective role of GAPDH acting as a chaperone in heme transfer to downstream areas.
Collapse
Affiliation(s)
- Yi Huang
- School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Pengfei Zhang
- School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Zhen Yang
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77004, United States
| | - Peipei Wang
- School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Hailing Li
- School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China; Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Wuhan, 430074, PR China
| | - Zhonghong Gao
- School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China; Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Wuhan, 430074, PR China.
| |
Collapse
|
21
|
Wang X, Liu W, Yin B, Sang Y, Liu Z, Dai Y, Duan X, Zhang G, Ding S, Tao Z. An isothermal strand displacement amplification strategy for nucleic acids using junction forming probes and colorimetric detection. Mikrochim Acta 2017. [DOI: 10.1007/s00604-017-2158-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
22
|
Wang L, Li M, Xie Y, Xu L, Ye R, Liu X. Preclinical efficacy of human Albumin in subarachnoid hemorrhage. Neuroscience 2017; 344:255-264. [DOI: 10.1016/j.neuroscience.2016.12.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 12/17/2016] [Accepted: 12/19/2016] [Indexed: 11/26/2022]
|
23
|
Watanabe Y, Ishimori K, Uchida T. Dual role of the active-center cysteine in human peroxiredoxin 1: Peroxidase activity and heme binding. Biochem Biophys Res Commun 2017; 483:930-935. [DOI: 10.1016/j.bbrc.2017.01.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 01/09/2017] [Indexed: 01/07/2023]
|
24
|
|
25
|
Ye H, Yang Z, Li H, Gao Z. NPY binds with heme to form a NPY–heme complex: enhancing peroxidase activity in free heme and promoting NPY nitration and inactivation. Dalton Trans 2017; 46:10315-10323. [DOI: 10.1039/c7dt01822c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
NPY binding with heme enhances the peroxidase activity of free heme, resulting in the important tyrosine nitration, which will attenuate its bioactivity.
Collapse
Affiliation(s)
- Huixian Ye
- School of Chemistry and Chemical Engineering
- Huazhong University of Science and Technology
- Wuhan 430074
- People's Republic of China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica
| | - Zhen Yang
- Department of Chemical and Biomolecular Engineering
- University of Houston
- Houston
- USA
| | - Hailing Li
- School of Chemistry and Chemical Engineering
- Huazhong University of Science and Technology
- Wuhan 430074
- People's Republic of China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica
| | - Zhonghong Gao
- School of Chemistry and Chemical Engineering
- Huazhong University of Science and Technology
- Wuhan 430074
- People's Republic of China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica
| |
Collapse
|
26
|
Salifu H, Wilson NO, Liu M, Dickinson-Copeland C, Yatich N, Keenan J, Turpin C, Jolly P, Gyasi R, Adjei AA, Stiles JK. Iron Supplementation Alters Heme and Heme Oxygenase 1 (HO-1) Levels In Pregnant Women in Ghana. SOJ MICROBIOLOGY & INFECTIOUS DISEASES 2016; 4. [PMID: 28124024 DOI: 10.15226/sojmid/4/2/00154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Iron supplementation is recommended for pregnant women to meet their iron requirement for a healthy pregnancy. The benefits and risks of universal iron supplementation during pregnancy in malaria endemic countries are currently being debated. As part of a broader study that focused on the effect of heme/HO-1 on pregnancy outcomes in malaria in pregnancy, we determined the association between iron supplementation and free heme levels in blood of pregnant women with and without malaria in Ghana. We hypothesized that pregnant women with malaria who took iron supplements will have higher levels of Heme/HO-1 than those who did not take iron supplements. METHODS A total of 337 women were recruited for this study. Blood samples were collected for malaria diagnosis and heme/HO-1 measurement. Quantification of heme was done using a heme colorimetric assay kit and HO-1 levels were performed using Enzyme-Linked Immunosorbent Assay (ELISA) on plasma samples. RESULTS Malaria positive iron supplemented women, in their third trimester, had significantly higher median levels of heme 59.3(43.1 - 60.4) than non-malaria iron supplemented women 35.7(33.0 - 62.2), p = 0.026. Also, malaria positive iron supplemented women had significant higher median levels of HO-16.2(IQR 4.9 - 8.1) than pregnant women who did not take iron supplements 2.9 (IQR 2.1 - 3.8), p = <0.001. CONCLUSION Although iron supplementation may be highly beneficial and improve pregnancy outcomes for iron deficient or anemic mothers, it is also likely that iron supplementation for pregnant women who are not iron deficient may put this group of women at risk for adverse pregnancy outcomes. Findings from this study sheds light on the effect of iron supplementation on malaria derived heme in pregnancy, which may inform how iron supplementation is recommended for pregnant women who are not iron deficient.
