1
|
Kasper R, Rodriguez-Alfonso A, Ständker L, Wiese S, Schneider EM. Major endothelial damage markers identified from hemadsorption filters derived from treated patients with septic shock - endoplasmic reticulum stress and bikunin may play a role. Front Immunol 2024; 15:1359097. [PMID: 38698864 PMCID: PMC11063272 DOI: 10.3389/fimmu.2024.1359097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
Introduction In septic patients the damage of the endothelial barrier is decisive leading to circulatory septic shock with disseminated vascular coagulation, edema and multiorgan failure. Hemadsorption therapy leads to rapid resolution of clinical symptoms. We propose that the isolation of proteins adsorbed to hemadsorption devices contributes to the identification of mediators responsible for endothelial barrier dysfunction. Material and methods Plasma materials enriched to hemadsorption filters (CytoSorb®) after therapy of patients in septic shock were fractionated and functionally characterized for their effect on cell integrity, viability, proliferation and ROS formation by human endothelial cells. Fractions were further studied for their contents of oxidized nucleic acids as well as peptides and proteins by mass spectrometry. Results Individual fractions exhibited a strong effect on endothelial cell viability, the endothelial layer morphology, and ROS formation. Fractions with high amounts of DNA and oxidized DNA correlated with ROS formation in the target endothelium. In addition, defined proteins such as defensins (HNP-1), SAA1, CXCL7, and the peptide bikunin were linked to the strongest additive effects in endothelial damage. Conclusion Our results indicate that hemadsorption is efficient to transiently remove strong endothelial damage mediators from the blood of patients with septic shock, which explains a rapid clinical improvement of inflammation and endothelial function. The current work indicates that a combination of stressors leads to the most detrimental effects. Oxidized ssDNA, likely derived from mitochondria, SAA1, the chemokine CXCL7 and the human neutrophil peptide alpha-defensin 1 (HNP-1) were unique for their significant negative effect on endothelial cell viability. However, the strongest damage effect occurred, when, bikunin - cleaved off from alpha-1-microglobulin was present in high relative amounts (>65%) of protein contents in the most active fraction. Thus, a relevant combination of stressors appears to be removed by hemadsorption therapy which results in fulminant and rapid, though only transient, clinical restitution.
Collapse
Affiliation(s)
- Robin Kasper
- Clinic of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm, Germany
| | - Armando Rodriguez-Alfonso
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, Germany
- Core Unit Mass Spectrometry and Proteomics (CUMP), Ulm University, Ulm, Germany
| | - Ludger Ständker
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics (CUMP), Ulm University, Ulm, Germany
| | - E. Marion Schneider
- Clinic of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
2
|
Gáll T, Nagy P, Garai D, Potor L, Balla GJ, Balla G, Balla J. Overview on hydrogen sulfide-mediated suppression of vascular calcification and hemoglobin/heme-mediated vascular damage in atherosclerosis. Redox Biol 2022; 57:102504. [PMID: 36240620 PMCID: PMC9576974 DOI: 10.1016/j.redox.2022.102504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 12/04/2022] Open
Abstract
Vulnerable atherosclerotic plaques with hemorrhage considerably contribute to cardiovascular morbidity and mortality. Calcification is the main characteristic of advanced atherosclerotic lesions and calcified aortic valve disease (CAVD). Lyses of red blood cells and hemoglobin (Hb) release occur in human hemorrhagic complicated lesions. During the interaction of cell-free Hb with plaque constituents, Hb is oxidized to ferric and ferryl states accompanied by oxidative changes of the globin moieties and heme release. Accumulation of both ferryl-Hb and metHb has been observed in atherosclerotic plaques. The oxidation hotspots in the globin chain are the cysteine and tyrosine amino acids associated with the generation of Hb dimers, tetramers and polymers. Moreover, fragmentation of Hb occurs leading to the formation of globin-derived peptides. A series of these pro-atherogenic cellular responses can be suppressed by hydrogen sulfide (H2S). Since H2S has been explored to exhibit a wide range of physiologic functions to maintain vascular homeostasis, it is not surprising that H2S may play beneficial effects in the progression of atherosclerosis. In the present review, we summarize the findings about the effects of H2S on atherosclerosis and CAVD with a special emphasis on the oxidation of Hb/heme in atherosclerotic plaque development and vascular calcification.
Collapse
Affiliation(s)
- Tamás Gáll
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Hungary; ELKH-UD Vascular Pathophysiology Research Group, 11003, University of Debrecen, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Péter Nagy
- Department of Molecular Immunology and Toxicology, National Institute of Oncology, Budapest, Hungary; Institute of Oncochemistry, University of Debrecen, Hungary
| | - Dorottya Garai
- Department of Molecular Immunology and Toxicology, National Institute of Oncology, Budapest, Hungary
| | - László Potor
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Hungary; ELKH-UD Vascular Pathophysiology Research Group, 11003, University of Debrecen, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | | | - György Balla
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Hungary; ELKH-UD Vascular Pathophysiology Research Group, 11003, University of Debrecen, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - József Balla
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Hungary; ELKH-UD Vascular Pathophysiology Research Group, 11003, University of Debrecen, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
3
|
Taiwo GA, Idowu M, Denvir J, Cervantes AP, Ogunade IM. Identification of Key Pathways Associated With Residual Feed Intake of Beef Cattle Based on Whole Blood Transcriptome Data Analyzed Using Gene Set Enrichment Analysis. Front Vet Sci 2022; 9:848027. [PMID: 35518641 PMCID: PMC9062580 DOI: 10.3389/fvets.2022.848027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/08/2022] [Indexed: 11/19/2022] Open
Abstract
We applied whole blood transcriptome analysis and gene set enrichment analysis to identify pathways associated with divergent selection for low or high RFI in beef cattle. A group of 56 crossbred beef steers (average BW = 261 ± 18.5 kg) were adapted to a high-forage total mixed ration in a confinement dry lot equipped with GrowSafe intake nodes for period of 49 d to determine their residual feed intake (RFI). After RFI determination, whole blood samples were collected from beef steers with the lowest RFI (most efficient; low-RFI; n = 8) and highest RFI (least efficient; high-RFI; n = 8). Prior to RNA extraction, whole blood samples collected were composited for each steer. Sequencing was performed on an Illumina NextSeq2000 equipped with a P3 flow. Gene set enrichment analysis (GSEA) was used to analyze differentially expressed gene sets and pathways between the two groups of steers. Results of GSEA revealed pathways associated with metabolism of proteins, cellular responses to external stimuli, stress, and heat stress were differentially inhibited (false discovery rate (FDR) < 0.05) in high-RFI compared to low-RFI beef cattle, while pathways associated with binding and uptake of ligands by scavenger receptors, scavenging of heme from plasma, and erythrocytes release/take up oxygen were differentially enriched (FDR < 0.05) in high-RFI, relative to low-RFI beef cattle. Taken together, our results revealed that beef steers divergently selected for low or high RFI revealed differential expressions of genes related to protein metabolism and stress responsiveness.
Collapse
Affiliation(s)
- Godstime A Taiwo
- Division of Animal and Nutritional Science, West Virginia University, Morgantown, WV, United States
| | - Modoluwamu Idowu
- Division of Animal and Nutritional Science, West Virginia University, Morgantown, WV, United States
| | - James Denvir
- Department of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Andres Pech Cervantes
- Agricultural Research Station, Fort Valley State University, Fort Valley, GA, United States
| | - Ibukun M Ogunade
- Division of Animal and Nutritional Science, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
4
|
Kristiansson A, Örbom A, Vilhelmsson Timmermand O, Ahlstedt J, Strand SE, Åkerström B. Kidney Protection with the Radical Scavenger α 1-Microglobulin (A1M) during Peptide Receptor Radionuclide and Radioligand Therapy. Antioxidants (Basel) 2021; 10:antiox10081271. [PMID: 34439519 PMCID: PMC8389303 DOI: 10.3390/antiox10081271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/05/2021] [Accepted: 08/07/2021] [Indexed: 02/07/2023] Open
Abstract
α1-Microglobulin (A1M) is an antioxidant found in all vertebrates, including humans. It has enzymatic reductase activity and can scavenge radicals and bind free heme groups. Infused recombinant A1M accumulates in the kidneys and has therefore been successful in protecting kidney injuries in different animal models. In this review, we focus on A1M as a radioprotector of the kidneys during peptide receptor radionuclide/radioligand therapy (PRRT/RLT). Patients with, e.g., neuroendocrine tumors or castration resistant prostate cancer can be treated by administration of radiolabeled small molecules which target and therefore enable the irradiation and killing of cancer cells through specific receptor interaction. The treatment is not curative, and kidney toxicity has been reported as a side effect since the small, radiolabeled substances are retained and excreted through the kidneys. In recent studies, A1M was shown to have radioprotective effects on cell cultures as well as having a similar biodistribution as the somatostatin analogue peptide 177Lu-DOTATATE after intravenous infusion in mice. Therefore, several animal studies were conducted to investigate the in vivo radioprotective potential of A1M towards kidneys. The results of these studies demonstrated that A1M co-infusion yielded protection against kidney toxicity and improved overall survival in mouse models. Moreover, two different mouse studies reported that A1M did not interfere with tumor treatment itself. Here, we give an overview of radionuclide therapy, the A1M physiology and the results from the radioprotector studies of the protein.
Collapse
Affiliation(s)
- Amanda Kristiansson
- Department of Clinical Sciences Lund, Oncology, Lund University, 221 00 Lund, Sweden; (A.Ö.); (O.V.T.); (S.-E.S.)
- Correspondence:
| | - Anders Örbom
- Department of Clinical Sciences Lund, Oncology, Lund University, 221 00 Lund, Sweden; (A.Ö.); (O.V.T.); (S.-E.S.)
| | - Oskar Vilhelmsson Timmermand
- Department of Clinical Sciences Lund, Oncology, Lund University, 221 00 Lund, Sweden; (A.Ö.); (O.V.T.); (S.-E.S.)
| | - Jonas Ahlstedt
- Department of Clinical Sciences Lund, CIPA, Lund University, 221 84 Lund, Sweden;
| | - Sven-Erik Strand
- Department of Clinical Sciences Lund, Oncology, Lund University, 221 00 Lund, Sweden; (A.Ö.); (O.V.T.); (S.-E.S.)
