1
|
Ytterbrink C, Shubbar E, Parris TZ, Langen B, Druid M, Schüler E, Strand SE, Åkerström B, Gram M, Helou K, Forssell-Aronsson E. Effects of Recombinant α 1-Microglobulin on Early Proteomic Response in Risk Organs after Exposure to 177Lu-Octreotate. Int J Mol Sci 2024; 25:7480. [PMID: 39000587 PMCID: PMC11242497 DOI: 10.3390/ijms25137480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Recombinant α1-microglobulin (A1M) is proposed as a protector during 177Lu-octreotate treatment of neuroendocrine tumors, which is currently limited by bone marrow and renal toxicity. Co-administration of 177Lu-octreotate and A1M could result in a more effective treatment by protecting healthy tissue, but the radioprotective action of A1M is not fully understood. The aim of this study was to examine the proteomic response of kidneys and bone marrow early after 177Lu-octreotate and/or A1M administration. Mice were injected with 177Lu-octreotate and/or A1M, while control mice received saline or A1M vehicle solution. Bone marrow, kidney medulla, and kidney cortex were sampled after 24 h or 7 d. The differential protein expression was analyzed with tandem mass spectrometry. The dosimetric estimation was based on 177Lu activity in the kidney. PHLDA3 was the most prominent radiation-responsive protein in kidney tissue. In general, no statistically significant difference in the expression of radiation-related proteins was observed between the irradiated groups. Most canonical pathways were identified in bone marrow from the 177Lu-octreotate+A1M group. Altogether, a tissue-dependent proteomic response followed exposure to 177Lu-octreotate alone or together with A1M. Combining 177Lu-octreotate with A1M did not inhibit the radiation-induced protein expression early after exposure, and late effects should be further studied.
Collapse
Affiliation(s)
- Charlotte Ytterbrink
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden; (C.Y.); (E.S.); (M.D.)
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (T.Z.P.); (K.H.)
| | - Emman Shubbar
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden; (C.Y.); (E.S.); (M.D.)
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (T.Z.P.); (K.H.)
| | - Toshima Z. Parris
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (T.Z.P.); (K.H.)
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Britta Langen
- Section of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Malin Druid
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden; (C.Y.); (E.S.); (M.D.)
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (T.Z.P.); (K.H.)
| | - Emil Schüler
- Department of Radiation Physics, Division of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
| | - Sven-Erik Strand
- Department of Clinical Sciences Lund, Oncology, Lund University, 221 00 Lund, Sweden;
| | - Bo Åkerström
- Department of Clinical Sciences Lund, Infection Medicine, Lund University, 221 00 Lund, Sweden;
| | - Magnus Gram
- Department of Clinical Sciences Lund, Pediatrics, Lund University, 221 00 Lund, Sweden;
- Department of Neonatology, Skåne University Hospital, 222 42 Lund, Sweden
- Biofilms—Research Center for Biointerfaces, Department of Biomedical Science, Faculty of Health and Society, Malmö University, 205 06 Malmö, Sweden
| | - Khalil Helou
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (T.Z.P.); (K.H.)
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Eva Forssell-Aronsson
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden; (C.Y.); (E.S.); (M.D.)
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (T.Z.P.); (K.H.)
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| |
Collapse
|
2
|
Burmakin M, Gilmour PS, Gram M, Shushakova N, Sandoval RM, Molitoris BA, Larsson TE. Therapeutic α-1-microglobulin ameliorates kidney ischemia-reperfusion injury. Am J Physiol Renal Physiol 2024; 327:F103-F112. [PMID: 38779750 DOI: 10.1152/ajprenal.00067.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/12/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
α-1-Microglobulin (A1M) is a circulating glycoprotein with antioxidant, heme-binding, and mitochondrial protection properties. The investigational drug RMC-035, a modified therapeutic A1M protein, was assessed for biodistribution and pharmacological activity in a broad set of in vitro and in vivo experiments, supporting its clinical development. Efficacy and treatment posology were assessed in various models of kidney ischemia and reperfusion injury (IRI). Real-time glomerular filtration rate (GFR), functional renal biomarkers, tubular injury biomarkers (NGAL and KIM-1), and histopathology were evaluated. Fluorescently labeled RMC-035 was used to assess biodistribution. RMC-035 demonstrated consistent and reproducible kidney protection in rat IRI models as well as in a model of IRI imposed on renal impairment and in a mouse IRI model, where it reduced mortality. Its pharmacological activity was most pronounced with combined dosing pre- and post-ischemia and weaker with either pre- or post-ischemia dosing alone. RMC-035 rapidly distributed to the kidneys via glomerular filtration and selective luminal uptake by proximal tubular cells. IRI-induced expression of kidney heme oxygenase-1 was inhibited by RMC-035, consistent with its antioxidative properties. RMC-035 also dampened IRI-associated inflammation and improved mitochondrial function, as shown by tubular autofluorescence. Taken together, the efficacy of RMC-035 is congruent with its targeted mechanism(s) and biodistribution profile, supporting its further clinical evaluation as a novel kidney-protective therapy.NEW & NOTEWORTHY A therapeutic A1M protein variant (RMC-035) is currently in phase 2 clinical development for renal protection in patients undergoing open-chest cardiac surgery. It targets several key pathways underlying kidney injury in this patient group, including oxidative stress, heme toxicity, and mitochondrial dysfunction. RMC-035 is rapidly eliminated from plasma, distributing to kidney proximal tubules, and demonstrates dose-dependent efficacy in numerous models of ischemia-reperfusion injury, particularly when administered before ischemia. These results support its continued clinical evaluation.