Collapse
Affiliation(s)
- Hassana Salifu
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine
| | - Nana O Wilson
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine
| | - Mingli Liu
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine
| | | | - Nelly Yatich
- University of Alabama, Birmingham Alabama, Department of Epidemiology
| | - John Keenan
- University of Alabama, Birmingham Alabama, Department of Epidemiology
| | - Cornelius Turpin
- University of Alabama, Birmingham Alabama, Department of Epidemiology
| | - Pauline Jolly
- University of Alabama, Birmingham Alabama, Department of Epidemiology
| | - Richard Gyasi
- University of Ghana Medical School, Department of Pathology, Korle-Bu Teaching Hospital, Accra, Ghana
| | - Andrew A Adjei
- University of Ghana Medical School, Department of Pathology, Korle-Bu Teaching Hospital, Accra, Ghana
| | - Jonathan K Stiles
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine
| |
Collapse
|
27
|
Ding Y, Xiao C, Wu Q, Xie Y, Li X, Hu H, Li L. The Mechanisms Underlying the Hypolipidaemic Effects of Grifola frondosa in the Liver of Rats. Front Microbiol 2016; 7:1186. [PMID: 27536279 PMCID: PMC4971090 DOI: 10.3389/fmicb.2016.01186] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 07/18/2016] [Indexed: 11/16/2022] Open
Abstract
The present study investigated the hypolipidaemic effects of Grifola frondosa and its regulation mechanism involved in lipid metabolism in liver of rats fed a high-cholesterol diet. The body weights and serum lipid levels of control rats, of hyperlipidaemic rats, and of hyperlipidaemic rats treated with oral G. frondosa were determined. mRNA expression and concentration of key lipid metabolism enzymes were investigated. Serum cholesterol, triacylglycerol, and low-density lipoprotein cholesterol levels were markedly decreased in hyperlipidaemic rats treated with G. frondosa compared with untreated hyperlipidaemic rats. mRNA expression of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), acyl-coenzyme A: cholesterol acyltransferase (ACAT2), apolipoprotein B (ApoB), fatty acid synthase (FAS), and acetyl-CoA carboxylase (ACC1) were significantly down-regulated, while expression of cholesterol 7-alpha-hydroxylase (CYP7A1) was significantly up-regulated in the livers of treated rats compared with untreated hyperlipidaemic rats. The concentrations of these enzymes also paralleled the observed changes in mRNA expression. Two-dimensional polyacrylamide gel electrophoresis (2-DE) and Matrix-Assisted Laser Desorption/Ionization Time of Flight Mass Spectrometry (MALDI-TOF-MS) were used to identify 20 proteins differentially expressed in livers of rats treated with G. frondosa compared with untreated hyperlipidemic rats. Of these 20 proteins, seven proteins were down-regulated, and 13 proteins were up-regulated. These findings indicate that the hypolipidaemic effects of G. frondosa reflected its modulation of key enzymes involved in cholesterol and triacylglycerol biosynthesis, absorption, and catabolic pathways. G. frondosa may exert anti-atherosclerotic effects by inhibiting LDL oxidation through down-regulation and up-regulating proteins expression in the liver of rats. Therefore, G. frondosa may produce both hypolipidaemic and anti-atherosclerotic effects, and potentially be of use as a functional food for the treatment or prevention of hyperlipidaemia and atherosclerosis.
Collapse
Affiliation(s)
- Yinrun Ding
- School of Bioscience and Bioengineering, South China University of TechnologyGuangzhou, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of MicrobiologyGuangzhou, China
- Department of Biology, Basic Medical College, Guangdong Medical UniversityZhangjiang, China
| | - Chun Xiao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of MicrobiologyGuangzhou, China
| | - Qingping Wu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of MicrobiologyGuangzhou, China
| | - Yizhen Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of MicrobiologyGuangzhou, China
| | - Xiangmin Li
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of MicrobiologyGuangzhou, China
| | - Huiping Hu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of MicrobiologyGuangzhou, China
| | - Liangqiu Li
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of MicrobiologyGuangzhou, China
| |
Collapse
|
28
|
Draaisma AM, Molicki JS, Verbeet N, Munneke R, Huysmans HA, Berger HM, Hazekamp MG. Increasing the antioxidative capacity of neonatal cardiopulmonary bypass prime solution: anin vitro study. Perfusion 2016; 18:357-62. [PMID: 14714771 DOI: 10.1191/0267659103pf693oa] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Inflammation and oxidative damage are believed to play an important role in the postoperative complications after cardiopulmonary bypass (CPB) in neonates. During the preparation of the prime, red blood cells (RBCs) release non-protein-bound iron (NPBI) and free haemoglobin/haem (Hb/haem). The presence of these prooxidants in the prime solution may increase oxidative stress in neonates undergoing CPB. The solution used as the basis of the prime solution may influence the degree of this oxidative stress. We investigated the NPBI and the Hb/haem binding capacities of two different prime solutions: a prime based on pasteurized human albumin and a prime based on fresh frozen plasma. The presence of NPBI and free Hb/haem were measured during and after the preparation of the prime solution. Only in the albumin prime was NPBI detectable. However, in both primes, the concentrations of free Hb/haem increased. Thus, to reduce the prooxidative effects of NPBI and free Hb/haem, RBCs should be added to the prime at the last possible moment. Adding fresh frozen plasma should be considered, as this would result in no detectable NPBI in the prime solution.
Collapse
Affiliation(s)
- Anjo M Draaisma
- Department of Extracorporeal Circulation, Leiden University Medical Centre, Leiden, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
29
|
Dong M, Zhang L, Li R, Li S, Jiang Y, Qiao Y, Duan Z, Li R, Wang Q, Wang H. Crosslinking catalysis-active center of hemin on the protein scaffold toward peroxidase mimic with powerful catalysis. RSC Adv 2016. [DOI: 10.1039/c6ra07139b] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Catalytic hemin (Hem) was cross-linked onto the protein scaffold of bovine serum albumin (BSA) forming a Hem–BSA composite with powerful catalysis.