- Department of Clinical Sciences Lund, Medical Radiation Physics, Lund University, 221 00 Lund, Sweden
| | - Bo Åkerström
- Department of Clinical Sciences Lund, Section for Infection Medicine, Lund University, 221 84 Lund, Sweden;
| |
Collapse
|
5
|
Ferryl Hemoglobin and Heme Induce A 1-Microglobulin in Hemorrhaged Atherosclerotic Lesions with Inhibitory Function against Hemoglobin and Lipid Oxidation. Int J Mol Sci 2021; 22:ijms22136668. [PMID: 34206377 PMCID: PMC8268598 DOI: 10.3390/ijms22136668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022] Open
Abstract
Infiltration of red blood cells into atheromatous plaques and oxidation of hemoglobin (Hb) and lipoproteins are implicated in the pathogenesis of atherosclerosis. α1-microglobulin (A1M) is a radical-scavenging and heme-binding protein. In this work, we examined the origin and role of A1M in human atherosclerotic lesions. Using immunohistochemistry, we observed a significant A1M immunoreactivity in atheromas and hemorrhaged plaques of carotid arteries in smooth muscle cells (SMCs) and macrophages. The most prominent expression was detected in macrophages of organized hemorrhage. To reveal a possible inducer of A1M expression in ruptured lesions, we exposed aortic endothelial cells (ECs), SMCs and macrophages to heme, Oxy- and FerrylHb. Both heme and FerrylHb, but not OxyHb, upregulated A1M mRNA expression in all cell types. Importantly, only FerrylHb induced A1M protein secretion in aortic ECs, SMCs and macrophages. To assess the possible function of A1M in ruptured lesions, we analyzed Hb oxidation and heme-catalyzed lipid peroxidation in the presence of A1M. We showed that recombinant A1M markedly inhibited Hb oxidation and heme-driven oxidative modification of low-density lipoproteins as well plaque lipids derived from atheromas. These results demonstrate the presence of A1M in atherosclerotic plaques and suggest its induction by heme and FerrylHb in the resident cells.
Collapse
|
6
|
Bergwik J, Kristiansson A, Allhorn M, Gram M, Åkerström B. Structure, Functions, and Physiological Roles of the Lipocalin α 1-Microglobulin (A1M). Front Physiol 2021; 12:645650. [PMID: 33746781 PMCID: PMC7965949 DOI: 10.3389/fphys.2021.645650] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/12/2021] [Indexed: 12/30/2022] Open
Abstract
α1-microglobulin (A1M) is found in all vertebrates including humans. A1M was, together with retinol-binding protein and β-lactoglobulin, one of the three original lipocalins when the family first was proposed in 1985. A1M is described as an antioxidant and tissue cleaning protein with reductase, heme- and radical-binding activities. These biochemical properties are driven by a strongly electronegative surface-exposed thiol group, C34, on loop 1 of the open end of the lipocalin barrel. A1M has been shown to have protective effects in vitro and in vivo in cell-, organ-, and animal models of oxidative stress-related medical conditions. The gene coding for A1M is unique among lipocalins since it is flanked downstream by four exons coding for another non-lipocalin protein, bikunin, and is consequently named α1-microglobulin-bikunin precursor gene (AMBP). The precursor is cleaved in the Golgi, and A1M and bikunin are secreted from the cell separately. Recent publications have suggested novel physiological roles of A1M in regulation of endoplasmic reticulum activities and erythrocyte homeostasis. This review summarizes the present knowledge of the structure and functions of the lipocalin A1M and presents a current model of its biological role(s).
Collapse
Affiliation(s)
- Jesper Bergwik
- Department of Clinical Sciences, Section for Infection Medicine, Lund University, Lund, Sweden
| | - Amanda Kristiansson
- Department of Clinical Sciences, Section for Infection Medicine, Lund University, Lund, Sweden.,Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Maria Allhorn
- Department of Clinical Sciences, Section for Infection Medicine, Lund University, Lund, Sweden
| | - Magnus Gram
- Department of Clinical Sciences, Pediatrics, Lund University, Lund, Sweden
| | - Bo Åkerström
- Department of Clinical Sciences, Section for Infection Medicine, Lund University, Lund, Sweden
| |
Collapse
|
7
|
Bergwik J, Kristiansson A, Welinder C, Göransson O, Hansson SR, Gram M, Erlandsson L, Åkerström B. Knockout of the radical scavenger α 1-microglobulin in mice results in defective bikunin synthesis, endoplasmic reticulum stress and increased body weight. Free Radic Biol Med 2021; 162:160-170. [PMID: 32092411 DOI: 10.1016/j.freeradbiomed.2020.02.019] [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: 11/19/2019] [Revised: 02/14/2020] [Accepted: 02/19/2020] [Indexed: 01/22/2023]
Abstract
α1-microglobulin (A1M) is a ubiquitous protein with reductase and radical- and heme-binding properties. The protein is mainly expressed in the liver and encoded by the α1-microglobulin-bikunin precursor (AMBP) gene together with the plasma proteinase inhibitor bikunin. The AMBP polypeptide is translated, glycosylated and the C-terminal bikunin part linked via a chondroitin sulfate glycosaminoglycan chain to one or two heavy chains in the endoplasmic reticulum (ER) and Golgi compartments. After proteolytic cleavage, the A1M protein and complexed bikunin parts are secreted separately. The complete physiological role of A1M, and the reason for the co-synthesis with bikunin, are both still unknown. The aim of this work was to develop an A1M knockout (A1M-KO) mouse model lacking expression of A1M, but with a preserved bikunin expression, and to study the phenotypic traits in these mice, with a focus on hepatic endoplasmic reticulum (ER) function. The bikunin expression was increased in the A1M-KO mouse livers, while the bikunin levels in plasma were decreased, indicating a defective biosynthesis of bikunin. The A1M-KO livers also showed an increased expression of transducers of the unfolded protein response (UPR), indicating an increased ER-stress in the livers. At twelve months of age, the A1M-KO mice also displayed an increased body weight, and an increased liver weight and lipid accumulation. Moreover, the KO mice showed an increased expression of endogenous antioxidants in the liver, but not in the kidneys. Together, these results suggest a physiological role of A1M as a regulator of the intracellular redox environment and more specifically the ER folding and posttranslational modification processes, particularly in the liver.
Collapse
Affiliation(s)
- Jesper Bergwik
- Section for Infection Medicine, Department of Clinical Sciences in Lund, Lund University, Lund, Sweden
| | - Amanda Kristiansson
- Section for Infection Medicine, Department of Clinical Sciences in Lund, Lund University, Lund, Sweden
| | - Charlotte Welinder
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Olga Göransson
- Protein Phosphorylation Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Stefan R Hansson
- Section for Obstetrics and Gynecology, Department of Clinical Sciences in Lund, Lund University, Lund, Sweden
| | - Magnus Gram
- Section for Infection Medicine, Department of Clinical Sciences in Lund, Lund University, Lund, Sweden
| | - Lena Erlandsson
- Section for Obstetrics and Gynecology, Department of Clinical Sciences in Lund, Lund University, Lund, Sweden
| | - Bo Åkerström
- Section for Infection Medicine, Department of Clinical Sciences in Lund, Lund University, Lund, Sweden.
| |
Collapse
|
8
|
Pregnant alpha-1-microglobulin (A1M) knockout mice exhibit features of kidney and placental damage, hemodynamic changes and intrauterine growth restriction. Sci Rep 2020; 10:20625. [PMID: 33244052 PMCID: PMC7691512 DOI: 10.1038/s41598-020-77561-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 11/11/2020] [Indexed: 11/09/2022] Open
Abstract
Alpha-1-microglobulin (A1M) is an antioxidant previously shown to be elevated in maternal blood during pregnancies complicated by preeclampsia and suggested to be important in the endogenous defense against oxidative stress. A knockout mouse model of A1M (A1Mko) was used in the present study to assess the importance of A1M during pregnancy in relation to the kidney, heart and placenta function. Systolic blood pressure (SBP) and heart rate (HR) were determined before and throughout gestation. The morphology of the organs was assessed by both light and electron microscopy. Gene expression profiles relating to vascular tone and oxidative stress were analyzed using RT-qPCR with validation of selected gene expression relating to vascular tone and oxidative stress response. Pregnant age-matched wild type mice were used as controls. In the A1Mko mice there was a significantly higher SBP before pregnancy that during pregnancy was significantly reduced compared to the control. In addition, the HR was higher both before and during pregnancy compared to the controls. Renal morphological abnormalities were more frequent in the A1Mko mice, and the gene expression profiles in the kidney and the heart showed downregulation of transcripts associated with vasodilation. Simultaneously, an upregulation of vasoconstrictors, blood pressure regulators, and genes for osmotic stress response, ion transport and reactive oxygen species (ROS) metabolism occurred. Fetal weight was lower in the A1Mko mice at E17.5. The vessels in the labyrinth zone of the placentas and the endoplasmic reticulum in the spongiotrophoblasts were collapsed. The gene profiles in the placenta showed downregulation of antioxidants, ROS metabolism and oxidative stress response genes. In conclusion, intact A1M expression is necessary for the maintenance of normal kidney, heart as well as placental structure and function for a normal pregnancy adaptation.
Collapse
|
9
|
Kristiansson A, Gram M, Flygare J, Hansson SR, Åkerström B, Storry JR. The Role of α 1-Microglobulin (A1M) in Erythropoiesis and Erythrocyte Homeostasis-Therapeutic Opportunities in Hemolytic Conditions. Int J Mol Sci 2020; 21:ijms21197234. [PMID: 33008134 PMCID: PMC7582998 DOI: 10.3390/ijms21197234] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 02/06/2023] Open
Abstract
α1-microglobulin (A1M) is a small protein present in vertebrates including humans. It has several physiologically relevant properties, including binding of heme and radicals as well as enzymatic reduction, that are used in the protection of cells and tissue. Research has revealed that A1M can ameliorate heme and ROS-induced injuries in cell cultures, organs, explants and animal models. Recently, it was shown that A1M could reduce hemolysis in vitro, observed with several different types of insults and sources of RBCs. In addition, in a recently published study, it was observed that mice lacking A1M (A1M-KO) developed a macrocytic anemia phenotype. Altogether, this suggests that A1M may have a role in RBC development, stability and turnover. This opens up the possibility of utilizing A1M for therapeutic purposes in pathological conditions involving erythropoietic and hemolytic abnormalities. Here, we provide an overview of A1M and its potential therapeutic effect in the context of the following erythropoietic and hemolytic conditions: Diamond-Blackfan anemia (DBA), 5q-minus myelodysplastic syndrome (5q-MDS), blood transfusions (including storage), intraventricular hemorrhage (IVH), preeclampsia (PE) and atherosclerosis.