Collapse
Affiliation(s)
- Mikhail Burmakin
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
- Guard Therapeutics International AB, Stockholm, Sweden
| | | | - Magnus Gram
- Pediatrics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Neonatology, Skåne University Hospital, Lund, Sweden
- Biofilms - Research Center for Biointerfaces, Department of Biomedical Science, Faculty of Health and Society, Malmö University, Malmö, Sweden
| | - Nelli Shushakova
- Renal Disease and Transplantation, Phenos GmbH, Hannover, Germany
- Department of Nephrology, Hannover Medical School, Hannover, Germany
| | - Ruben M Sandoval
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Bruce A Molitoris
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Tobias E Larsson
- Guard Therapeutics International AB, Stockholm, Sweden
- Division of Nephrology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
3
|
Li N, Han L, Wang X, Qiao O, Zhang L, Gong Y. Biotherapy of experimental acute kidney injury: emerging novel therapeutic strategies. Transl Res 2023; 261:69-85. [PMID: 37329950 DOI: 10.1016/j.trsl.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Acute kidney injury (AKI) is a complex and heterogeneous disease with high incidence and mortality, posing a serious threat to human life and health. Usually, in clinical practice, AKI is caused by crush injury, nephrotoxin exposure, ischemia-reperfusion injury, or sepsis. Therefore, most AKI models for pharmacological experimentation are based on this. The current research promises to develop new biological therapies, including antibody therapy, non-antibody protein therapy, cell therapy, and RNA therapy, that could help mitigate the development of AKI. These approaches can promote renal repair and improve systemic hemodynamics after renal injury by reducing oxidative stress, inflammatory response, organelles damage, and cell death, or activating cytoprotective mechanisms. However, no candidate drugs for AKI prevention or treatment have been successfully translated from bench to bedside. This article summarizes the latest progress in AKI biotherapy, focusing on potential clinical targets and novel treatment strategies that merit further investigation in future pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Ning Li
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Nankai District, Tianjin, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Lu Han
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Nankai District, Tianjin, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Xinyue Wang
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Nankai District, Tianjin, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Ou Qiao
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Nankai District, Tianjin, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Li Zhang
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Nankai District, Tianjin, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Yanhua Gong
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Nankai District, Tianjin, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China.
| |
Collapse
|
4
|
Alattar AG, Kristiansson A, Karlsson H, Vallius S, Ahlstedt J, Forssell-Aronsson E, Åkerström B, Strand SE, Flygare J, Gram M. Recombinant α 1-Microglobulin (rA1M) Protects against Hematopoietic and Renal Toxicity, Alone and in Combination with Amino Acids, in a 177Lu-DOTATATE Mouse Radiation Model. Biomolecules 2023; 13:928. [PMID: 37371508 DOI: 10.3390/biom13060928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
177Lu-DOTATATE peptide receptor radionuclide therapy (PRRT) is used clinically to treat metastasized or unresectable neuroendocrine tumors (NETs). Although 177Lu-DOTATATE is mostly well tolerated in patients, bone marrow suppression and long-term renal toxicity are still side effects that should be considered. Amino acids are often used to minimize renal radiotoxicity, however, they are associated with nausea and vomiting in patients. α1-microglobulin (A1M) is an antioxidant with heme- and radical-scavenging abilities. A recombinant form (rA1M) has previously been shown to be renoprotective in preclinical models, including in PRRT-induced kidney damage. Here, we further investigated rA1M's renal protective effect in a mouse 177Lu-DOTATATE model in terms of administration route and dosing regimen and as a combined therapy with amino acids (Vamin). Moreover, we investigated the protective effect of rA1M on peripheral blood and bone marrow cells, as well as circulatory biomarkers. Intravenous (i.v.) administration of rA1M reduced albuminuria levels and circulatory levels of the oxidative stress-related protein fibroblast growth factor-21 (FGF-21). Dual injections of rA1M (i.e., at 0 and 24 h post-177Lu-DOTATATE administration) preserved bone marrow cellularity and peripheral blood reticulocytes. Administration of Vamin, alone or in combination with rA1M, did not show any protection of bone marrow cellularity or peripheral reticulocytes. In conclusion, this study suggests that rA1M, administered i.v. for two consecutive days in conjunction with 177Lu-DOTATATE, may reduce hematopoietic and kidney toxicity during PRRT with 177Lu-DOTATATE.