Collapse
|
30
|
Wang XF, Zhang XY, Gao X, Liu XX, Wang YH. Proteomic Profiling of a Respiratory Syncytial Virus-Infected Rat Pneumonia Model. Jpn J Infect Dis 2016; 69:285-92. [DOI: 10.7883/yoken.jjid.2015.244] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Xue-Feng Wang
- The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine
| | - Xiu-Ying Zhang
- The Graduate College of Liaoning University of Traditional Chinese Medicine
| | - Xuejuan Gao
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, Jinan University
| | - Xiao-Xue Liu
- The Graduate College of Liaoning University of Traditional Chinese Medicine
| | - Yi-Huan Wang
- The Graduate College of Liaoning University of Traditional Chinese Medicine
| |
Collapse
|
31
|
Human Albumin Improves Long-Term Behavioral Sequelae After Subarachnoid Hemorrhage Through Neurovascular Remodeling. Crit Care Med 2015; 43:e440-9. [PMID: 26181220 DOI: 10.1097/ccm.0000000000001193] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Subarachnoid hemorrhage results in significant long-lasting neurologic sequelae. Here, we investigated whether human albumin improves long-term outcomes in experimental subarachnoid hemorrhage and whether neurovascular remodeling is involved in the protection of albumin. DESIGN Laboratory investigation. SETTING Hospital research laboratory. SUBJECTS Male Sprague-Dawley rats. INTERVENTIONS Rats underwent subarachnoid hemorrhage by endovascular perforation. Albumin of either 0.63 or 1.25 g/kg was injected IV immediately after the surgery. Modified Garcia test, beam-walking test, novel object recognition, and Morris water maze were employed to determine the behavioral deficits. The effects of albumin on early neurovascular dysfunction and chronic synaptic plasticity were also studied. MEASUREMENTS AND MAIN RESULTS Both doses of albumin significantly improved the sensorimotor scores (F = 31.277; p = 0.001) and cognitive performance (F = 7.982; p = 0.001 in novel object recognition test; and F = 3.431; p = 0.026 in the latency analysis of Morris water maze test) for at least 40 days after subarachnoid hemorrhage. There were remarkable microvasculature hypoperfusion, intracranial pressure rise, early vasoconstriction, neural apoptosis, and degeneration in subarachnoid hemorrhage rats, with albumin significantly attenuating such neurovascular dysfunction. Furthermore, albumin markedly prevented blood-brain barrier disruption, as indicated by less blood-brain barrier leakage, preserved blood-brain barrier-related proteins, and dampened gelatinase activities. The expressions of key synaptic elements were up-regulated with albumin supplementation in both acute and chronic phases. Accordingly, a higher dendritic spine density was observed in the prefrontal and hippocampal areas of albumin-treated subarachnoid hemorrhage animals. CONCLUSIONS Albumin at low-to-moderate doses markedly improves long-term neurobehavioral sequelae after subarachnoid hemorrhage, which may involve an integrated process of neurovascular remodeling.
Collapse
|
32
|
Gaudreault V, Wirbel J, Jardim A, Rohrbach P, Scorza T. Red Blood Cells Preconditioned with Hemin Are Less Permissive to Plasmodium Invasion In Vivo and In Vitro. PLoS One 2015; 10:e0140805. [PMID: 26465787 PMCID: PMC4605744 DOI: 10.1371/journal.pone.0140805] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
Malaria is a parasitic disease that causes severe hemolytic anemia in Plasmodium-infected hosts, which results in the release and accumulation of oxidized heme (hemin). Although hemin impairs the establishment of Plasmodium immunity in vitro and in vivo, mice preconditioned with hemin develop lower parasitemia when challenged with Plasmodium chabaudi adami blood stage parasites. In order to understand the mechanism accounting for this resistance as well as the impact of hemin on eryptosis and plasma levels of scavenging hemopexin, red blood cells were labeled with biotin prior to hemin treatment and P. c. adami infection. This strategy allowed discriminating hemin-treated from de novo generated red blood cells and to follow the infection within these two populations of cells. Fluorescence microscopy analysis of biotinylated-red blood cells revealed increased P. c. adami red blood cells selectivity and a decreased permissibility of hemin-conditioned red blood cells for parasite invasion. These effects were also apparent in in vitro P. falciparum cultures using hemin-preconditioned human red blood cells. Interestingly, hemin did not alter the turnover of red blood cells nor their replenishment during in vivo infection. Our results assign a function for hemin as a protective agent against high parasitemia, and suggest that the hemolytic nature of blood stage human malaria may be beneficial for the infected host.
Collapse
Affiliation(s)
- Véronique Gaudreault
- Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, Québec, Canada
| | - Jakob Wirbel
- Institute of parasitology, McGill University, Montréal, Québec, Canada
| | - Armando Jardim
- Institute of parasitology, McGill University, Montréal, Québec, Canada
| | - Petra Rohrbach
- Institute of parasitology, McGill University, Montréal, Québec, Canada
| | - Tatiana Scorza
- Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
33
|
Moosavi-Movahedi Z, Gharibi H, Hadi-Alijanvand H, Akbarzadeh M, Esmaili M, Atri MS, Sefidbakht Y, Bohlooli M, Nazari K, Javadian S, Hong J, Saboury AA, Sheibani N, Moosavi-Movahedi AA. Caseoperoxidase, mixed β-casein-SDS-hemin-imidazole complex: a nano artificial enzyme. J Biomol Struct Dyn 2015; 33:2619-32. [PMID: 25562503 DOI: 10.1080/07391102.2014.1003196] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
A novel peroxidase-like artificial enzyme, named "caseoperoxidase", was biomimetically designed using a nano artificial amino acid apo-protein hydrophobic pocket. This four-component nano artificial enzyme containing heme-imidazole-β-casein-SDS exhibited high activity growth and k(cat) performance toward the native horseradish peroxidase demonstrated by the steady state kinetics using UV-vis spectrophotometry. The hydrophobicity and secondary structure of the caseoperoxidase were studied by ANS fluorescence and circular dichroism spectroscopy. Camel β-casein (Cβ-casein) was selected as an appropriate apo-protein for the heme active site because of its innate flexibility and exalted hydrophobicity. This selection was confirmed by homology modeling method. Heme docking into the newly obtained Cβ-casein structure indicated one heme was mainly incorporated with Cβ-casein. The presence of a main electrostatic site for the active site in the Cβ-casein was also confirmed by experimental methods through Wyman binding potential and isothermal titration calorimetry. The existence of Cβ-casein protein in this biocatalyst lowered the suicide inactivation and provided a suitable protective role for the heme active-site. Additional experiments confirmed the retention of caseoperoxidase structure and function as an artificial enzyme.