Collapse
Affiliation(s)
- Amanda Kristiansson
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden;
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, 221 84 Lund, Sweden;
- Correspondence:
| | - Magnus Gram
- Department of Clinical Sciences Lund, Pediatrics, Lund University, 221 84 Lund, Sweden;
| | - Johan Flygare
- Department of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, 221 84 Lund, Sweden;
| | - Stefan R. Hansson
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences Lund, Lund University, 221 84 Lund, Sweden;
| | - Bo Åkerström
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden;
| | - Jill R. Storry
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, 221 84 Lund, Sweden;
- Department of Clinical Immunology and Transfusion Medicine, Office of Medical Services, 221 85 Lund, Sweden
| |
Collapse
|
10
|
α 1-Microglobulin (A1M) Protects Human Proximal Tubule Epithelial Cells from Heme-Induced Damage In Vitro. Int J Mol Sci 2020; 21:ijms21165825. [PMID: 32823731 PMCID: PMC7461577 DOI: 10.3390/ijms21165825] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress is associated with many renal disorders, both acute and chronic, and has also been described to contribute to the disease progression. Therefore, oxidative stress is a potential therapeutic target. The human antioxidant α1-microglobulin (A1M) is a plasma and tissue protein with heme-binding, radical-scavenging and reductase activities. A1M can be internalized by cells, localized to the mitochondria and protect mitochondrial function. Due to its small size, A1M is filtered from the blood into the glomeruli, and taken up by the renal tubular epithelial cells. A1M has previously been described to reduce renal damage in animal models of preeclampsia, radiotherapy and rhabdomyolysis, and is proposed as a pharmacological agent for the treatment of kidney damage. In this paper, we examined the in vitro protective effects of recombinant human A1M (rA1M) in human proximal tubule epithelial cells. Moreover, rA1M was found to protect against heme-induced cell-death both in primary cells (RPTEC) and in a cell-line (HK-2). Expression of stress-related genes was upregulated in both cell cultures in response to heme exposure, as measured by qPCR and confirmed with in situ hybridization in HK-2 cells, whereas co-treatment with rA1M counteracted the upregulation. Mitochondrial respiration, analyzed with the Seahorse extracellular flux analyzer, was compromised following exposure to heme, but preserved by co-treatment with rA1M. Finally, heme addition to RPTE cells induced an upregulation of the endogenous cellular expression of A1M, via activation of the nuclear factor erythroid 2-related factor 2 (Nrf2)-pathway. Overall, data suggest that A1M/rA1M protects against stress-induced damage to tubule epithelial cells that, at least partly, can be attributed to maintaining mitochondrial function.
Collapse
|
11
|
Bergwik J, Åkerström B. α 1-Microglobulin Binds Illuminated Flavins and Has a Protective Effect Against Sublethal Riboflavin-Induced Damage in Retinal Epithelial Cells. Front Physiol 2020; 11:295. [PMID: 32300309 PMCID: PMC7142231 DOI: 10.3389/fphys.2020.00295] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/16/2020] [Indexed: 12/28/2022] Open
Abstract
Riboflavin (vitamin B2) is an important constituent of the prosthetic groups flavin adenine dinucleotide (FAD) and flavin mononucleotide (FMN), which are utilized as electron-carriers in energy metabolism. Excitation by UV-light leads to the generation of riboflavin radicals and reactive oxygen species (ROS), which can oxidize a wide range of biomolecules. The human protein α1-microglobulin (A1M) is a reductase and a radical scavenger, which can protect cells and matrix against oxidative damage. Here, we provide evidence of a molecular interaction between illuminated riboflavin and A1M, similar to the radical scavenging reactions previously seen between A1M and other organic radicals. Binding between riboflavin and A1M was demonstrated by gel migration shift, UV-absorbance and fluorescence spectrum analysis. The reaction between A1M and UV-light illuminated riboflavin involved covalent modification of A1M and proteolytic release of an N-terminal part of the protein. Furthermore, A1M also inhibited the ROS-induced photoreduction reaction of riboflavin, in a reaction involving the free thiol group in position C34. Finally, the results show a protective effect of A1M, analyzed by gene expression rates of stress genes, against sublethal damage in retinal epithelial cells in culture. Together, our results suggest a new role of A1M as a scavenger of riboflavin radicals and ROS produced during illumination of riboflavin.
Collapse
Affiliation(s)
- Jesper Bergwik
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Bo Åkerström
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
12
|
Murtoniemi K, Kalapotharakos G, Vahlberg T, Räikkonen K, Kajantie E, Hämäläinen E, Åkerström B, Villa PM, Hansson SR, Laivuori H. Longitudinal changes in plasma hemopexin and alpha-1-microglobulin concentrations in women with and without clinical risk factors for pre-eclampsia. PLoS One 2019; 14:e0226520. [PMID: 31841544 PMCID: PMC6913989 DOI: 10.1371/journal.pone.0226520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/26/2019] [Indexed: 02/07/2023] Open
Abstract
Recent studies have shown increased concentration of fetal hemoglobin (HbF) in pre-eclamptic women. Plasma hemopexin (Hpx) and alpha-1-microglobulin (A1M) are hemoglobin scavenger proteins that protect against toxic effects of free heme released in the hemoglobin degradation process. We used an enzyme-linked immunosorbent assay to analyze maternal plasma Hpx and A1M concentrations at 12–14, 18–20 and 26–28 weeks of gestation in three groups: 1) 51 women with a low risk for pre-eclampsia (LRW), 2) 49 women with a high risk for pre-eclampsia (PE) who did not develop PE (HRW) and 3) 42 women with a high risk for PE who developed PE (HRPE). The study had three aims: 1) to investigate whether longitudinal differences exist between study groups, 2) to examine if Hpx and A1M concentrations develop differently in pre-eclamptic women with small for gestational age (SGA) fetuses vs. pre-eclamptic women with appropriate for gestational age fetuses, and 3) to examine if longitudinal Hpx and A1M profiles differ by PE subtype (early-onset vs. late-onset and severe vs. non-severe PE). Repeated measures analysis of variance was used to analyze differences in Hpx and A1M concentrations between the groups. We found that the differences in longitudinal plasma Hpx and A1M concentrations in HRW compared to HRPE and to LRW may be associated with reduced risk of PE regardless of clinical risk factors. In women who developed PE, a high A1M concentration from midgestation to late second trimester was associated with SGA. There were no differences in longitudinal Hpx and A1M concentrations from first to late second trimester in high-risk women who developed early-onset or. late-onset PE or in women who developed severe or. non-severe PE.
Collapse
Affiliation(s)
- Katja Murtoniemi
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, University of Helsinki, Finland
- Department of Obstetrics and Gynaecology, University of Turku and Turku University Hospital, Turku, Finland
- * E-mail:
| | - Grigorios Kalapotharakos
- Skåne University Hospital, Department of Clinical Sciences Lund, Department of Obstetrics and Gynecology, Lund University, Lund, Sweden
| | - Tero Vahlberg
- Department of Clinical Medicine, Biostatistics, University of Turku and Turku University Hospital, Turku, Finland
| | - Katri Räikkonen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eero Kajantie
- PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- National Institute for Health and Welfare, Helsinki, Finland
- Children`s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Esa Hämäläinen
- Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland
| | - Bo Åkerström
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Pia M. Villa
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Stefan R. Hansson
- Skåne University Hospital, Department of Clinical Sciences Lund, Department of Obstetrics and Gynecology, Lund University, Lund, Sweden
| | - Hannele Laivuori
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, University of Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital and Tampere University, Faculty of Medicine and Health Technology, Tampere, Finland
| |
Collapse
|
13
|
Kristiansson A, Ahlstedt J, Holmqvist B, Brinte A, Tran TA, Forssell-Aronsson E, Strand SE, Gram M, Åkerström B. Protection of Kidney Function with Human Antioxidation Protein α 1-Microglobulin in a Mouse 177Lu-DOTATATE Radiation Therapy Model. Antioxid Redox Signal 2019; 30:1746-1759. [PMID: 29943622 PMCID: PMC6477591 DOI: 10.1089/ars.2018.7517] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
AIMS Peptide receptor radionuclide therapy (PRRT) is in clinical use today to treat metastatic neuroendocrine tumors. Infused, radiolabeled, somatostatin analog peptides target tumors that are killed by irradiation damage. The peptides, however, are also retained in kidneys due to glomerular filtration, and the administered doses must be limited to avoid kidney damage. The human radical scavenger and antioxidant, α1-microglobulin (A1M), has previously been shown to protect bystander tissue against irradiation damage and has pharmacokinetic and biodistribution properties similar to somatostatin analogs. In this study, we have investigated if A1M can be used as a renal protective agent in PRRT. RESULTS We describe nephroprotective effects of human recombinant A1M on the short- and long-term renal damage observed following lutetium 177 (177Lu)-DOTATATE (150 MBq) exposure in BALB/c mice. After 1, 4, and 8 days (short term), 177Lu-DOTATATE injections resulted in increased formation of DNA double-strand breaks in the renal cortex, upregulated expression of apoptosis and stress response-related genes, and proteinuria (albumin in urine), all of which were significantly suppressed by coadministration of A1M (7 mg/kg). After 6, 12, and 24 weeks (long term), 177Lu-DOTATATE injections resulted in increased animal death, kidney lesions, glomerular loss, upregulation of stress genes, proteinuria, and plasma markers of reduced kidney function, all of which were suppressed by coadministration of A1M. Innovation and Conclusion: This study demonstrates that A1M effectively inhibits radiation-induced renal damage. The findings suggest that A1M may be used as a radioprotector during clinical PRRT, potentially facilitating improved tumor control and enabling more patients to receive treatment.
Collapse
Affiliation(s)
- Amanda Kristiansson
- 1 Division of Infection Medicine, Department of Clinical Sciences in Lund, Lund University , Lund, Sweden
| | - Jonas Ahlstedt
- 1 Division of Infection Medicine, Department of Clinical Sciences in Lund, Lund University , Lund, Sweden
| | | | | | - Thuy A Tran
- 3 Lund University Bioimaging Center , Lund, Sweden .,4 Department of Clinical Neuroscience, Karolinska Institutet , Stockholm, Sweden
| | - Eva Forssell-Aronsson
- 5 Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Center, University of Gothenburg , Sweden
| | - Sven-Erik Strand
- 6 Medical Radiation Physics, Department of Clinical Sciences in Lund, Lund University , Lund, Sweden
| | - Magnus Gram
- 1 Division of Infection Medicine, Department of Clinical Sciences in Lund, Lund University , Lund, Sweden .,7 Pediatrics, Department of Clinical Sciences in Lund, Skane University Hospital, Lund University , Lund, Sweden
| | - Bo Åkerström
- 1 Division of Infection Medicine, Department of Clinical Sciences in Lund, Lund University , Lund, Sweden
| |
Collapse
|
14
|
Kalapotharakos G, Murtoniemi K, Åkerström B, Hämäläinen E, Kajantie E, Räikkönen K, Villa P, Laivuori H, Hansson SR. Plasma Heme Scavengers Alpha-1-Microglobulin and Hemopexin as Biomarkers in High-Risk Pregnancies. Front Physiol 2019; 10:300. [PMID: 31019465 PMCID: PMC6458234 DOI: 10.3389/fphys.2019.00300] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 03/06/2019] [Indexed: 12/27/2022] Open
Abstract
Women with established preeclampsia (PE) have increased plasma concentration of free fetal hemoglobin. We measured two hemoglobin scavenger system proteins, hemopexin (Hpx) and alpha-1-microglobulin (A1M) in maternal plasma using enzyme-linked immunosorbent assay during the late second trimester of pregnancy in women with high and low risk of developing PE. In total 142 women were included in nested case-control study: 42 women diagnosed with PE and 100 controls (49 randomly selected high-risk and 51 low-risk controls). The concentration of plasma A1M in high-risk controls was higher compared to low-risk controls. Women with severe PE had higher plasma A1M levels compared to women with non-severe PE. In conclusion, the concentration of plasma A1M is increased in the late second trimester in high-risk controls, suggesting activation of endogenous protective system against oxidative stress.