Collapse
Affiliation(s)
- Abdul Ghani Alattar
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, 221 84 Lund, Sweden
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, 221 84 Lund, Sweden
| | - Amanda Kristiansson
- Pediatrics, Department of Clinical Sciences Lund, Skåne University Hospital, Lund University, 221 84 Lund, Sweden
| | - Helena Karlsson
- Pediatrics, Department of Clinical Sciences Lund, Skåne University Hospital, Lund University, 221 84 Lund, Sweden
| | - Suvi Vallius
- Pediatrics, Department of Clinical Sciences Lund, Skåne University Hospital, Lund University, 221 84 Lund, Sweden
| | - Jonas Ahlstedt
- Department of Clinical Sciences Lund, CIPA, Lund University, 221 84 Lund, Sweden
| | - Eva Forssell-Aronsson
- Department of Medical Radiation Sciences, Sahlgrenska Cancer Center, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Bo Åkerström
- Department of Clinical Sciences Lund, Section for Infection Medicine, Lund University, 221 84 Lund, Sweden
| | - Sven-Erik Strand
- Department of Clinical Sciences Lund, Oncology, Lund University, 222 42 Lund, Sweden
- Department of Clinical Sciences Lund, Medical Radiation Physics, Lund University, 221 85 Lund, Sweden
| | - Johan Flygare
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, 221 84 Lund, Sweden
| | - Magnus Gram
- Pediatrics, Department of Clinical Sciences Lund, Skåne University Hospital, Lund University, 221 84 Lund, Sweden
| |
Collapse
|
5
|
Sjögren SE, Chen J, Mattebo A, Alattar AG, Karlsson H, Siva K, Soneji S, Tedgård U, Chen JJ, Gram M, Flygare J. Targeting elevated heme levels to treat a mouse model for Diamond-Blackfan Anemia. Exp Hematol 2022; 105:50-61. [PMID: 34757171 PMCID: PMC10499113 DOI: 10.1016/j.exphem.2021.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 10/19/2021] [Accepted: 10/25/2021] [Indexed: 11/26/2022]
Abstract
Diamond-Blackfan anemia (DBA) is a rare genetic disorder in which patients present a scarcity of erythroid precursors in an otherwise normocellular bone marrow. Most, but not all, patients carry mutations in ribosomal proteins such as RPS19, suggesting that compromised mRNA translation and ribosomal stress are pathogenic mechanisms causing depletion of erythroid precursors. To gain further insight to disease mechanisms in DBA, we performed a custom short hairpin RNA (shRNA) based screen against 750 genes hypothesized to affect DBA pathophysiology. Among the hits were two shRNAs against the erythroid specific heme-regulated eIF2α kinase (HRI), which is a negative regulator of mRNA translation. This study shows that shRNA-mediated HRI silencing or loss of one HRI allele improves expansion of Rps19-deficient erythroid precursors, as well as improves the anemic phenotype in Rps19-deficient animals. We found that Rps19-deficient erythroblasts have elevated levels of unbound intracellular heme, which is normalized by HRI heterozygosity. Additionally, targeting elevated heme levels by treating cells with the heme scavenger alpha-1-microglobulin (A1M), increased proliferation of Rps19-deficient erythroid precursors and decreased heme levels in a disease-specific manner. HRI heterozygosity, but not A1M treatment, also decreased the elevated p53 activity observed in Rps19-deficient cells, indicating that p53 activation is caused by ribosomal stress and aberrant mRNA translation and not heme overload in Rps19-deficiency. Together, these findings suggest that targeting elevated heme levels is a promising new treatment strategy for DBA.
Collapse
Affiliation(s)
- Sara E Sjögren
- Department of molecular medicine and gene therapy, Lund University, Lund, Sweden; Stem cell center, Lund University, Lund, Sweden
| | - Jun Chen
- Department of molecular medicine and gene therapy, Lund University, Lund, Sweden; Stem cell center, Lund University, Lund, Sweden
| | - Alexander Mattebo
- Department of molecular medicine and gene therapy, Lund University, Lund, Sweden; Stem cell center, Lund University, Lund, Sweden
| | - Abdul G Alattar
- Department of molecular medicine and gene therapy, Lund University, Lund, Sweden; Stem cell center, Lund University, Lund, Sweden; Department of Pediatrics, Lund University, Lund, Sweden
| | - Helena Karlsson
- Department of Infection Medicine, Lund University, Lund, Sweden; Guard Therapeutics AB, Lund, Sweden
| | - Kavitha Siva
- Department of molecular medicine and gene therapy, Lund University, Lund, Sweden; Stem cell center, Lund University, Lund, Sweden
| | - Shamit Soneji
- Stem cell center, Lund University, Lund, Sweden; Division of Molecular Hematology, Lund University, Lund, Sweden
| | - Ulf Tedgård
- Department of Pediatric Hematology and Oncology, Skåne University Hospital, Lund, Sweden
| | - Jane-Jane Chen
- Institute for Biomedical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, US
| | - Magnus Gram
- Department of Infection Medicine, Lund University, Lund, Sweden; Guard Therapeutics AB, Lund, Sweden
| | - Johan Flygare
- Department of molecular medicine and gene therapy, Lund University, Lund, Sweden; Stem cell center, Lund University, Lund, Sweden.