Collapse
Affiliation(s)
| | - Hussein Gharibi
- b Faculty of Science, Department of Chemistry , Tarbiat Modares University , Tehran , Iran
| | - Hamid Hadi-Alijanvand
- c Department of Biological Sciences , Institute for Advanced Studies in Basic Sciences (IASBS) , Zanjan , Iran
| | - Mohammad Akbarzadeh
- d Institute of Biochemistry and Biophysics, University of Tehran , Tehran , Iran
| | - Mansoore Esmaili
- d Institute of Biochemistry and Biophysics, University of Tehran , Tehran , Iran
| | - Maliheh S Atri
- d Institute of Biochemistry and Biophysics, University of Tehran , Tehran , Iran
| | - Yahya Sefidbakht
- d Institute of Biochemistry and Biophysics, University of Tehran , Tehran , Iran
| | - Mousa Bohlooli
- d Institute of Biochemistry and Biophysics, University of Tehran , Tehran , Iran
| | | | - Soheila Javadian
- b Faculty of Science, Department of Chemistry , Tarbiat Modares University , Tehran , Iran
| | - Jun Hong
- f College of Life Science , Henan University , Kaifeng 475000 , China
| | - Ali A Saboury
- d Institute of Biochemistry and Biophysics, University of Tehran , Tehran , Iran.,g Center of Excellence in Biothermodynamics, University of Tehran , Tehran , Iran
| | - Nader Sheibani
- h Department of Ophthalmology and Visual Sciences , Biomedical Engineering, University of Wisconsin School of Medicine and Public Health , Madison , WI , USA.,i McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health , Madison , WI , USA
| | - Ali A Moosavi-Movahedi
- d Institute of Biochemistry and Biophysics, University of Tehran , Tehran , Iran.,g Center of Excellence in Biothermodynamics, University of Tehran , Tehran , Iran.,i McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health , Madison , WI , USA
| |
Collapse
|
34
|
Lu N, Li J, Tian R, Peng YY. Key Roles for Tyrosine 10 in Aβ–Heme Complexes and Its Relevance to Oxidative Stress. Chem Res Toxicol 2014; 28:365-72. [DOI: 10.1021/tx5003035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Naihao Lu
- Key Laboratory of Functional Small Organic Molecule,
Ministry of
Education and College of Life Science, ‡Key Laboratory of Green Chemistry,
Jiangxi Province and College of Chemistry and Chemical Engineering, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi 330022, China
| | - Jiayu Li
- Key Laboratory of Functional Small Organic Molecule,
Ministry of
Education and College of Life Science, ‡Key Laboratory of Green Chemistry,
Jiangxi Province and College of Chemistry and Chemical Engineering, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi 330022, China
| | - Rong Tian
- Key Laboratory of Functional Small Organic Molecule,
Ministry of
Education and College of Life Science, ‡Key Laboratory of Green Chemistry,
Jiangxi Province and College of Chemistry and Chemical Engineering, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi 330022, China
| | - Yi-Yuan Peng
- Key Laboratory of Functional Small Organic Molecule,
Ministry of
Education and College of Life Science, ‡Key Laboratory of Green Chemistry,
Jiangxi Province and College of Chemistry and Chemical Engineering, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi 330022, China
| |
Collapse
|
35
|
Zhao X, Aronowski J. Nrf2 to pre-condition the brain against injury caused by products of hemolysis after ICH. Transl Stroke Res 2014; 4:71-5. [PMID: 23378859 DOI: 10.1007/s12975-012-0245-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Brain damage caused by intracerebral hemorrhage (ICH) is mediated in part by the toxicity of extravascular blood deposited in brain parenchyma during the hematoma formation. In this paper we discuss the therapeutic benefits and potential mechanisms associated with the activation of transcription factor Nrf2 regarding its role in defending brain tissue against toxicity of blood, a component of secondary injury. We emphasize the pleiotropic capacity of Nrf2 as it recruits multiple pathways aiming at reducing deleterious effects of blood lysis products.
Collapse
Affiliation(s)
- Xiurong Zhao
- University of Texas Medical School - Houston; Department of Neurology, Stroke Program
| | | |
Collapse
|
36
|
Sands SA, Williams R, Marshall S, LeVine SM. Perivascular iron deposits are associated with protein nitration in cerebral experimental autoimmune encephalomyelitis. Neurosci Lett 2014; 582:133-8. [PMID: 24846416 DOI: 10.1016/j.neulet.2014.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/01/2014] [Accepted: 05/02/2014] [Indexed: 12/12/2022]
Abstract
Nitration of proteins, which is thought to be mediated by peroxynitrite, is a mechanism of tissue damage in multiple sclerosis (MS). However, protein nitration can also be catalyzed by iron, heme or heme-associated molecules independent of peroxynitrite. Since microhemorrhages and perivascular iron deposits are present in the CNS of MS patients, we sought to determine if iron is associated with protein nitration. A cerebral model of experimental autoimmune encephalomyelitis (cEAE) was utilized since this model has been shown to have perivascular iron deposits similar to those present in MS. Histochemical staining for iron was used together with immunohistochemistry for nitrotyrosine, eNOS, or iNOS on cerebral sections. Leakage of the blood-brain barrier (BBB) was studied by albumin immunohistochemistry. Iron deposits were colocalized with nitrotyrosine staining around vessels in cEAE mice while control animals revealed minimal staining. This finding supports the likelihood that nitrotyrosine formation was catalyzed by iron or iron containing molecules. Examples of iron deposits were also observed in association with eNOS and iNOS, which could be one source of substrates for this reaction. Extravasation of albumin was present in cEAE mice, but not in control animals. Extravasated albumin may act to limit tissue injury by binding iron and/or heme as well as being a target of nitration, but the protection is incomplete. In summary, iron-catalyzed nitration of proteins is a likely mechanism of tissue damage in MS.