Collapse
Affiliation(s)
- Grigorios Kalapotharakos
- Department of Clinical Sciences Lund, Skåne University Hospital, Lund, Sweden.,Department of Obstetrics and Gynecology, Lund University, Lund, Sweden
| | - Katja Murtoniemi
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Department of Obstetrics and Gynecology, Turku University Hospital and University of Turku, Turku, Finland
| | - Bo Åkerström
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Esa Hämäläinen
- HUSLAB, Helsinki University Hospital, Helsinki, Finland.,Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland
| | - Eero Kajantie
- National Institute for Health and Welfare, Helsinki, Finland.,Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Unit for Pediatrics, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pia Villa
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Hannele Laivuori
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.,Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland.,Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Stefan R Hansson
- Department of Clinical Sciences Lund, Skåne University Hospital, Lund, Sweden.,Department of Obstetrics and Gynecology, Lund University, Lund, Sweden
| |
Collapse
|
15
|
Åkerström B, Rosenlöf L, Hägerwall A, Rutardottir S, Ahlstedt J, Johansson ME, Erlandsson L, Allhorn M, Gram M. rA1M-035, a Physicochemically Improved Human Recombinant α 1-Microglobulin, Has Therapeutic Effects in Rhabdomyolysis-Induced Acute Kidney Injury. Antioxid Redox Signal 2019; 30:489-504. [PMID: 29471681 PMCID: PMC6338582 DOI: 10.1089/ars.2017.7181] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIMS Human α1-microglobulin (A1M) is an endogenous reductase and radical- and heme-binding protein with physiological antioxidant protective functions. Recombinant human A1M (rA1M) has been shown to have therapeutic properties in animal models of preeclampsia, a pregnancy disease associated with oxidative stress. Recombinant A1M, however, lacks glycosylation, and shows lower solubility and stability than A1M purified from human plasma. The aims of this work were to (i) use site-directed mutagenesis to improve the physicochemical properties of rA1M, (ii) demonstrate that the physicochemically improved rA1M displays full in vitro cell protective effects as recombinant wild-type A1M (rA1M-wt), and (iii) show its therapeutic potential in vivo against acute kidney injury (AKI), another disease associated with oxidative stress. RESULTS A novel recombinant A1M-variant (rA1M-035) with three amino acid substitutions was constructed, successfully expressed, and purified. rA1M-035 had improved solubility and stability compared with rA1M-wt, and showed intact in vitro heme-binding, reductase, antioxidation, and cell protective activities. Both rA1M-035 and rA1M-wt showed, for the first time, potential in vivo protective effects on kidneys using a mouse rhabdomyolysis glycerol injection model of AKI. INNOVATION A novel recombinant A1M-variant, rA1M-035, was engineered. This protein showed improved solubility and stability compared with rA1M-wt, full in vitro functional activity, and potential protection against AKI in an in vivo rhabdomyolysis mouse model. CONCLUSION The new rA1M-035 is a better drug candidate than rA1M-wt for treatment of AKI and preeclampsia in human patients.
Collapse
Affiliation(s)
- Bo Åkerström
- 1 Sections for Infection Medicine and Department of Clinical Sciences, Lund University , Lund, Sweden
| | | | | | | | - Jonas Ahlstedt
- 1 Sections for Infection Medicine and Department of Clinical Sciences, Lund University , Lund, Sweden
| | - Maria E Johansson
- 1 Sections for Infection Medicine and Department of Clinical Sciences, Lund University , Lund, Sweden
| | - Lena Erlandsson
- 3 Sections for Obstetrics and Gynecology, Department of Clinical Sciences, Lund University , Lund, Sweden
| | - Maria Allhorn
- 1 Sections for Infection Medicine and Department of Clinical Sciences, Lund University , Lund, Sweden
| | - Magnus Gram
- 1 Sections for Infection Medicine and Department of Clinical Sciences, Lund University , Lund, Sweden
| |
Collapse
|
16
|
Marshall SA, Cox AG, Parry LJ, Wallace EM. Targeting the vascular dysfunction: Potential treatments for preeclampsia. Microcirculation 2018; 26:e12522. [PMID: 30556222 DOI: 10.1111/micc.12522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/22/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022]
Abstract
Preeclampsia is a pregnancy-specific disorder, primarily characterized by new-onset hypertension in combination with a variety of other maternal or fetal signs. The pathophysiological mechanisms underlying the disease are still not entirely clear. Systemic maternal vascular dysfunction underlies the clinical features of preeclampsia. It is a result of oxidative stress and the actions of excessive anti-angiogenic factors, such as soluble fms-like tyrosine kinase, soluble endoglin, and activin A, released by a dysfunctional placenta. The vascular dysfunction then leads to impaired regulation and secretion of relaxation factors and an increase in sensitivity/production of constrictors. This results in a more constricted vasculature rather than the relaxed vasodilated state associated with normal pregnancy. Currently, the only effective "treatment" for preeclampsia is delivery of the placenta and therefore the baby. Often, this means a preterm delivery to save the life of the mother, with all the attendant risks and burdens associated with fetal prematurity. To lessen this burden, there is a pressing need for more effective treatments that target the maternal vascular dysfunction that underlies the hypertension. This review details the vascular effects of key drugs undergoing clinical assessment as potential treatments for women with preeclampsia.
Collapse
Affiliation(s)
- Sarah A Marshall
- Departments of Obstetrics and Gynaecology and Medicine, School of Clinical Sciences, The Ritchie Centre, Monash University, Clayton, Victoria, Australia
| | - Annie G Cox
- Departments of Obstetrics and Gynaecology and Medicine, School of Clinical Sciences, The Ritchie Centre, Monash University, Clayton, Victoria, Australia
| | - Laura J Parry
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Euan M Wallace
- Departments of Obstetrics and Gynaecology and Medicine, School of Clinical Sciences, The Ritchie Centre, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
17
|
Larsson S, Åkerström B, Gram M, Lohmander LS, Struglics A. α1-Microglobulin Protects Against Bleeding-Induced Oxidative Damage in Knee Arthropathies. Front Physiol 2018; 9:1596. [PMID: 30505280 PMCID: PMC6250851 DOI: 10.3389/fphys.2018.01596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/24/2018] [Indexed: 11/13/2022] Open
Abstract
Knee injury increases the risk of developing knee osteoarthritis (OA). Recent evidence suggests involvement of oxidative stress induced by inflammation and bleeding in the joint. This study investigates the role in this process of α1-microglobulin (A1M), a plasma and tissue antioxidant protein with reducing function, and heme- and radical-binding properties. We studied matched knee synovial fluid (sf) and serum (s) samples from 122 subjects (mean age 40 years, 31% females): 10 were knee healthy references, 13 had acute inflammatory arthritis (AIA), 79 knee injury 0–10 years prior to sampling, and 20 knee OA. Using immunoassays, we measured sf-A1M and s-A1M, sf-hemoglobin (sf-Hb), sf-total free heme (sf-Heme), and sf-carbonyl groups (sf-Carbonyl). We explored associations by partial correlation, or linear regression models with adjustments for age, sex and diagnosis, and evaluated diagnostic capacity by area under the receiver operator characteristics curve (AUC). The AIA group had 1.2- to 1.7-fold higher sf-A1M and s-A1M concentrations compared to the other diagnostic groups; other biomarkers showed no between-group differences. sf-A1M and s-A1M were with AUC of 0.76 and 0.78, respectively, diagnostic for AIA. In the injury group, the amount of bleeding in the joint was inversely correlated to time after injury when measured as sf-Heme (r = -0.41, p < 0.001), but not when measured as sf-Hb (r = -0.19, p = 0.098). A similar inverse association with time after injury was noted for sf-A1M (r = -0.30, p = 0.007), but not for s-A1M and sf-Carbonyl. Linear regression models showed that sf-Heme was more strongly associated with sf-A1M and sf-Carbonyl than sf-Hb. Independent of diagnosis, sf-Heme explained 5.7% of the variability in sf-A1M and 3.0% in the variability in sf-Carbonyl, but appeared unrelated to s-A1M. High sf-A1M and low sf-Heme or sf-Hb were independently associated with low sf-Carbonyl. In conclusion, our results demonstrate that independent of disease, Hb and heme within a knee joint correlates with an increased sf-A1M concentration that appears to be protective of oxidative damage, i.e., a reduction in carbonyl groups. High concentrations of A1M in synovial fluid and serum was further diagnostic for AIA.
Collapse
Affiliation(s)
- Staffan Larsson
- Department of Clinical Sciences Lund, Orthopaedics, Faculty of Medicine, Lund University, Lund, Sweden
- *Correspondence: Staffan Larsson,
| | - Bo Åkerström
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Magnus Gram
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - L. Stefan Lohmander
- Department of Clinical Sciences Lund, Orthopaedics, Faculty of Medicine, Lund University, Lund, Sweden
| | - André Struglics
- Department of Clinical Sciences Lund, Orthopaedics, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
18
|
Gunnarsson R, Åkerström B, Hansson SR, Gram M. Recombinant alpha-1-microglobulin: a potential treatment for preeclampsia. Drug Discov Today 2017; 22:736-743. [DOI: 10.1016/j.drudis.2016.12.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 11/21/2016] [Accepted: 12/08/2016] [Indexed: 01/31/2023]
|
19
|
Anderson UD, Gram M, Ranstam J, Thilaganathan B, Kerström B, Hansson SR. Fetal hemoglobin, α1-microglobulin and hemopexin are potential predictive first trimester biomarkers for preeclampsia. Pregnancy Hypertens 2016; 6:103-9. [PMID: 27155336 DOI: 10.1016/j.preghy.2016.02.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/26/2016] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Overproduction of cell-free fetal hemoglobin (HbF) in the preeclamptic placenta has been recently implicated as a new etiological factor of preeclampsia. In this study, maternal serum levels of HbF and the endogenous hemoglobin/heme scavenging systems were evaluated as predictive biomarkers for preeclampsia in combination with uterine artery Doppler ultrasound. STUDY DESIGN Case-control study including 433 women in early pregnancy (mean 13.7weeks of gestation) of which 86 subsequently developed preeclampsia. The serum concentrations of HbF, total cell-free hemoglobin, hemopexin, haptoglobin and α1-microglobulin were measured in maternal serum. All patients were examined with uterine artery Doppler ultrasound. Logistic regression models were developed, which included the biomarkers, ultrasound indices, and maternal risk factors. RESULTS There were significantly higher serum concentrations of HbF and α1-microglobulin and significantly lower serum concentrations of hemopexin in patients who later developed preeclampsia. The uterine artery Doppler ultrasound results showed significantly higher pulsatility index values in the preeclampsia group. The optimal prediction model was obtained by combining HbF, α1-microglobulin and hemopexin in combination with the maternal characteristics parity, diabetes and pre-pregnancy hypertension. The optimal sensitivity for all preeclampsia was 60% at 95% specificity. CONCLUSIONS Overproduction of placentally derived HbF and depletion of hemoglobin/heme scavenging mechanisms are involved in the pathogenesis of preeclampsia. The combination of HbF and α1-microglobulin and/or hemopexin may serve as a prediction model for preeclampsia in combination with maternal risk factors and/or uterine artery Doppler ultrasound.