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Bergwik J, Kristiansson A, Larsson J, Ekström S, Åkerström B, Allhorn M. Binding of the human antioxidation protein α 1-microglobulin (A1M) to heparin and heparan sulfate. Mapping of binding site, molecular and functional characterization, and co-localization in vivo and in vitro. Redox Biol 2021; 41:101892. [PMID: 33607500 PMCID: PMC7900767 DOI: 10.1016/j.redox.2021.101892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 12/27/2022] Open
Abstract
Heparin and heparan sulfate (HS) are linear sulfated disaccharide polymers. Heparin is found mainly in mast cells, while heparan sulfate is found in connective tissue, extracellular matrix and on cell membranes in most tissues. α1-microglobulin (A1M) is a ubiquitous protein with thiol-dependent antioxidant properties, protecting cells and matrix against oxidative damage due to its reductase activities and radical- and heme-binding properties. In this work, it was shown that A1M binds to heparin and HS and can be purified from human plasma by heparin affinity chromatography and size exclusion chromatography. The binding strength is inversely dependent of salt concentration and proportional to the degree of sulfation of heparin and HS. Potential heparin binding sites, located on the outside of the barrel-shaped A1M molecule, were determined using hydrogen deuterium exchange mass spectrometry (HDX-MS). Immunostaining of endothelial cells revealed pericellular co-localization of A1M and HS and the staining of A1M was almost completely abolished after treatment with heparinase. A1M and HS were also found to be co-localized in vivo in the lungs, aorta, kidneys and skin of mice. The redox-active thiol group of A1M was unaffected by the binding to HS, and the cell protection and heme-binding abilities of A1M were slightly affected. The discovery of the binding of A1M to heparin and HS provides new insights into the biological role of A1M and represents the basis for a novel method for purification of A1M from plasma.
Collapse
Affiliation(s)
- Jesper Bergwik
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden.
| | - Amanda Kristiansson
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jörgen Larsson
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Simon Ekström
- Swedish National Infrastructure for Biological Mass Spectrometry (BioMS), Lund University, Lund, Sweden
| | - Bo Åkerström
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Maria Allhorn
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
9
|
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
|
10
|
Heidari Beigvand H, Heidari K, Hashemi B, Saberinia A. The Value of Lactate Dehydrogenase in Predicting Rhabdomyolysis-Induced Acute Renal Failure; a Narrative Review. ARCHIVES OF ACADEMIC EMERGENCY MEDICINE 2021; 9:e24. [PMID: 34027419 PMCID: PMC8126348 DOI: 10.22037/aaem.v9i1.1096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Introduction Determining the diagnostic value of available biomarkers in predicting rhabdomyolysis-induced acute kidney injury (AKI) is a priority. This study aimed to review the current evidence about the value of lactate dehydrogenase (LDH) in this regard. Methods In this narrative review, the papers in PubMed, Embase, and web of science were studied. The keywords prognosis, prognoses, prognostic, LDH, rhabdomyolysis, emergency patients, and acute kidney failure or AKI had been selected from MeSH medical dictionary. Related papers written in English and published from November 2007 to December 2020 were selected. Results Finally, 14 articles were accepted for analysis. Among the selected articles, four were randomized clinical trials, seven were cross-sectional, and three were case-control studies. The results of the present review showed that abuse of illegal drugs is the most common cause of rhabdomyolysis. AKI is the most serious complication of rhabdomyolysis reported in the studies. These studies have shown a three-fold increase in AKI following drug-induced rhabdomyolysis. The review of the included articles shows that high LDH can predicts AKI, especially in critical and emergency situations such as rhabdomyolysis where there is a risk of death if diagnosed late. These studies show that LDH increases in the presence of renal failure and tissue damage. Conclusion Serum LDH is an appropriate and cost-effective prognostic indicator that can be used for risk classification of patients at risk for rhabdomyolysis-induced AKI.