Collapse
Affiliation(s)
- Scott A Sands
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Rachel Williams
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Sylvester Marshall
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Steven M LeVine
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA.
| |
Collapse
|
37
|
Chiabrando D, Vinchi F, Fiorito V, Mercurio S, Tolosano E. Heme in pathophysiology: a matter of scavenging, metabolism and trafficking across cell membranes. Front Pharmacol 2014; 5:61. [PMID: 24782769 PMCID: PMC3986552 DOI: 10.3389/fphar.2014.00061] [Citation(s) in RCA: 308] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/18/2014] [Indexed: 01/19/2023] Open
Abstract
Heme (iron-protoporphyrin IX) is an essential co-factor involved in multiple biological processes: oxygen transport and storage, electron transfer, drug and steroid metabolism, signal transduction, and micro RNA processing. However, excess free-heme is highly toxic due to its ability to promote oxidative stress and lipid peroxidation, thus leading to membrane injury and, ultimately, apoptosis. Thus, heme metabolism needs to be finely regulated. Intracellular heme amount is controlled at multiple levels: synthesis, utilization by hemoproteins, degradation and both intracellular and intercellular trafficking. This review focuses on recent findings highlighting the importance of controlling intracellular heme levels to counteract heme-induced oxidative stress. The contributions of heme scavenging from the extracellular environment, heme synthesis and incorporation into hemoproteins, heme catabolism and heme transport in maintaining adequate intracellular heme content are discussed. Particular attention is put on the recently described mechanisms of heme trafficking through the plasma membrane mediated by specific heme importers and exporters. Finally, the involvement of genes orchestrating heme metabolism in several pathological conditions is illustrated and new therapeutic approaches aimed at controlling heme metabolism are discussed.
Collapse
Affiliation(s)
- Deborah Chiabrando
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin Turin, Italy
| | - Francesca Vinchi
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin Turin, Italy
| | - Veronica Fiorito
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin Turin, Italy
| | - Sonia Mercurio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin Turin, Italy
| | - Emanuela Tolosano
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin Turin, Italy
| |
Collapse
|
38
|
Lu N, He Y, Chen C, Tian R, Xiao Q, Peng YY. Tyrosine can protect against oxidative stress through ferryl hemoglobin reduction. Toxicol In Vitro 2014; 28:847-55. [PMID: 24698734 DOI: 10.1016/j.tiv.2014.03.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 02/19/2014] [Accepted: 03/21/2014] [Indexed: 10/25/2022]
Abstract
The toxic mechanism of hemoglobin (Hb) under oxidative stress is linked to the formations of highly cytotoxic ferryl species and subsequently heme-to-protein cross-linked derivative of Hb (Hb-X). In this study, we have examined the effects of free tyrosine and its analogues (3-chlorotyrosine, phenylalanine) on the stability of ferryl hemoglobin and the formation of Hb-X. The results showed that free tyrosine (not phenylalanine, 10-500 μM) was an efficient reducing agent of ferryl species and also effective at preventing the formation of cytotoxic Hb-X. Meanwhile, the dimeric tyrosine was formed as the oxidation product of tyrosine during Hb redox reaction. Compared with free tyrosine, 3-chlorotyrosine, an oxidation product of tyrosine and a proposed biomarker for hypochlorous acid (HOCl) in vivo, exhibited stronger antioxidant properties in Hb-induced oxidative stress, which was consistent with its more efficient ability in the reduction of ferryl species. These results showed that the presence of tyrosine and its derivative in vivo and vitro could ameliorate oxidative damage through ferryl heme reduction. The antioxidant ability, therefore, may provide new insights into the nutritional and physiological significance of free tyrosine with redox active heme proteins-related oxidative stress.
Collapse
Affiliation(s)
- Naihao Lu
- Jiangxi Key Laboratory of Functional Organic Molecules, Jiangxi Science and Technology Normal University, Nanchang 330013, China; Key Laboratory of Functional Small Organic Molecule, Ministry of Education and College of Life Science, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi 330022, China.
| | - Yingjie He
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education and College of Life Science, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi 330022, China; Key Laboratory of Green Chemistry, Jiangxi Province and College of Chemistry and Chemical Engineering, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi 330022, China
| | - Chao Chen
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education and College of Life Science, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi 330022, China
| | - Rong Tian
- Key Laboratory of Green Chemistry, Jiangxi Province and College of Chemistry and Chemical Engineering, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi 330022, China
| | - Qiang Xiao
- Jiangxi Key Laboratory of Functional Organic Molecules, Jiangxi Science and Technology Normal University, Nanchang 330013, China.
| | - Yi-Yuan Peng
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education and College of Life Science, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi 330022, China; Key Laboratory of Green Chemistry, Jiangxi Province and College of Chemistry and Chemical Engineering, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi 330022, China
| |
Collapse
|
39
|
Heme-induced neutrophil extracellular traps contribute to the pathogenesis of sickle cell disease. Blood 2014; 123:3818-27. [PMID: 24620350 DOI: 10.1182/blood-2013-10-529982] [Citation(s) in RCA: 264] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Sickle cell disease (SCD) is characterized by recurring episodes of vascular occlusion in which neutrophil activation plays a major role. The disease is associated with chronic hemolysis with elevated cell-free hemoglobin and heme. The ensuing depletion of heme scavenger proteins leads to nonspecific heme uptake and heme-catalyzed generation of reactive oxygen species. Here, we have identified a novel role for heme in the induction of neutrophil extracellular trap (NET) formation in SCD. NETs are decondensed chromatin decorated by granular enzymes and are released by activated neutrophils. In humanized SCD mice, we have detected NETs in the lungs and soluble NET components in plasma. The presence of NETs was associated with hypothermia and death of these mice, which could be prevented and delayed, respectively, by dismantling NETs with DNase I treatment. We have identified heme as the plasma factor that stimulates neutrophils to release NETs in vitro and in vivo. Increasing or decreasing plasma heme concentrations can induce or prevent, respectively, in vivo NET formation, indicating that heme plays a crucial role in stimulating NET release in SCD. Our results thus suggest that NETs significantly contribute to SCD pathogenesis and can serve as a therapeutic target for treating SCD.