Collapse
Affiliation(s)
- Ulrik Dolberg Anderson
- Section of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Sweden; Skåne University Hospital, Malmö/Lund, Sweden.
| | - Magnus Gram
- Department of Clinical Sciences, Lund, Infection Medicine, Lund University, Sweden
| | - Jonas Ranstam
- Department of Clinical Sciences, RC Syd, Lund University, Sweden
| | - Basky Thilaganathan
- Fetal Medicine Unit, St. George's University Hospital, London, United Kingdom
| | - Bo Kerström
- Department of Clinical Sciences, Lund, Infection Medicine, Lund University, Sweden
| | - Stefan R Hansson
- Section of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Sweden; Skåne University Hospital, Malmö/Lund, Sweden
| |
Collapse
|
20
|
Ahlstedt J, Tran TA, Strand SE, Gram M, Åkerström B. Human Anti-Oxidation Protein A1M--A Potential Kidney Protection Agent in Peptide Receptor Radionuclide Therapy. Int J Mol Sci 2015; 16:30309-20. [PMID: 26694383 PMCID: PMC4691176 DOI: 10.3390/ijms161226234] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 11/28/2015] [Accepted: 12/11/2015] [Indexed: 11/16/2022] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) has been in clinical use for 15 years to treat metastatic neuroendocrine tumors. PRRT is limited by reabsorption and retention of the administered radiolabeled somatostatin analogues in the proximal tubule. Consequently, it is essential to develop and employ methods to protect the kidneys during PRRT. Today, infusion of positively charged amino acids is the standard method of kidney protection. Other methods, such as administration of amifostine, are still under evaluation and show promising results. α1-microglobulin (A1M) is a reductase and radical scavenging protein ubiquitously present in plasma and extravascular tissue. Human A1M has antioxidation properties and has been shown to prevent radiation-induced in vitro cell damage and protect non-irradiated surrounding cells. It has recently been shown in mice that exogenously infused A1M and the somatostatin analogue octreotide are co-localized in proximal tubules of the kidney after intravenous infusion. In this review we describe the current situation of kidney protection during PRRT, discuss the necessity and implications of more precise dosimetry and present A1M as a new, potential candidate for renal protection during PRRT and related targeted radionuclide therapies.
Collapse
Affiliation(s)
- Jonas Ahlstedt
- Section for Infection Medicine, Department of Clinical Sciences in Lund, Lund University, Lund 221 84, Sweden.
| | - Thuy A Tran
- Lund University Bioimaging Center, Lund University, Lund 221 84, Sweden.
| | - Sven-Erik Strand
- Section of Medical Radiation Physics, Department of Clinical Sciences in Lund, Lund University, Lund 221 84, Sweden.
| | - Magnus Gram
- Section for Infection Medicine, Department of Clinical Sciences in Lund, Lund University, Lund 221 84, Sweden.
| | - Bo Åkerström
- Section for Infection Medicine, Department of Clinical Sciences in Lund, Lund University, Lund 221 84, Sweden.
| |
Collapse
|
21
|
Proteomic profiling reveals α1-antitrypsin, α1-microglobulin, and clusterin as preeclampsia-related serum proteins in pregnant women. Taiwan J Obstet Gynecol 2015; 54:499-504. [DOI: 10.1016/j.tjog.2014.01.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2014] [Indexed: 11/23/2022] Open
|
22
|
Nääv Å, Erlandsson L, Axelsson J, Larsson I, Johansson M, Wester-Rosenlöf L, Mörgelin M, Casslén V, Gram M, Åkerström B, Hansson SR. A1M Ameliorates Preeclampsia-Like Symptoms in Placenta and Kidney Induced by Cell-Free Fetal Hemoglobin in Rabbit. PLoS One 2015; 10:e0125499. [PMID: 25955715 PMCID: PMC4425457 DOI: 10.1371/journal.pone.0125499] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 03/17/2015] [Indexed: 11/18/2022] Open
Abstract
Preeclampsia is one of the most serious pregnancy-related diseases and clinically manifests as hypertension and proteinuria after 20 gestational weeks. The worldwide prevalence is 3-8% of pregnancies, making it the most common cause of maternal and fetal morbidity and mortality. Preeclampsia lacks an effective therapy, and the only “cure” is delivery. We have previously shown that increased synthesis and accumulation of cell-free fetal hemoglobin (HbF) in the placenta is important in the pathophysiology of preeclampsia. Extracellular hemoglobin (Hb) and its metabolites induce oxidative stress, which may lead to acute renal failure and vascular dysfunction seen in preeclampsia. The human endogenous protein, α1-microglobulin (A1M), removes cell-free heme-groups and induces natural tissue repair mechanisms. Exogenously administered A1M has been shown to alleviate the effects of Hb-induced oxidative stress in rat kidneys. Here we attempted to establish an animal model mimicking the human symptoms at stage two of preeclampsia by administering species-specific cell-free HbF starting mid-gestation until term, and evaluated the therapeutic effect of A1M on the induced symptoms. Female pregnant rabbits received HbF infusions i.v. with or without A1M every second day from gestational day 20. The HbF-infused animals developed proteinuria and a significantly increased glomerular sieving coefficient in kidney that was ameliorated by co-administration of A1M. Transmission electron microscopy analysis of kidney and placenta showed both intracellular and extracellular tissue damages after HbF-treatment, while A1M co-administration resulted in a significant reduction of the structural and cellular changes. Neither of the HbF-treated animals displayed any changes in blood pressure during pregnancy. In conclusion, infusion of cell-free HbF in the pregnant rabbits induced tissue damage and organ failure similar to those seen in preeclampsia, and was restored by co-administration of A1M. This study provides preclinical evidence supporting further examination of A1M as a potential new therapy for preeclampsia.
Collapse
Affiliation(s)
- Åsa Nääv
- Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- * E-mail:
| | - Lena Erlandsson
- Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Josefin Axelsson
- Nephrology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Irene Larsson
- Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Martin Johansson
- Clinical Pathology, Department of Laboratory Medicine, Lund University, Malmö, Sweden
| | - Lena Wester-Rosenlöf
- Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Matthias Mörgelin
- Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Vera Casslén
- Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Magnus Gram
- Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Bo Åkerström
- Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Stefan R. Hansson
- Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
23
|
Hansson SR, Nääv Å, Erlandsson L. Oxidative stress in preeclampsia and the role of free fetal hemoglobin. Front Physiol 2015; 5:516. [PMID: 25628568 PMCID: PMC4292435 DOI: 10.3389/fphys.2014.00516] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/16/2014] [Indexed: 02/04/2023] Open
Abstract
Preeclampsia is a leading cause of pregnancy complications and affects 3-7% of pregnant women. This review summarizes the current knowledge of a new potential etiology of the disease, with a special focus on hemoglobin-induced oxidative stress. Furthermore, we also suggest hemoglobin as a potential target for therapy. Gene and protein profiling studies have shown increased expression and accumulation of free fetal hemoglobin in the preeclamptic placenta. Predominantly due to oxidative damage to the placental barrier, fetal hemoglobin leaks over to the maternal circulation. Free hemoglobin and its metabolites are toxic in several ways; (a) ferrous hemoglobin (Fe(2+)) binds strongly to the vasodilator nitric oxide (NO) and reduces the availability of free NO, which results in vasoconstriction, (b) hemoglobin (Fe(2+)) with bound oxygen spontaneously generates free oxygen radicals, and (c) the heme groups create an inflammatory response by inducing activation of neutrophils and cytokine production. The endogenous protein α1-microglobulin, with radical and heme binding properties, has shown both ex vivo and in vivo to have the ability to counteract free hemoglobin-induced placental and kidney damage. Oxidative stress in general, and more specifically fetal hemoglobin-induced oxidative stress, could play a key role in the pathology of preeclampsia seen both in the placenta and ultimately in the maternal endothelium.
Collapse
Affiliation(s)
- Stefan R. Hansson
- Department of Obstetrics and Gynecology, Institute for Clinical Sciences, Lund UniversityLund, Sweden
| | | | | |
Collapse
|
24
|
Åkerström B, Gram M. A1M, an extravascular tissue cleaning and housekeeping protein. Free Radic Biol Med 2014; 74:274-82. [PMID: 25035076 DOI: 10.1016/j.freeradbiomed.2014.06.025] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/26/2014] [Accepted: 06/27/2014] [Indexed: 02/02/2023]
Abstract
Alpha-1-microglobulin (A1M) is a small protein found intra- and extracellularly in all tissues of vertebrates. The protein was discovered 40 years ago and its physiological role remained unknown for a long time. A series of recent publications have demonstrated that A1M is a vital part of tissue housekeeping. A strongly electronegative free thiol group forms the structural basis of heme-binding, reductase, and radical-trapping properties. A rapid flow of liver-produced A1M through blood and extravascular compartments ensures clearing of biological fluids from heme and free radicals and repair of oxidative lesions. After binding, both the radicals and the A1M are electroneutral and therefore do not present any further oxidative stress to tissues. The biological cleaning cycle is completed by glomerular filtration, renal degradation, and urinary excretion of A1M heavily modified by covalently linked radicals and heme groups. Based on its role as a tissue housekeeping cleaning factor, A1M constitutes a potential therapeutic drug candidate in treatment or prophylaxis of diseases or conditions that are associated with pathological oxidative stress elements.
Collapse
Affiliation(s)
- Bo Åkerström
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden.
| | - Magnus Gram
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
25
|
Fetal hemoglobin in preeclampsia: a new causative factor, a tool for prediction/diagnosis and a potential target for therapy. Curr Opin Obstet Gynecol 2014; 25:448-55. [PMID: 24185004 DOI: 10.1097/gco.0000000000000022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE OF REVIEW Preeclampsia, one of the leading causes of pregnancy complications, affects 3-7% of pregnant women. This review summarizes the present knowledge of a new potential cause of the disease and suggests a method for its prediction/diagnosis and a possible treatment, both based on the recent findings on the involvement of fetal hemoglobin (HbF) and the heme and radical scavenging protein A1M (alpha-1-microglobulin). RECENT FINDINGS Gene and protein profiling studies have independently shown that increased amount of free HbF is accumulated in the preeclampsia placenta. As a result of a predominantly oxidative damage to the blood-placenta barrier, HbF leaks over to the maternal blood circulation. Elevated levels can be measured already in the first trimester, and later in pregnancy, the levels correlate with the blood pressure in women with preeclampsia. Ex-vivo data show that the human protein A1M, an endogeneous antioxidation protection protein, can prevent Hb-induced damage to the placenta, restore the blood-placental barrier and prevent maternal tissue damage. SUMMARY Free HbF may provide both a predictive and a diagnostic clinical biomarker from the first trimester. A1M has the potential as a future pharmacological treatment for preeclampsia.