Collapse
Affiliation(s)
- Hazhir Heidari Beigvand
- Preventive Medicine and Public Health Research Center, Psychosocial Health Research Institute, Community and Family Medicine Department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kamran Heidari
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behrooz Hashemi
- Emergency Department, Shohadaye Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amin Saberinia
- Emergency Department, Shohadaye Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Emergency Department, Bahonar Hospital, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
11
|
177Lu-PSMA-617 Therapy in Mice, with or without the Antioxidant α 1-Microglobulin (A1M), Including Kidney Damage Assessment Using 99mTc-MAG3 Imaging. Biomolecules 2021; 11:biom11020263. [PMID: 33579037 PMCID: PMC7916794 DOI: 10.3390/biom11020263] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 12/31/2022] Open
Abstract
Anti-prostate specific membrane antigen (PSMA) radioligand therapy is promising but not curative in castration resistant prostate cancer. One way to broaden the therapeutic index could be to administer higher doses in combination with radioprotectors, since administered radioactivity is kept low today in order to avoid side-effects from a high absorbed dose to healthy tissue. Here, we investigated the human radical scavenger α1-microglobulin (A1M) together with 177-Lutetium (177Lu) labeled PSMA-617 in preclinical models with respect to therapeutic efficacy and kidney toxicity. Nude mice with subcutaneous LNCaP xenografts were injected with 50 or 100 MBq of [177Lu]Lu-PSMA-617, with or without injections of recombinant A1M (rA1M) (at T = 0 and T = 24 h). Kidney absorbed dose was calculated to 7.36 Gy at 4 days post a 100 MBq injection. Activity distribution was imaged with Single-Photon Emission Computed Tomography (SPECT) at 24 h. Tumor volumes were measured continuously, and kidneys and blood were collected at termination (3–4 days and 3–4 weeks after injections). In a parallel set of experiments, mice were given [177Lu]Lu-PSMA-617 and rA1M as above and dynamic technetium-99m mercaptoacetyltriglycine ([99mTc]Tc-MAG3) SPECT imaging was performed prior to injection, and 3- and 6-months post injection. Blood and urine were continuously sampled. At termination (6 months) the kidneys were resected. Biomarkers of kidney function, expression of stress genes and kidney histopathology were analyzed. [177Lu]Lu-PSMA-617 uptake, in tumors and kidneys, as well as treatment efficacy did not differ between rA1M and vehicle groups. In mice given rA1M, [99mTc]Tc-MAG3 imaging revealed a significantly higher slope of initial uptake at three months compared to mice co-injected with [177Lu]Lu-PSMA-617 and vehicle. Little or no change compared to control was seen in urine albumin, serum/plasma urea levels, RT-qPCR analysis of stress response genes and in the kidney histopathological evaluation. In conclusion, [99mTc]Tc-MAG3 imaging presented itself as a sensitive tool to detect changes in kidney function revealing that administration of rA1M has a potentially positive effect on kidney perfusion and tubular function when combined with [177Lu]Lu-PSMA-617 therapy. Furthermore, we could show that rA1M did not affect anti-PSMA radioligand therapy efficacy.
Collapse
|
12
|
Kristiansson A, Bergwik J, Alattar AG, Flygare J, Gram M, Hansson SR, Olsson ML, Storry JR, Allhorn M, Åkerström B. Human radical scavenger α 1-microglobulin protects against hemolysis in vitro and α 1-microglobulin knockout mice exhibit a macrocytic anemia phenotype. Free Radic Biol Med 2021; 162:149-159. [PMID: 32092412 DOI: 10.1016/j.freeradbiomed.2020.02.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [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: 12/21/2022]
Abstract
During red blood cell (RBC) lysis hemoglobin and heme leak out of the cells and cause damage to the endothelium and nearby tissue. Protective mechanisms exist; however, these systems are not sufficient in diseases with increased extravascular hemolysis e.g. hemolytic anemia. α1-microglobulin (A1M) is a ubiquitous reductase and radical- and heme-binding protein with antioxidation properties. Although present in the circulation in micromolar concentrations, its function in blood is unclear. Here, we show that A1M provides RBC stability. A1M-/- mice display abnormal RBC morphology, reminiscent of macrocytic anemia conditions, i.e. fewer, larger and more heterogeneous cells. Recombinant human A1M (rA1M) reduced in vitro hemolysis of murine RBC against spontaneous, osmotic and heme-induced stress. Moreover, A1M is taken up by human RBCs both in vitro and in vivo. Similarly, rA1M also protected human RBCs against in vitro spontaneous, osmotic, heme- and radical-induced hemolysis as shown by significantly reduced leakage of hemoglobin and LDH. Addition of rA1M resulted in decreased hemolysis compared to addition of the heme-binding protein hemopexin and the radical-scavenging and reducing agents ascorbic acid and Trolox (vitamin E). Furthermore, rA1M significantly reduced spontaneous and heme-induced fetal RBC cell death. Addition of A1M to human whole blood resulted in a significant reduction of hemolysis, whereas removal of A1M from whole blood resulted in increased hemolysis. We conclude that A1M has a protective function in reducing hemolysis which is neither specific to the origin of hemolytic insult, nor species specific.
Collapse
Affiliation(s)
- Amanda Kristiansson
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden.