Collapse
|
40
|
Martínez-Sernández V, Mezo M, González-Warleta M, Perteguer MJ, Muiño L, Guitián E, Gárate T, Ubeira FM. The MF6p/FhHDM-1 major antigen secreted by the trematode parasite Fasciola hepatica is a heme-binding protein. J Biol Chem 2013; 289:1441-56. [PMID: 24280214 DOI: 10.1074/jbc.m113.499517] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Blood-feeding parasites have developed biochemical mechanisms to control heme intake and detoxification. Here we show that a major antigen secreted by Fasciola hepatica, previously reported as MF6p, of unknown function (gb|CCA61804.1), and as FhHDM-1, considered to be a helminth defense molecule belonging to the family of cathelicidin-like proteins (gb|ADZ24001.1), is in fact a heme-binding protein. The heme-binding nature of the MF6p/FhHDM-1 protein was revealed in two independent experiments: (i) immunopurification of the secreted protein·heme complexes with mAb MF6 and subsequent analysis by C8 reversed-phase HPLC and MS/MS spectrometry and (ii) analysis of the binding ability of the synthetic protein to hemin in vitro. By immunohistochemistry analysis, we have observed that MF6p/FhHDM-1 is produced by parenchymal cells and transported to other tissues (e.g. vitellaria and testis). Interestingly, MF6p/FhHDM-1 is absent both in the intestinal cells and in the lumen of cecum, but it can be released through the tegumental surface to the external medium, where it binds to free heme molecules regurgitated by the parasite after hemoglobin digestion. Proteins that are close analogs of the Fasciola MF6p/FhHDM-1 are present in other trematodes, including Clonorchis, Opistorchis, Paragonimus, Schistosoma, and Dicrocoelium. Using UV-visible spectroscopy and immunoprecipitation techniques, we observed that synthetic MF6p/FhHDM-1 binds to hemin with 1:1 stoichiometry and an apparent Kd of 1.14 × 10(-6) M(-1). We also demonstrated that formation of synthetic MF6p/FhHDM-1·hemin complexes inhibited hemin degradation by hydrogen peroxide and hemin peroxidase-like activity in vitro. Our results suggest that MF6p/FhHDM-1 may be involved in heme homeostasis in trematodes.
Collapse
Affiliation(s)
- Victoria Martínez-Sernández
- From the Laboratorio de Parasitología, Facultad de Farmacia, Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Huang Y, Shuai Y, Li H, Gao Z. Tyrosine residues play an important role in heme detoxification by serum albumin. Biochim Biophys Acta Gen Subj 2013; 1840:970-6. [PMID: 24252277 DOI: 10.1016/j.bbagen.2013.11.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 10/16/2013] [Accepted: 11/09/2013] [Indexed: 11/18/2022]
Abstract
BACKGROUND Serum albumin binds avidly to heme to form heme-serum albumin complex, also called methemalbumin, and this binding is thought to protect against the potentially toxic effects of heme. However, the mechanism of detoxification has not been fully elucidated. METHODS SDS-PAGE and Western blot were used to determine the efficiency of methemalbumin on catalyzing protein carbonylation and nitration. HPLC was used to test the formation of heme to protein cross-linked methemalbumin. RESULTS The peroxidase activity of heme increased upon human serum albumin (HSA) binding. Methemalbumin showed higher efficiency in catalyzing tyrosine oxidation than free heme in the presence of H2O2. Methemalbumin catalyzed self-nitration and significantly promoted the nitration of tyrosine in coexistent protein, but decreased the carbonylation of coexistent protein compared with heme. The heme to protein cross-linked form of methemalbumin suggested that HSA trapped the free radical accompanied by the formation of ferryl heme. When tyrosine residues in HSA were modified by iodination, HSA lost of protection effect on protein carbonylation. The low concentration of glutathione could effectively inhibit tyrosine nitration, but had no effect on protein carbonylation. CONCLUSION HSA protects against the toxic effect of heme by transferring the free radical to tyrosine residues in HSA, therefore protecting surrounding proteins from irreversible oxidation, rather than by direct inhibiting the peroxidase activity. The increased tyrosine radicals can be reduced by endogenic antioxidants such as GSH. GENERAL SIGNIFICANCE This investigation indicated the important role of tyrosine residues in heme detoxification by HSA and suggested a possible novel mechanism.
Collapse
Affiliation(s)
- Yi Huang
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China
| | - Youxia Shuai
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China
| | - Hailing Li
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China
| | - Zhonghong Gao
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China.