Collapse
|
26
|
Wester-Rosenlöf L, Casslén V, Axelsson J, Edström-Hägerwall A, Gram M, Holmqvist M, Johansson ME, Larsson I, Ley D, Marsal K, Mörgelin M, Rippe B, Rutardottir S, Shohani B, Åkerström B, Hansson SR. A1M/α1-microglobulin protects from heme-induced placental and renal damage in a pregnant sheep model of preeclampsia. PLoS One 2014; 9:e86353. [PMID: 24489717 PMCID: PMC3904882 DOI: 10.1371/journal.pone.0086353] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 12/06/2013] [Indexed: 02/03/2023] Open
Abstract
Preeclampsia (PE) is a serious pregnancy complication that manifests as hypertension and proteinuria after the 20(th) gestation week. Previously, fetal hemoglobin (HbF) has been identified as a plausible causative factor. Cell-free Hb and its degradation products are known to cause oxidative stress and tissue damage, typical of the PE placenta. A1M (α1-microglobulin) is an endogenous scavenger of radicals and heme. Here, the usefulness of A1M as a treatment for PE is investigated in the pregnant ewe PE model, in which starvation induces PE symptoms via hemolysis. Eleven ewes, in late pregnancy, were starved for 36 hours and then treated with A1M (n = 5) or placebo (n = 6) injections. After injections, the ewes were re-fed and observed for additional 72 hours. They were monitored for blood pressure, proteinuria, blood cell distribution and clinical and inflammation markers in plasma. Before termination, the utero-placental circulation was analyzed with Doppler velocimetry and the kidney glomerular function was analyzed by Ficoll sieving. At termination, blood, kidney and placenta samples were collected and analyzed for changes in gene expression and tissue structure. The starvation resulted in increased amounts of the hemolysis marker bilirubin in the blood, structural damages to the placenta and kidneys and an increased glomerular sieving coefficient indicating a defect filtration barrier. Treatment with A1M ameliorated these changes without signs of side-effects. In conclusion, A1M displayed positive therapeutic effects in the ewe starvation PE model, and was well tolerated. Therefore, we suggest A1M as a plausible treatment for PE in humans.
Collapse
Affiliation(s)
| | - Vera Casslén
- Department of Obstetrics and Gynecology, Lund University, Lund, Sweden
| | | | | | - Magnus Gram
- Department of Infection Medicine, Lund University, Lund, Sweden
| | - Madlene Holmqvist
- Department of Obstetrics and Gynecology, Lund University, Lund, Sweden
| | | | - Iréne Larsson
- Department of Obstetrics and Gynecology, Lund University, Lund, Sweden
| | - David Ley
- Department of Pediatrics, Lund University, Lund, Sweden
| | - Karel Marsal
- Department of Obstetrics and Gynecology, Lund University, Lund, Sweden
| | | | - Bengt Rippe
- Department of Nephrology, Lund University, Lund, Sweden
| | | | - Behnaz Shohani
- Department of Obstetrics and Gynecology, Lund University, Lund, Sweden
| | - Bo Åkerström
- Department of Infection Medicine, Lund University, Lund, Sweden
- * E-mail: (BÅ); (SRH)
| | - Stefan R. Hansson
- Department of Obstetrics and Gynecology, Lund University, Lund, Sweden
- * E-mail: (BÅ); (SRH)
| |
Collapse
|
27
|
Sverrisson K, Axelsson J, Rippe A, Gram M, Åkerström B, Hansson SR, Rippe B. Extracellular fetal hemoglobin induces increases in glomerular permeability: inhibition with α1-microglobulin and tempol. Am J Physiol Renal Physiol 2013; 306:F442-8. [PMID: 24338823 DOI: 10.1152/ajprenal.00502.2013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Extracellular fetal hemoglobin (HbF) and adult hemoglobin (HbA) are proinflammatory and generate ROS. Increased plasma levels of extracellular HbF have recently been reported to occur in early preeclampsia. α1-Microglobulin (A1M) is a physiological heme-binding protein and radical scavenger that has been shown to counteract vascular permeability increases induced by HbA in the perfused placenta. The present study was performed to investigate whether HbF and HbA will increase glomerular permeability in vivo and to test whether A1M and tempol, a ROS scavenger, can prevent their effects. Anesthetized Wistar rats were continuously infused intravenously with either HbA, HbF, or cyano-inactivated HbF together with FITC-Ficoll-70/400, inulin, and (51)Cr-labeled EDTA for 2 h. Plasma samples and urine samples (left ureter) were taken repeatedly and analyzed by high-performance size exclusion chromatography to assess glomerular sieving coefficients for Ficoll of radius 10-80 Å. In separate experiments, A1M or tempol was given before and during Hb infusions. Extracellular HbF caused rapid, transient increases in glomerular permeability to large Ficoll molecules (50-80Å), contrary to the effects of HbA and cyano-inactivated HbF. For HbF, glomerular sieving coefficients for Ficoll of radius 60Å increased from 3.85 ± 0.85 × 10(-5) to 2.60 ± 0.96 × 10(-4) at 15 min, changes that were abrogated by tempol and reduced by A1M. In conclusion, our data demonstrate that extracellular HbF, infused systemically, can acutely increase glomerular permeability through inducing oxidative stress.
Collapse
Affiliation(s)
- Kristinn Sverrisson
- Dept. of Nephrology, Lund Univ., Skåne Univ. Hospital, Lund S-211 85, Sweden.
| | | | | | | | | | | | | |
Collapse
|
28
|
Gram M, Sveinsdottir S, Ruscher K, Hansson SR, Cinthio M, Akerström B, Ley D. Hemoglobin induces inflammation after preterm intraventricular hemorrhage by methemoglobin formation. J Neuroinflammation 2013; 10:100. [PMID: 23915174 PMCID: PMC3750409 DOI: 10.1186/1742-2094-10-100] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 07/26/2013] [Indexed: 12/22/2022] Open
Abstract
Background Cerebral intraventricular hemorrhage (IVH) is a major cause of severe neurodevelopmental impairment in preterm infants. To date, no therapy is available that prevents infants from developing serious neurological disability following IVH. Thus, to develop treatment strategies for IVH, it is essential to characterize the initial sequence of molecular events that leads to brain damage. In this study, we investigated extracellular hemoglobin (Hb) as a causal initiator of inflammation in preterm IVH. Methods Using a preterm rabbit pup model, we investigated the molecular mechanisms and events following IVH. We also characterized the concentrations of cell-free Hb metabolites and pro-inflammatory mediators in the cerebrospinal fluid (CSF) of preterm human infants and rabbit pups. Finally, Hb metabolites were evaluated as causal initiators of inflammation in primary rabbit astrocyte cell cultures. Results Following IVH in preterm rabbit pups, the intraventricular CSF concentration of cell-free methemoglobin (metHb) increased from 24 to 72 hours and was strongly correlated with the concentration of TNFα at 72 hours (r2 = 0.896, P <0.001). Also, the mRNA expression of TNFα, IL-1β, and Toll-like receptor-4 and TNFα protein levels were significantly increased in periventricular tissue at 72 hours, which was accompanied by extensive astrocyte activation (that is, glial fibrillary acidic protein (GFAP)staining). Furthermore, exposure of primary rabbit astrocyte cell cultures to metHb caused a dose-dependent increase in TNFα mRNA and protein levels, which was not observed following exposure to oxyhemoglobin (oxyHb) or hemin. Finally, a positive correlation (r2 = 0.237, P <0.03) between metHb and TNFα concentrations was observed in the CSF of preterm human infants following IVH. Conclusions Following preterm IVH, increased metHb formation in the intraventricular space induces expression of pro-inflammatory cytokines. Thus, the formation of metHb might be a crucial initial event in the development of brain damage following preterm IVH. Accordingly, removal, scavenging, or neutralization of Hb could present a therapeutic opportunity and plausible approach to decreasing the damage in the immature brain following preterm IVH.
Collapse
Affiliation(s)
- Magnus Gram
- Division of Infection Medicine, Lund University, S-221 84 Lund, Sweden.
| | | | | | | | | | | | | |
Collapse
|
29
|
Hong J, Zhao YX, Xiao BL, Moosavi-Movahedi AA, Ghourchian H, Sheibani N. Direct electrochemistry of hemoglobin immobilized on a functionalized multi-walled carbon nanotubes and gold nanoparticles nanocomplex-modified glassy carbon electrode. SENSORS 2013; 13:8595-611. [PMID: 23881129 PMCID: PMC3758613 DOI: 10.3390/s130708595] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 06/17/2013] [Accepted: 06/30/2013] [Indexed: 12/02/2022]
Abstract
Direct electron transfer of hemoglobin (Hb) was realized by immobilizing Hb on a carboxyl functionalized multi-walled carbon nanotubes (FMWCNTs) and gold nanoparticles (AuNPs) nanocomplex-modified glassy carbon electrode. The ultraviolet-visible absorption spectrometry (UV-Vis), transmission electron microscopy (TEM) and Fourier transform infrared (FTIR) methods were utilized for additional characterization of the AuNPs and FMWCNTs. The cyclic voltammogram of the modified electrode has a pair of well-defined quasi-reversible redox peaks with a formal potential of −0.270 ± 0.002 V (vs. Ag/AgCl) at a scan rate of 0.05 V/s. The heterogeneous electron transfer constant (ks) was evaluated to be 4.0 ± 0.2 s−1. The average surface concentration of electro-active Hb on the surface of the modified glassy carbon electrode was calculated to be 6.8 ± 0.3 × 10−10 mol cm−2. The cathodic peak current of the modified electrode increased linearly with increasing concentration of hydrogen peroxide (from 0.05 nM to 1 nM) with a detection limit of 0.05 ± 0.01 nM. The apparent Michaelis-Menten constant (Kmapp) was calculated to be 0.85 ± 0.1 nM. Thus, the modified electrode could be applied as a third generation biosensor with high sensitivity, long-term stability and low detection limit.
Collapse
Affiliation(s)
- Jun Hong
- School of Life Sciences, Henan University, JinMing Road, Kaifeng 475000, China; E-Mails: (Y.-X.Z.); (B.-L.X.)