| | - Jesper Bergwik
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Abdul Ghani Alattar
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Department of Laboratory Medicine, Lund University, Lund, Sweden; Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Johan Flygare
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Magnus Gram
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Stefan R Hansson
- Department of Obstetrics and Gynecology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Martin L Olsson
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden; Department of Clinical Immunology and Transfusion Medicine, Office of Medical Services, Lund, Sweden
| | - Jill R Storry
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden; Department of Clinical Immunology and Transfusion Medicine, Office of Medical Services, Lund, Sweden
| | - Maria Allhorn
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Bo Åkerström
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
13
|
Abstract
The concept of engineering robust protein scaffolds for novel binding functions emerged 20 years ago, one decade after the advent of recombinant antibody technology. Early examples were the Affibody, Monobody (Adnectin), and Anticalin proteins, which were derived from fragments of streptococcal protein A, from the tenth type III domain of human fibronectin, and from natural lipocalin proteins, respectively. Since then, this concept has expanded considerably, including many other protein templates. In fact, engineered protein scaffolds with useful binding specificities, mostly directed against targets of biomedical relevance, constitute an area of active research today, which has yielded versatile reagents as laboratory tools. However, despite strong interest from basic science, only a handful of those protein scaffolds have undergone biopharmaceutical development up to the clinical stage. This includes the abovementioned pioneering examples as well as designed ankyrin repeat proteins (DARPins). Here we review the current state and clinical validation of these next-generation therapeutics.
Collapse
Affiliation(s)
| | - Arne Skerra
- Lehrstuhl für Biologische Chemie, Technische Universität München, 85354 Freising, Germany;
| |
Collapse
|
14
|
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
|
15
|
Romantsik O, Bruschettini M, Ley D. Intraventricular Hemorrhage and White Matter Injury in Preclinical and Clinical Studies. Neoreviews 2020; 20:e636-e652. [PMID: 31676738 DOI: 10.1542/neo.20-11-e636] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Germinal matrix-intraventricular hemorrhage (IVH) occurs in nearly half of infants born at less than 26 weeks' gestation. Up to 50% of survivors with IVH develop cerebral palsy, cognitive deficits, behavioral disorders, posthemorrhagic ventricular dilatation, or a combination of these sequelae. After the initial bleeding and the primary brain injury, inflammation and secondary brain injury might lead to periventricular leukomalacia or diffuse white matter injury. Potential factors that are involved include microglia and astrocyte activation, degradation of blood components with release of "toxic" products, infiltration of the brain by systemic immune cells, death of neuronal and glial cells, and arrest of preoligodendrocyte maturation. In addition, impairment of the blood-brain barrier may play a major role in the pathophysiology. A wide range of animal models has been used to explore causes and mechanisms leading to IVH-induced brain injury. Preclinical studies have identified potential targets for enhancing brain repair. However, little has been elucidated about the effectiveness of potential interventions in clinical studies. A systematic review of available preclinical and clinical studies might help identify research gaps and which types of interventions may be prioritized. Future trials should report clinically robust and long-term outcomes after IVH.
Collapse
Affiliation(s)
- Olga Romantsik
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skane University Hospital, Lund, Sweden
| | - Matteo Bruschettini
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skane University Hospital, Lund, Sweden
| | - David Ley
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skane University Hospital, Lund, Sweden
| |
Collapse
|
16
|
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
|
17
|
Carlsson MLR, Kristiansson A, Bergwik J, Kanagarajan S, Bülow L, Åkerström B, Zhu LH. Expression, Purification and Initial Characterization of Functional α 1-Microglobulin (A1M) in Nicotiana benthamiana. FRONTIERS IN PLANT SCIENCE 2020; 11:593773. [PMID: 33363557 PMCID: PMC7752767 DOI: 10.3389/fpls.2020.593773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/12/2020] [Indexed: 05/08/2023]
Abstract
α1-Microglobulin (A1M) is a small glycoprotein that belongs to the lipocalin protein family. A major biological role of A1M is to protect cells and tissues against oxidative damage by clearing free heme and reactive oxygen species. Because of this, the protein has attracted great interest as a potential pharmaceutical candidate for treatment of acute kidney injury and preeclampsia. The aim of this study was to explore the possibility of expressing human A1M in plants through transient gene expression, as an alternative or complement to other expression systems. E. coli, insect and mammalian cell culture have previously been used for recombinant A1M (rA1M) or A1M production, but these systems have various drawbacks, including additional complication and expense in refolding for E. coli, while insect produced rA1M is heavily modified with chromophores and mammalian cell culture has been used only in analytical scale. For that purpose, we have used a viral vector (pJL-TRBO) delivered by Agrobacterium for expression of three modified A1M gene variants in the leaves of N. benthamiana. The results showed that these modified rA1M protein variants, A1M-NB1, A1M-NB2 and A1M-NB3, targeted to the cytosol, ER and extracellular space, respectively, were successfully expressed in the leaves, which was confirmed by SDS-PAGE and Western blot analysis. The cytosol accumulated A1M-NB1 was selected for further analysis, as it appeared to have a higher yield than the other variants, and was purified with a yield of ca. 50 mg/kg leaf. The purified protein had the expected structural and functional properties, displaying heme-binding capacity and capacity of protecting red blood cells against stress-induced cell death. The protein also carried bound chromophores, a characteristic feature of A1M and an indicator of a capacity to bind small molecules. The study showed that expression of the functional protein in N. benthamiana may be an attractive alternative for production of rA1M for pharmaceutical purposes and a basis for future research on A1M structure and function.