| |
Collapse
|
42
|
Kumar A, Ganini D, Deterding LJ, Ehrenshaft M, Chatterjee S, Mason RP. Immuno-spin trapping of heme-induced protein radicals: Implications for heme oxygenase-1 induction and heme degradation. Free Radic Biol Med 2013; 61:265-72. [PMID: 23624303 PMCID: PMC3851609 DOI: 10.1016/j.freeradbiomed.2013.04.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 04/13/2013] [Accepted: 04/16/2013] [Indexed: 01/29/2023]
Abstract
Heme, in the presence of hydrogen peroxide, can act as a peroxidase. Intravascular hemolysis results in a massive release of heme into the plasma in several pathophysiological conditions such as hemolytic anemia, malaria, and sickle cell disease. Heme is known to induce heme oxygenase-1(HO-1) expression, and the extent of induction depends on the ratio of albumin to heme in plasma. HO-1 degrades heme and ultimately generates the antioxidant bilirubin. Heme also causes oxidative stress in cells, but whether it causes protein-radical formation has not yet been studied. In the literature, two purposes for the degradation of heme by HO-1 are discussed. One is the production of the antioxidant bilirubin and the other is the prevention of heme-dependent adverse effects. Here we have investigated heme-induced protein-radical formation, which might have pathophysiological consequences, and have used immuno-spin trapping to establish the formation of heme-induced protein radicals in two systems: human serum albumin (HSA)/H2O2 and human plasma/H2O2.We found that excess heme catalyzed the formation of HSA radicals in the presence of hydrogen peroxide. When heme and hydrogen peroxide were added to human plasma, heme was found to oxidize proteins, primarily and predominantly HSA; however, when HSA-depleted plasma was used, heme triggered the oxidation of several other proteins, including transferrin. Thus, HSA in plasma protected other proteins from heme/H2O2-induced oxidation. The antioxidants ascorbate and uric acid significantly attenuated protein-radical formation induced by heme/H2O2; however, bilirubin did not confer significant protection. Based on these findings, we conclude that heme is degraded by HO-1 because it is a catalyst of protein-radical formation and not merely to produce the relatively inefficient antioxidant bilirubin.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Free Radical Metabolism Group, Laboratories of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA.
| | - Douglas Ganini
- Free Radical Metabolism Group, Laboratories of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Leesa J Deterding
- Laboratory of Structural Biology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Marilyn Ehrenshaft
- Free Radical Metabolism Group, Laboratories of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Saurabh Chatterjee
- Department of Environmental Health Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Ronald P Mason
- Free Radical Metabolism Group, Laboratories of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA
| |
Collapse
|
43
|
Hahl P, Davis T, Washburn C, Rogers JT, Smith A. Mechanisms of neuroprotection by hemopexin: modeling the control of heme and iron homeostasis in brain neurons in inflammatory states. J Neurochem 2013; 125:89-101. [PMID: 23350672 DOI: 10.1111/jnc.12165] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 01/18/2013] [Accepted: 01/18/2013] [Indexed: 12/21/2022]
Abstract
Hemopexin provides neuroprotection in mouse models of stroke and intracerebral hemorrhage and protects neurons in vitro against heme or reactive oxygen species (ROS) toxicity via heme oxygenase-1 (HO1) activity. To model human brain neurons experiencing hemorrhages and inflammation, we used human neuroblastoma cells, heme-hemopexin complexes, and physiologically relevant ROS, for example, H(2)O(2) and HOCl, to provide novel insights into the underlying mechanism whereby hemopexin safely maintains heme and iron homeostasis. Human amyloid precursor protein (hAPP), needed for iron export from neurons, is induced ~twofold after heme-hemopexin endocytosis by iron from heme catabolism via the iron-regulatory element of hAPP mRNA. Heme-hemopexin is relatively resistant to damage by ROS and retains its ability to induce the cytoprotective HO1 after exposure to tert-butylhydroperoxide, although induction is impaired, but not eliminated, by exposure to high concentrations of H(2)O(2) in vitro. Apo-hemopexin, which predominates in non-hemolytic states, resists damage by H(2)O(2) and HOCl, except for the highest concentrations likely in vivo. Heme-albumin and albumin are preferential targets for ROS; thus, albumin protects hemopexin in biological fluids like CSF and plasma where it is abundant. These observations provide strong evidence that hemopexin will be neuroprotective after traumatic brain injury, with heme release in the CNS, and during the ensuing inflammation. Hemopexin sequesters heme, thus preventing unregulated heme uptake that leads to toxicity; it safely delivers heme to neuronal cells; and it activates the induction of proteins including HO1 and hAPP that keep heme and iron at safe levels in neurons.
Collapse
Affiliation(s)
- Peter Hahl
- School of Biological Sciences, University of Missouri - Kansas City, Kansas City, MO, USA
| | | | | | | | | |
Collapse
|
44
|
Karnaukhova E, Krupnikova SS, Rajabi M, Alayash AI. Heme binding to human alpha-1 proteinase inhibitor. Biochim Biophys Acta Gen Subj 2012; 1820:2020-9. [PMID: 23000493 DOI: 10.1016/j.bbagen.2012.09.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 08/20/2012] [Accepted: 09/14/2012] [Indexed: 11/27/2022]
Abstract
BACKGROUND Heme is a unique prosthetic group of various hemoproteins that perform diverse biological functions; however, in its free form heme is intrinsically toxic in vivo. Due to its potential toxicity, heme binding to plasma proteins is an important safety issue in regard to protein therapeutics derived from human blood. While heme binding by hemopexin, albumin and α(1)-microglobulin has been extensively studied, the role of other plasma proteins remains largely unknown. METHODS We examined two acute-phase plasma proteins, haptoglobin (Hp) and alpha-1 proteinase inhibitor (α(1)-PI) for possible interactions with heme and bilirubin (BR), the final product of heme degradation, using various techniques: UV/Vis spectroscopy, fluorescence, circular dichroism (CD), and surface plasmon resonance (SPR). RESULTS According to our data, Hp exhibits a very weak association with both heme and BR; α(1)-PI's affinity to BR is also very low. However, α(1)-PI's affinity to heme (K(D) 2.0×10(-8)M) is of the same order of magnitude as that of albumin (1.26×10(-8)M). The data for α(1)-PI binding with protoporphyrin IX (PPIX) suggest that the elimination of the iron atom from the porphyrin structure results in almost 350-fold lower affinity (K(D) 6.93×10(-6)M), thus indicating that iron is essential for the heme coordination with the α(1)-PI. CONCLUSIONS This work demonstrates for the first time that human α(1)-PI is a heme binding protein with an affinity to heme comparable to that of albumin. GENERAL SIGNIFICANCE Our data may have important implications for safety and efficacy of plasma protein therapeutics.