- Authors to whom correspondence should be addressed; E-Mails: (J.H.); (A.A.M.-M.); Tel.: +86-137-8116-1597 (J.H.); Fax: +86-378-388-6258 (J.H.); Tel.: +98-21-640-3957 (A.A.M.-M.); Fax: +98-21-640-4680 (A.A.M.-M.)
| | - Ying-Xue Zhao
- School of Life Sciences, Henan University, JinMing Road, Kaifeng 475000, China; E-Mails: (Y.-X.Z.); (B.-L.X.)
| | - Bao-Lin Xiao
- School of Life Sciences, Henan University, JinMing Road, Kaifeng 475000, China; E-Mails: (Y.-X.Z.); (B.-L.X.)
| | - Ali Akbar Moosavi-Movahedi
- Institute of Biochemistry and Biophysics, University of Tehran, Enquelab Avenue, P.O. Box 13145-1384, Tehran, Iran; E-Mail:
- Authors to whom correspondence should be addressed; E-Mails: (J.H.); (A.A.M.-M.); Tel.: +86-137-8116-1597 (J.H.); Fax: +86-378-388-6258 (J.H.); Tel.: +98-21-640-3957 (A.A.M.-M.); Fax: +98-21-640-4680 (A.A.M.-M.)
| | - Hedayatollah Ghourchian
- Institute of Biochemistry and Biophysics, University of Tehran, Enquelab Avenue, P.O. Box 13145-1384, Tehran, Iran; E-Mail:
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, 600 Highland Avenue, K6/456 CSC, Madison, WI 53792-4673, USA; E-Mail:
| |
Collapse
|
30
|
Olsson MG, Rosenlöf LW, Kotarsky H, Olofsson T, Leanderson T, Mörgelin M, Fellman V, Åkerström B. The radical-binding lipocalin A1M binds to a Complex I subunit and protects mitochondrial structure and function. Antioxid Redox Signal 2013; 18:2017-28. [PMID: 23157686 DOI: 10.1089/ars.2012.4658] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIMS During cell death, energy-consuming cell degradation and recycling programs are performed. Maintenance of energy delivery during cell death is therefore crucial, but the mechanisms to keep the mitochondrial functions intact during these processes are poorly understood. We have investigated the hypothesis that the heme- and radical-binding ubiquitous protein α1-microglobulin (A1M) is involved in protection of the mitochondria against oxidative insult during cell death. RESULTS Using blood cells, keratinocytes, and liver cells, we show that A1M binds with high affinity to apoptosis-induced cells and is localized to mitochondria. The mitochondrial Complex I subunit NDUFAB1 was identified as a major molecular target of the A1M binding. Furthermore, A1M was shown to inhibit the swelling of mitochondria, and to reverse the severely abrogated ATP-production of mitochondria when exposed to heme and reactive oxygen species (ROS). INNOVATION Import of the radical- and heme-binding protein A1M from the extracellular compartment confers protection of the mitochondrial structure and function during cellular insult. CONCLUSION A1M binds to a subunit of Complex I and has a role in assisting the mitochondria to maintain its energy delivery during cell death. A1M may also, at the same time, counteract and eliminate the ROS generated by the mitochondrial respiration to prevent oxidative damage to surrounding healthy tissue.
Collapse
Affiliation(s)
- Magnus G Olsson
- Division of Infection Medicine, Lund University, Lund, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
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: 70] [Impact Index Per Article: 5.4] [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
|
32
|
Chorley BN, Campbell MR, Wang X, Karaca M, Sambandan D, Bangura F, Xue P, Pi J, Kleeberger SR, Bell DA. Identification of novel NRF2-regulated genes by ChIP-Seq: influence on retinoid X receptor alpha. Nucleic Acids Res 2012; 40:7416-29. [PMID: 22581777 PMCID: PMC3424561 DOI: 10.1093/nar/gks409] [Citation(s) in RCA: 445] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 04/18/2012] [Accepted: 04/19/2012] [Indexed: 12/14/2022] Open
Abstract
Cellular oxidative and electrophilic stress triggers a protective response in mammals regulated by NRF2 (nuclear factor (erythroid-derived) 2-like; NFE2L2) binding to deoxyribonucleic acid-regulatory sequences near stress-responsive genes. Studies using Nrf2-deficient mice suggest that hundreds of genes may be regulated by NRF2. To identify human NRF2-regulated genes, we conducted chromatin immunoprecipitation (ChIP)-sequencing experiments in lymphoid cells treated with the dietary isothiocyanate, sulforaphane (SFN) and carried out follow-up biological experiments on candidates. We found 242 high confidence, NRF2-bound genomic regions and 96% of these regions contained NRF2-regulatory sequence motifs. The majority of binding sites were near potential novel members of the NRF2 pathway. Validation of selected candidate genes using parallel ChIP techniques and in NRF2-silenced cell lines indicated that the expression of about two-thirds of the candidates are likely to be directly NRF2-dependent including retinoid X receptor alpha (RXRA). NRF2 regulation of RXRA has implications for response to retinoid treatments and adipogenesis. In mouse, 3T3-L1 cells' SFN treatment affected Rxra expression early in adipogenesis, and knockdown of Nrf2-delayed Rxra expression, both leading to impaired adipogenesis.
Collapse
Affiliation(s)
- Brian N. Chorley
- Environmental Genomics Section, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences, National Institutes of Health, The Hamner Institutes and Environmental Genetics Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Michelle R. Campbell
- Environmental Genomics Section, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences, National Institutes of Health, The Hamner Institutes and Environmental Genetics Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Xuting Wang
- Environmental Genomics Section, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences, National Institutes of Health, The Hamner Institutes and Environmental Genetics Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Mehmet Karaca
- Environmental Genomics Section, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences, National Institutes of Health, The Hamner Institutes and Environmental Genetics Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Deepa Sambandan
- Environmental Genomics Section, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences, National Institutes of Health, The Hamner Institutes and Environmental Genetics Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Fatu Bangura
- Environmental Genomics Section, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences, National Institutes of Health, The Hamner Institutes and Environmental Genetics Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Peng Xue
- Environmental Genomics Section, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences, National Institutes of Health, The Hamner Institutes and Environmental Genetics Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Jingbo Pi
- Environmental Genomics Section, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences, National Institutes of Health, The Hamner Institutes and Environmental Genetics Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Steven R. Kleeberger
- Environmental Genomics Section, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences, National Institutes of Health, The Hamner Institutes and Environmental Genetics Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Douglas A. Bell
- Environmental Genomics Section, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences, National Institutes of Health, The Hamner Institutes and Environmental Genetics Section, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| |
Collapse
|
33
|
Cederlund M, Ghosh F, Arnér K, Andréasson S, Akerström B. Vitreous levels of oxidative stress biomarkers and the radical-scavenger α1-microglobulin/A1M in human rhegmatogenous retinal detachment. Graefes Arch Clin Exp Ophthalmol 2012; 251:725-32. [PMID: 22829194 DOI: 10.1007/s00417-012-2113-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 06/25/2012] [Accepted: 07/01/2012] [Indexed: 11/26/2022] Open
Abstract
PURPOSE To explore oxidative stress and the radical scavenger α(1)-microglobulin (A1M) in the vitreous body of human eyes with primary rhegmatogenous retinal detachment (RRD). METHODS Levels of carbonyl groups, a marker of oxidative stress, and A1M were measured by ELISA and RIA in 14 vitreous samples derived from patients suffering from RRD, and compared with 14 samples from macula hole (MH) patients. Carbonyl group and A1M levels in RRD samples were statistically related to detachment characteristics. Analysis of total protein level, SDS-PAGE, and Western blotting of A1M was also performed. In a separate experiment, mRNA expression of A1M was measured by RT-PCR in rat retina explants. RESULTS Levels of carbonyl groups and A1M varied widely in RRD vitreous samples, but were significantly higher in samples derived from eyes with large detachment area and macula-off status, while the presence of vitreous hemorrhage did not show any significant correlation. Compared with MH samples, RRD samples displayed significantly higher levels of A1M, whereas changes in total protein levels and carbonyl groups were not significant. Novel forms of A1M, not previously seen in plasma, were found in the vitreous body by Western blotting. Furthermore, A1M expression was seen in rat retina explants and was upregulated after 24 h of culturing. CONCLUSION Oxidative stress is a prominent feature of human eyes with primary RRD, and is directly related to detachment severity. Affected eyes can launch a protective response in the form of the radical scavenger A1M possibly derived from the retina. The results thus indicate potential therapeutic cell loss prevention in RRD by employing the endogeneous radical scavenger A1M.
Collapse
Affiliation(s)
- Martin Cederlund
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, BMC B14, 221 84 Lund, Sweden
| | | | | | | | | |
Collapse
|
34
|
Anderson U, Olsson M, Kristensen K, Åkerström B, Hansson S. Review: Biochemical markers to predict preeclampsia. Placenta 2012; 33 Suppl:S42-7. [DOI: 10.1016/j.placenta.2011.11.021] [Citation(s) in RCA: 136] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 11/21/2011] [Accepted: 11/23/2011] [Indexed: 12/01/2022]
|
35
|
Feng Q, Liu K, Fu J, Zhang Y, Zheng Z, Wang C, Du Y, Ye W. Direct electrochemistry of hemoglobin based on nano-composite film of gold nanopaticles and poly (diallyldimethylammonium chloride) functionalized graphene. Electrochim Acta 2012. [DOI: 10.1016/j.electacta.2011.11.048] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
36
|
Olsson MG, Allhorn M, Larsson J, Cederlund M, Lundqvist K, Schmidtchen A, Sørensen OE, Mörgelin M, Akerström B. Up-regulation of A1M/α1-microglobulin in skin by heme and reactive oxygen species gives protection from oxidative damage. PLoS One 2011; 6:e27505. [PMID: 22096585 PMCID: PMC3214066 DOI: 10.1371/journal.pone.0027505] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 10/18/2011] [Indexed: 01/02/2023] Open
Abstract
During bleeding the skin is subjected to oxidative insults from free heme and radicals, generated from extracellular hemoglobin. The lipocalin α1-microglobulin (A1M) was recently shown to have reductase properties, reducing heme-proteins and other substrates, and to scavenge heme and radicals. We investigated the expression and localization of A1M in skin and the possible role of A1M in the protection of skin tissue from damage induced by heme and reactive oxygen species. Skin explants, keratinocyte cultures and purified collagen I were exposed to heme, reactive oxygen species, and/or A1M and investigated by biochemical methods and electron microscopy. The results demonstrate that A1M is localized ubiquitously in the dermal and epidermal layers, and that the A1M-gene is expressed in keratinocytes and up-regulated after exposure to heme and reactive oxygen species. A1M inhibited the heme- and reactive oxygen species-induced ultrastructural damage, up-regulation of antioxidation and cell cycle regulatory genes, and protein carbonyl formation in skin and keratinocytes. Finally, A1M bound to purified collagen I (Kd = 0.96×10−6 M) and could inhibit and repair the destruction of collagen fibrils by heme and reactive oxygen species. The results suggest that A1M may have a physiological role in protection of skin cells and matrix against oxidative damage following bleeding.