Collapse
Affiliation(s)
- Magnus L. R. Carlsson
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Alnarp, Sweden
| | - Amanda Kristiansson
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jesper Bergwik
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Selvaraju Kanagarajan
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Alnarp, Sweden
| | - Leif Bülow
- Division of Pure and Applied Biochemistry, Department of Chemistry, Lund University, Lund, Sweden
| | - Bo Åkerström
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Li-Hua Zhu
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Alnarp, Sweden
- *Correspondence: Li-Hua Zhu,
| |
Collapse
|
18
|
Romantsik O, Agyemang AA, Sveinsdóttir S, Rutardóttir S, Holmqvist B, Cinthio M, Mörgelin M, Gumus G, Karlsson H, Hansson SR, Åkerström B, Ley D, Gram M. The heme and radical scavenger α 1-microglobulin (A1M) confers early protection of the immature brain following preterm intraventricular hemorrhage. J Neuroinflammation 2019; 16:122. [PMID: 31174551 PMCID: PMC6554963 DOI: 10.1186/s12974-019-1486-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/25/2019] [Indexed: 12/31/2022] Open
Abstract
Background Germinal matrix intraventricular hemorrhage (GM-IVH) is associated with cerebro-cerebellar damage in very preterm infants, leading to neurodevelopmental impairment. Penetration, from the intraventricular space, of extravasated red blood cells and extracellular hemoglobin (Hb), to the periventricular parenchyma and the cerebellum has been shown to be causal in the development of brain injury following GM-IVH. Furthermore, the damage has been described to be associated with the cytotoxic nature of extracellular Hb-metabolites. To date, there is no therapy available to prevent infants from developing either hydrocephalus or serious neurological disability. Mechanisms previously described to cause brain damage following GM-IVH, i.e., oxidative stress and Hb-metabolite toxicity, suggest that the free radical and heme scavenger α1-microglobulin (A1M) may constitute a potential neuroprotective intervention. Methods Using a preterm rabbit pup model of IVH, where IVH was induced shortly after birth in pups delivered by cesarean section at E29 (3 days prior to term), we investigated the brain distribution of recombinant A1M (rA1M) following intracerebroventricular (i.c.v.) administration at 24 h post-IVH induction. Further, short-term functional protection of i.c.v.-administered human A1M (hA1M) following IVH in the preterm rabbit pup model was evaluated. Results Following i.c.v. administration, rA1M was distributed in periventricular white matter regions, throughout the fore- and midbrain and extending to the cerebellum. The regional distribution of rA1M was accompanied by a high co-existence of positive staining for extracellular Hb. Administration of i.c.v.-injected hA1M was associated with decreased structural tissue and mitochondrial damage and with reduced mRNA expression for proinflammatory and inflammatory signaling-related genes induced by IVH in periventricular brain tissue. Conclusions The results of this study indicate that rA1M/hA1M is a potential candidate for neuroprotective treatment following preterm IVH. Electronic supplementary material The online version of this article (10.1186/s12974-019-1486-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Olga Romantsik
- Pediatrics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | | | | | | | | | - Magnus Cinthio
- Department of Electrical Measurements, Lund University, Lund, Sweden
| | - Mattias Mörgelin
- Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Gulcin Gumus
- Fetal i+D Fetal Medicine Research Center, BCNatal Barcelona Center for Maternal-Fetal and Neonatal Medicine, University of Barcelona, Barcelona, Spain
| | | | - Stefan R Hansson
- Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Bo Åkerström
- Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - David Ley
- Pediatrics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Magnus Gram
- Pediatrics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden. .,Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden. .,A1M Pharma AB, Lund, Sweden.
| |
Collapse
|
19
|
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
|
20
|
Grimes S, Bombay K, Lanes A, Walker M, Corsi DJ. Potential biological therapies for severe preeclampsia: a systematic review and meta-analysis. BMC Pregnancy Childbirth 2019; 19:163. [PMID: 31072315 PMCID: PMC6509856 DOI: 10.1186/s12884-019-2268-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/27/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Preeclampsia remains a significant danger to both mother and child and current prevention and treatment management strategies are limited. The objective of this systematic review was to investigate the current literature on evidence for the use of the regenerative capacity of mesenchymal stem cell (MSC) therapy, the anticoagulant activity of antithrombin (AT), or the free radical scavenging activity of alpha-1-microglobulin (A1M) as potential novel treatments for severe preeclampsia and Hemolysis, Elevated Liver enzymes, Low Platelet count (HELLP). METHOD We conducted a systematic review of potential biological therapies for preeclampsia. We screened MEDLINE and Embase from inception through May 2017 for studies using AT, A1M or MSCs as potential treatments for preeclampsia and/or HELLP. A meta-analysis was performed to pool data from randomized control trials (RCTs) with homogenous outcomes using the inverse variance method. The Newcastle-Ottawa Scale, the Cochrane risk of bias tool for RCTs, and SYRCLE's risk of bias tool for animal studies were used to investigate potential bias of studies. RESULTS The literature search retrieved a total of 1015 articles, however, only 17 studies met the selection criteria: AT (n = 9, 8 human and 1 animal); A1M (n = 4, 3 animal and 1 ex-vivo); and, MSCs (n = 4, 3 animal and 1 ex-vivo). A meta-analysis of AT therapy versus placebo and a meta-analysis for AT therapy with heparin versus heparin alone did not show significant differences between study groups. Animal and ex-vivo studies demonstrated significant benefits in relevant outcomes for A1M and MSCs versus control treatments. Most RCT studies were rated as having a low risk of bias across categories with some studies showing an unclear risk of bias in some categories. The two cohort studies both received a total of four out of nine stars (a rating of "poor" quality). Most animal studies had an unclear risk of bias across most categories, with some studies having a low risk of bias in some categories. CONCLUSIONS The findings of this review are strengthened by rigorous systematic search and review of the literature. Results of our meta-analyses do not currently warrant further exploration of AT as a treatment of preeclampsia in human trials. Results of animal and ex-vivo studies of A1M and MSCs were encouraging and supportive of initiating human investigations.