Collapse
Affiliation(s)
- Elena Karnaukhova
- Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
45
|
Olsson MG, Allhorn M, Bülow L, Hansson SR, Ley D, Olsson ML, Schmidtchen A, Akerström B. Pathological conditions involving extracellular hemoglobin: molecular mechanisms, clinical significance, and novel therapeutic opportunities for α(1)-microglobulin. Antioxid Redox Signal 2012; 17:813-46. [PMID: 22324321 DOI: 10.1089/ars.2011.4282] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hemoglobin (Hb) is the major oxygen (O(2))-carrying system of the blood but has many potentially dangerous side effects due to oxidation and reduction reactions of the heme-bound iron and O(2). Extracellular Hb, resulting from hemolysis or exogenous infusion, is shown to be an important pathogenic factor in a growing number of diseases. This review briefly outlines the oxidative/reductive toxic reactions of Hb and its metabolites. It also describes physiological protection mechanisms that have evolved against extracellular Hb, with a focus on the most recently discovered: the heme- and radical-binding protein α(1)-microglobulin (A1M). This protein is found in all vertebrates, including man, and operates by rapidly clearing cytosols and extravascular fluids of heme groups and free radicals released from Hb. Five groups of pathological conditions with high concentrations of extracellular Hb are described: hemolytic anemias and transfusion reactions, the pregnancy complication pre-eclampsia, cerebral intraventricular hemorrhage of premature infants, chronic inflammatory leg ulcers, and infusion of Hb-based O(2) carriers as blood substitutes. Finally, possible treatments of these conditions are discussed, giving a special attention to the described protective effects of A1M.
Collapse
|
46
|
Grunwald EW, Richards MP. Effects of hemopexin on hemin and hemoglobin-mediated lipid oxidation in washed fish muscle. Lebensm Wiss Technol 2012. [DOI: 10.1016/j.lwt.2011.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
47
|
Lu N, Chen W, Zhu J, Peng YY. Enhancement of nitrite on heme-induced oxidative reactions: A potential toxicological implication. Toxicol In Vitro 2012; 26:81-5. [DOI: 10.1016/j.tiv.2011.10.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 07/17/2011] [Accepted: 10/20/2011] [Indexed: 10/15/2022]
|
48
|
Human serum albumin: from bench to bedside. Mol Aspects Med 2011; 33:209-90. [PMID: 22230555 DOI: 10.1016/j.mam.2011.12.002] [Citation(s) in RCA: 1319] [Impact Index Per Article: 94.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 12/21/2011] [Indexed: 02/07/2023]
Abstract
Human serum albumin (HSA), the most abundant protein in plasma, is a monomeric multi-domain macromolecule, representing the main determinant of plasma oncotic pressure and the main modulator of fluid distribution between body compartments. HSA displays an extraordinary ligand binding capacity, providing a depot and carrier for many endogenous and exogenous compounds. Indeed, HSA represents the main carrier for fatty acids, affects pharmacokinetics of many drugs, provides the metabolic modification of some ligands, renders potential toxins harmless, accounts for most of the anti-oxidant capacity of human plasma, and displays (pseudo-)enzymatic properties. HSA is a valuable biomarker of many diseases, including cancer, rheumatoid arthritis, ischemia, post-menopausal obesity, severe acute graft-versus-host disease, and diseases that need monitoring of the glycemic control. Moreover, HSA is widely used clinically to treat several diseases, including hypovolemia, shock, burns, surgical blood loss, trauma, hemorrhage, cardiopulmonary bypass, acute respiratory distress syndrome, hemodialysis, acute liver failure, chronic liver disease, nutrition support, resuscitation, and hypoalbuminemia. Recently, biotechnological applications of HSA, including implantable biomaterials, surgical adhesives and sealants, biochromatography, ligand trapping, and fusion proteins, have been reported. Here, genetic, biochemical, biomedical, and biotechnological aspects of HSA are reviewed.
Collapse
|
49
|
Expression of virulence factors by Staphylococcus aureus grown in serum. Appl Environ Microbiol 2011; 77:8097-105. [PMID: 21926198 DOI: 10.1128/aem.05316-11] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Staphylococcus aureus produces many virulence factors, including toxins, immune-modulatory factors, and exoenzymes. Previous studies involving the analysis of virulence expression were mainly performed by in vitro experiments using bacterial medium. However, when S. aureus infects a host, the bacterial growth conditions are quite different from those in a medium, which may be related to the different expression of virulence factors in the host. In this study, we investigated the expression of virulence factors in S. aureus grown in calf serum. The expression of many virulence factors, including hemolysins, enterotoxins, proteases, and iron acquisition factors, was significantly increased compared with that in bacterial medium. In addition, the expression of RNA III, a global regulon for virulence expression, was significantly increased. This effect was partially restored by the addition of 300 μM FeCl₃ into serum, suggesting that iron depletion is associated with the increased expression of virulence factors in serum. In chemically defined medium without iron, a similar effect was observed. In a mutant with agr inactivated grown in serum, the expression of RNA III, psm, and sec4 was not increased, while other factors were still induced in the mutant, suggesting that another regulatory factor(s) is involved. In addition, we found that serum albumin is a major factor for the capture of free iron to prevent the supply of iron to bacteria grown in serum. These results indicate that S. aureus expresses virulence factors in adaptation to the host environment.
Collapse
|
50
|
Westberg JA, Jiang J, Andersson LC. Stanniocalcin 1 binds hemin through a partially conserved heme regulatory motif. Biochem Biophys Res Commun 2011; 409:266-9. [DOI: 10.1016/j.bbrc.2011.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 05/01/2011] [Indexed: 10/18/2022]
|