Collapse
Affiliation(s)
- Magnus G Olsson
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Anderson UD, Olsson MG, Rutardóttir S, Centlow M, Kristensen KH, Isberg PE, Thilaganathan B, Åkerström B, Hansson SR. Fetal hemoglobin and α1-microglobulin as first- and early second-trimester predictive biomarkers for preeclampsia. Am J Obstet Gynecol 2011; 204:520.e1-5. [PMID: 21439542 DOI: 10.1016/j.ajog.2011.01.058] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2010] [Revised: 11/08/2010] [Accepted: 01/26/2011] [Indexed: 10/18/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate fetal hemoglobin (HbF) and α(1)-microglobulin (A1M) in maternal serum as first-trimester biomarkers for preeclampsia (PE). STUDY DESIGN The design was a case-control study. We included 96 patients in the first trimester of pregnancy (60 with PE and 36 controls). Venous serum samples were analyzed for HbF and total hemoglobin (Hb) by enzyme-linked immunosorbent assay and for A1M by radioimmunoassay. Sensitivity and specificity was calculated by logistic regression and receiver operating characteristic curve analysis. RESULTS The HbF/Hb ratio and A1M concentration were significantly elevated in serum from women with subsequent development of PE (P < .0001). The optimal sensitivity and specificity was obtained using the biomarkers in combination; 69% sensitivity for a 5% screen positive rate and 90% sensitivity for a 23% screen positive rate. CONCLUSION The study suggests that HbF/Hb ratio in combination with A1M is predictive biomarkers for PE.
Collapse
|
38
|
May K, Rosenlöf L, Olsson MG, Centlow M, Mörgelin M, Larsson I, Cederlund M, Rutardottir S, Siegmund W, Schneider H, Akerström B, Hansson SR. Perfusion of human placenta with hemoglobin introduces preeclampsia-like injuries that are prevented by α1-microglobulin. Placenta 2011; 32:323-32. [PMID: 21356557 DOI: 10.1016/j.placenta.2011.01.017] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 01/28/2011] [Accepted: 01/31/2011] [Indexed: 12/11/2022]
Abstract
BACKGROUND Preeclamptic women have increased plasma levels of free fetal hemoglobin (HbF), increased gene expression of placental HbF and accumulation of free HbF in the placental vascular lumen. Free hemoglobin (Hb) is pro-inflammatory, and causes oxidative stress and tissue damage. METHODOLOGY To show the impact of free Hb in PE, we used the dual ex vivo placental perfusion model. Placentas were perfused with Hb and investigated for physical parameters, Hb leakage, gene expression and morphology. The protective effects of α(1)-microglobulin (A1M), a heme- and radical-scavenger and antioxidant, was investigated. RESULTS Hb-addition into the fetal circulation led to a significant increase of the perfusion pressure and the feto-maternal leakage of free Hb. Morphological damages similar to the PE placentas were observed. Gene array showed up-regulation of genes related to immune response, apoptosis, and oxidative stress. Simultaneous addition of A1M to the maternal circulation inhibited the Hb leakage, morphological damage and gene up-regulation. Furthermore, perfusion with Hb and A1M induced a significant up-regulation of extracellular matrix genes. SIGNIFICANCE The ex vivo Hb-perfusion of human placenta resulted in physiological and morphological changes and a gene expression profile similar to what is observed in PE placentas. These results underline the potentially important role of free Hb in PE etiology. The damaging effects were counteracted by A1M, suggesting a role of this protein as a new potential PE therapeutic agent.
Collapse
Affiliation(s)
- K May
- Division of Obstetrics and Gynecology, Department of Clinical Sciences, Lund University Hospital, Lund University, Lund, Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Olsson MG, Nilsson EJC, Rutardóttir S, Paczesny J, Pallon J, Åkerström B. Bystander Cell Death and Stress Response is Inhibited by the Radical Scavenger α1-Microglobulin in Irradiated Cell Cultures. Radiat Res 2010; 174:590-600. [DOI: 10.1667/rr2213.1] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
40
|
Taneja S, Sen S, Gupta VK, Aggarwal R, Jameel S. Plasma and urine biomarkers in acute viral hepatitis E. Proteome Sci 2009; 7:39. [PMID: 19860894 PMCID: PMC2773234 DOI: 10.1186/1477-5956-7-39] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Accepted: 10/27/2009] [Indexed: 11/21/2022] Open
Abstract
Background Hepatitis E, caused by the hepatitis E virus (HEV), is endemic to developing countries where it manifests as waterborne outbreaks and sporadic cases. Though generally self-limited with a low mortality rate, some cases progress to fulminant hepatic failure (FHF) with high mortality. With no identified predictive or diagnostic markers, the events leading to disease exacerbation are not known. Our aim is to use proteomic tools to identify biomarkers of acute and fulminant hepatitis E. Results We analyzed proteins in the plasma and urine of hepatitis E patients and healthy controls by two-dimensional Differential Imaging Gel Electrophoresis (DIGE) and mass spectrometry, and identified over 30 proteins to be differentially expressed during acute hepatitis E. The levels of one plasma protein, transthyretin, and one urine protein, alpha-1-microglobulin (α1m), were then quantitated by enzyme immunoassay (EIA) in clinical samples from a larger group of patients and controls. The results showed decreased plasma transthyretin levels (p < 0.005) and increased urine α1m levels (p < 0.001) in acute hepatitis E patients, compared to healthy controls. Preliminary results also showed lower urine zinc alpha glycoprotein levels in fulminant hepatitis E compared to acute disease; this remains to be confirmed with more fulminant cases. Conclusion Our results demonstrate the utility of characterizing plasma and urine proteomes for signatures of the host response to HEV infection. We predict that plasma transthyretin and urine α1m could be reliable biomarkers of acute hepatitis E. Besides the utility of this approach to biomarker discovery, proteome-level changes in human biofluids would also guide towards a better understanding of host-virus interaction and disease.
Collapse
Affiliation(s)
- Shikha Taneja
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India.
| | | | | | | | | |
Collapse
|
41
|
Olsson MG, Olofsson T, Tapper H, Akerstrom B. The lipocalin alpha1-microglobulin protects erythroid K562 cells against oxidative damage induced by heme and reactive oxygen species. Free Radic Res 2008; 42:725-36. [PMID: 18712632 DOI: 10.1080/10715760802337265] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alpha(1)-microglobulin is a 26 kDa plasma and tissue glycoprotein that belongs to the lipocalin protein superfamily. Recent reports show that it is a reductase and radical scavenger and that it binds heme and has heme-degrading properties. This study has investigated the protective effects of alpha(1)-microglobulin against oxidation by heme and reactive oxygen species in the human erythroid cell line, K562. The results show that alpha(1)-microglobulin prevents intracellular oxidation and up-regulation of heme oxygenase-1 induced by heme, hydrogen peroxide and Fenton reaction-generated hydroxyl radicals in the culture medium. It also reduces the cytosol of non-oxidized cells. Endogeneous expression of alpha(1)-microglobulin was up-regulated by these oxidants and silencing of the alpha(1)-microglobulin expression increased the cytosol oxidation. alpha(1)-microglobulin also inhibited cell death caused by heme and cleared cells from bound heme. Binding of heme to alpha(1)-microglobulin increased the radical reductase activity of the protein as compared to the apo-protein. Finally, alpha(1)-microglobulin was localized mainly at the cell surface both when administered exogeneously and in non-treated cells. The results suggest that alpha(1)-microglobulin is involved in the defence against oxidative cellular injury caused by haemoglobin and heme and that the protein may employ both heme-scavenging and one-electron reduction of radicals to achieve this.
Collapse
Affiliation(s)
- Magnus G Olsson
- Division of Infection Medicine, Lund University, Lund, Sweden
| | | | | | | |
Collapse
|
42
|
Akerström B, Maghzal GJ, Winterbourn CC, Kettle AJ. The Lipocalin α1-Microglobulin Has Radical Scavenging Activity. J Biol Chem 2007; 282:31493-503. [PMID: 17766242 DOI: 10.1074/jbc.m702624200] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The lipocalin alpha(1)-microglobulin (alpha(1)m) is a 26-kDa glycoprotein present in plasma and in interstitial fluids of all tissues. The protein was recently shown to have reductase properties, reducing heme-proteins and other substrates, and was also reported to be involved in binding and scavenging of heme and tryptophan metabolites. To investigate its possible role as a reductant of organic radicals, we have studied the interaction of alpha(1)m with the synthetic radical, 2,2'-azino-bis-(3-ethylbenzthiazoline-6-sulfonic acid) (ABTS radical). The lipocalin readily reacted with the ABTS radical forming reduced ABTS. The apparent rate constant for this reaction was 6.3 +/- 2.5 x 10(3) M(-1) s(-1). A second reaction product with an intense purple color and an absorbance maximum at 550 nm was formed at a similar rate. This was shown by liquid chromatography/mass spectrometry to be derived from covalent attachment of a portion of ABTS radical to tyrosine residues on alpha(1)m. The relative yields of reduced ABTS and the purple ABTS derivative bound to alpha(1)m were approximately 2:1. Both reactions were dependent on the thiolate group of the cysteine residue in position 34 of the alpha(1)m polypeptide. Our results indicate that alpha(1)m is involved in a sequential reduction of ABTS radicals followed by trapping of these radicals by covalent attachment. In combination with the reported physiological properties of the protein, our results suggest that alpha(1)m may be a radical reductant and scavenger in vivo.
Collapse
Affiliation(s)
- Bo Akerström
- Department of Clinical Sciences, Lund University, 22184 Lund, Sweden.
| | | | | | | |
Collapse
|
43
|
Nordberg J, Allhorn M, Winqvist I, Akerström B, Olsson ML. Quantitative and qualitative evaluation of plasma and urine alpha1-microglobulin in healthy donors and patients with different haemolytic disorders and haemochromatosis. Clin Chim Acta 2007; 386:31-7. [PMID: 17706626 DOI: 10.1016/j.cca.2007.07.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2007] [Revised: 07/18/2007] [Accepted: 07/18/2007] [Indexed: 11/27/2022]
Abstract
BACKGROUND The haem-binding protein alpha(1)-microglobulin (alpha(1)m) is involved in protection against oxidative damage induced by extracellular haem/haemoglobin. A carboxy-terminally truncated form of alpha(1)m (t-alpha(1)m), formed by reactions with haemoglobin, degrades haem into a yellow-brown chromophore linked to the protein. The aim of this work was to investigate if t-alpha(1)m is present in urine from a large cohort and if urinary and plasma alpha(1)m/t-alpha(1)m concentrations are changed in patients with haemolytic disorders and haemochromatosis. METHODS Urine and blood from patients (n=20) and a control group (n=22) were investigated for alpha(1)m and t-alpha(1)m by gel electrophoresis, Western blotting and radioimmunoassay. Data were compared to clinical chemistry data and medical records. RESULTS Two thirds of all studied subjects displayed t-alpha(1)m in urine but the t-alpha(1)m/alpha(1)m ratio was not increased in patients. Instead, significantly elevated ratios were found in females compared to males. Patients with intravascular or extravascular haemolysis showed higher alpha(1)m, albumin and beta(2)-microglobulin/creatinine ratios in urine indicating glomerulo-tubular dysfunction. CONCLUSIONS The demonstration of t-alpha(1)m in urine of this cohort may be of importance in quantitative clinical chemistry. Whilst impaired kidney function due to intravascular haemolysis is well-known to occur, it is an unexpected finding in a group of patients with extravascular haemolysis.
Collapse
|