Collapse
Affiliation(s)
- Sophia Grimes
- OMNI Research Group, Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario Canada
| | - Kira Bombay
- OMNI Research Group, Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario Canada
| | - Andrea Lanes
- OMNI Research Group, Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario Canada
- School of Epidemiology, Public Health and Preventive Medicine, University of Ottawa, Ottawa, Ontario Canada
| | - Mark Walker
- OMNI Research Group, Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario Canada
- School of Epidemiology, Public Health and Preventive Medicine, University of Ottawa, Ottawa, Ontario Canada
- Department of Obstetrics, Gynecology & Newborn Care, University of Ottawa, Ottawa, Ontario Canada
| | - Daniel J. Corsi
- OMNI Research Group, Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario Canada
- School of Epidemiology, Public Health and Preventive Medicine, University of Ottawa, Ottawa, Ontario Canada
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, Ontario Canada
- OMNI Research Group, Centre for Practice Changing Research, Ottawa Hospital Research Institute, L1242, Box 241, 501 Smyth Road, Ottawa, ON K1H 8L6 Canada
| |
Collapse
|
21
|
Li Z, Zhou G, Jiang L, Xiang H, Cao Y. Effect of STOX1 on recurrent spontaneous abortion by regulating trophoblast cell proliferation and migration via the PI3K/AKT signaling pathway. J Cell Biochem 2019; 120:8291-8299. [PMID: 30548667 PMCID: PMC6590170 DOI: 10.1002/jcb.28112] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/29/2018] [Indexed: 01/24/2023]
Abstract
STOX1 is a transcription factor that is implicated in the high prevalence of human gestational diseases. It has been studied in various types of gestational diseases using different molecular and cellular biological technologies. However, the effect and detailed mechanism of storkhead box 1 (STOX1) in recurrent spontaneous abortion (RSA) remain unknown. This study aimed to explore the effect and detailed mechanism of STOX1 in human trophoblast cells. The result showed that downregulation of STOX1 by short hairpin RNA (shRNA) led to a decrease in proliferation and migration in HTR-8/SVneo cells, while it induced the apoptosis of HTR-8/SVneo cells. Moreover, the result showed that trophoblast cells expressed lower levels of pAKT and p85 subunits after treatment with STOX1 shRNA when compared with control. However, overexpression of STOX1 obviously increased the pAKT and p85 protein expressions. Transfection of pcDNA-AKT plasmid increased cell proliferation and migration in HTR-8/SVneo cells while suppressed the apoptosis of HTR-8/SVneo cells. Furthermore, inhibition of the PI3K/Akt pathway by a specific inhibitor promoted cell apoptosis and aggravatedly suppressed cell proliferation and migration of HTR-8/SVneo cells. On the other hand, upregulation of the PI3K/Akt pathway could increase the relative expression level of Bcl-2 and decrease the relative expression levels of Bax and Bim, while inhibition of the PI3K/Akt pathway led to adverse results. Our results demonstrated that inhibition of STOX1 could suppress trophoblast cell proliferation and migration, while promote apoptosis through inhibiting the PI3K/Akt signaling pathway. These findings might provide a new fundamental mechanism for regulating RSA and could be used to prevent and treat RSA in clinic.
Collapse
Affiliation(s)
- Zhifang Li
- Reproductive Medicine Center, The First Affiliated Hospital, Anhui Medical UniversityHefeiChina,Anqing Municipal Hospital, Anhui Medical UniversityAnqingChina
| | - Guiju Zhou
- Reproductive Medicine Center, The First Affiliated Hospital, Anhui Medical UniversityHefeiChina,Department Gynecology, The Second Affiliated Hospital, Anhui Medical UniversityHefeiChina
| | - Longfan Jiang
- Anqing Municipal Hospital, Anhui Medical UniversityAnqingChina
| | - Huifen Xiang
- Reproductive Medicine Center, The First Affiliated Hospital, Anhui Medical UniversityHefeiChina
| | - Yunxia Cao
- Reproductive Medicine Center, The First Affiliated Hospital, Anhui Medical UniversityHefeiChina
| |
Collapse
|