1
|
Ruminski PG, Rettig MP, DiPersio JF. Development of VLA4 and CXCR4 Antagonists for the Mobilization of Hematopoietic Stem and Progenitor Cells. Biomolecules 2024; 14:1003. [PMID: 39199390 PMCID: PMC11353233 DOI: 10.3390/biom14081003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 09/01/2024] Open
Abstract
The treatment of patients diagnosed with hematologic malignancies typically includes hematopoietic stem cell transplantation (HSCT) as part of a therapeutic standard of care. The primary graft source of hematopoietic stem and progenitor cells (HSPCs) for HSCT is mobilized from the bone marrow into the peripheral blood of allogeneic donors or patients. More recently, these mobilized HSPCs have also been the source for gene editing strategies to treat diseases such as sickle-cell anemia. For a HSCT to be successful, it requires the infusion of a sufficient number of HSPCs that are capable of adequate homing to the bone marrow niche and the subsequent regeneration of stable trilineage hematopoiesis in a timely manner. Granulocyte-colony-stimulating factor (G-CSF) is currently the most frequently used agent for HSPC mobilization. However, it requires five or more daily infusions to produce an adequate number of HSPCs and the use of G-CSF alone often results in suboptimal stem cell yields in a significant number of patients. Furthermore, there are several undesirable side effects associated with G-CSF, and it is contraindicated for use in sickle-cell anemia patients, where it has been linked to serious vaso-occlusive and thrombotic events. The chemokine receptor CXCR4 and the cell surface integrin α4β1 (very late antigen 4 (VLA4)) are both involved in the homing and retention of HSPCs within the bone marrow microenvironment. Preclinical and/or clinical studies have shown that targeted disruption of the interaction of the CXCR4 or VLA4 receptors with their endogenous ligands within the bone marrow niche results in the rapid and reversible mobilization of HSPCs into the peripheral circulation and is synergistic when combined with G-CSF. In this review, we discuss the roles CXCR4 and VLA4 play in bone marrow homing and retention and will summarize more recent development of small-molecule CXCR4 and VLA4 inhibitors that, when combined, can synergistically improve the magnitude, quality and convenience of HSPC mobilization for stem cell transplantation and ex vivo gene therapy after the administration of just a single dose. This optimized regimen has the potential to afford a superior alternative to G-CSF for HSPC mobilization.
Collapse
Affiliation(s)
| | | | - John F. DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Ave., St Louis, MO 63105, USA
| |
Collapse
|
2
|
Chen K, Zhu P, Chu M, Tao H, Wang Q, Lv S, Huang L, Geng D. What do osteoporosis and osteoarthritis have in common? An integrated study of overlapping differentially expressed genes in bone mesenchymal stem cells of osteoporosis and osteoarthritis. Gene 2024; 893:147914. [PMID: 37865148 DOI: 10.1016/j.gene.2023.147914] [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: 07/13/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
OBJECTIVE For identification of aberrantly expressed genes in mesenchymal stem cells of osteoporosis (OP) and osteoarthritis (OA), Gene Expression Omnibus (GEO) datasets were integrated to investigate the intersection point. METHODS GSE35958 (osteoporosis) and GSE19664 (osteoarthritis) datasets were obtained from GEO database. The abnormally expressed genes were analyzed by GEO2R. Functional enrichment was explored by Metascape database and R software. The String database and Cytoscape software were used to build the protein-protein interaction network and identify hub genes. GSE35957 and GSE116925 were used as verification datasets. Single-cell analysis and pseudotime analysis were undertaken. CTDbase, Network Analyst, HPA database, HERB database and MIRW database were used to research the information, tissue and cell distribution, regulation, interaction and ingredients targeting the hub genes. Additionally, in vitro experiments such as RT-PCR, ALP staining and immunofluorescence were undertaken as verification tests. RESULTS Ten hub genes were identified in this study. All these genes play an important role in bone or cartilage generation. They have diagnostic values and therapeutic potential for OA and OP. Single-cell analysis visualized the cell distribution and pseudotime distribution of these genes. Some potential therapeutic ingredients of these genes were identified, such as curcumin, wogonin and glycerin. In vitro experiments, RT-PCR results showed that COL9A3 and MMP3 were downregulated and PTH1R was upregulated during osteogenic induction of BMSC. Immunohistochemical results showed the expression trend of MMP3 and COL2A1. CONCLUSION Ten abnormal hub genes of osteoporosis and osteoarthritis were identified successfully by this study. They were important regulatory genes for healthy bone and cartilage. These genes could be the common connections between osteoporosis and osteoarthritis as well as treatment targets. Further study of the regulatory mechanism and treatment effects of these genes would be valuable. The results of this study could contribute to further research.
Collapse
Affiliation(s)
- Kai Chen
- Department of Orthopedics, Hai'an People's Hospital, Zhongba Road 17, Hai'an, Jiangsu 226600, China; Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu 215000, China
| | - Pengfei Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu 215000, China
| | - Miao Chu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu 215000, China; Department of Orthopedics, Yixing People's Hospital, Tongzhenguan Road 75, Yixing, Jiangsu 214200, China
| | - Huaqiang Tao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu 215000, China
| | - Qiufei Wang
- Department of Orthopedics, Changshu First People's Hospital, Shuyuan Road 1, Changshu, Jiangsu 215500, China
| | - Shujun Lv
- Department of Orthopedics, Hai'an People's Hospital, Zhongba Road 17, Hai'an, Jiangsu 226600, China.
| | - Lixin Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu 215000, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu 215000, China.
| |
Collapse
|
3
|
Barresi V, Di Bella V, Lo Nigro L, Privitera AP, Bonaccorso P, Scuderi C, Condorelli DF. Temporary serine protease inhibition and the role of SPINK2 in human bone marrow. iScience 2023; 26:106949. [PMID: 37378330 PMCID: PMC10291479 DOI: 10.1016/j.isci.2023.106949] [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: 11/11/2022] [Revised: 03/23/2023] [Accepted: 05/20/2023] [Indexed: 06/29/2023] Open
Abstract
Protease temporary inhibitors are true substrates that bind the catalytic site with high affinity but are slowly degraded, thus acting as inhibitor for a defined time window. Serine peptidase inhibitor Kazal type (SPINK) family is endowed with such functional property whose physiological meaning is poorly explored. High expression of SPINK2 in some hematopoietic malignancies prompted us to investigate its role in adult human bone marrow. We report here the physiological expression of SPINK2 in hematopoietic stem and progenitor cells (HSPCs) and mobilized cluster differentiation 34 (CD34)+ cells. We determined the SPINK2 degradation constant and derived a mathematical relationship predicting the zone of inhibited target protease activity surrounding the SPINK2-secreting HSPCs. Analysis of putative target proteases for SPINK2 revealed the expression of PRSS2 and PRSS57 in HSPCs. Our combined results suggest that SPINK2 and its target serine proteases might play a role in the intercellular communication within the hematopoietic stem cell niche.
Collapse
Affiliation(s)
- Vincenza Barresi
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, 95123 Catania, Italy
| | - Virginia Di Bella
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, 95123 Catania, Italy
| | - Luca Lo Nigro
- Cytogenetic-Cytofluorimetric-Molecular Biology Lab, 95123 Catania, Italy
- Center of Pediatric Hematology-Oncology, Azienda Policlinico – San Marco, 95123 Catania, Italy
| | - Anna Provvidenza Privitera
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, 95123 Catania, Italy
| | - Paola Bonaccorso
- Cytogenetic-Cytofluorimetric-Molecular Biology Lab, 95123 Catania, Italy
- Center of Pediatric Hematology-Oncology, Azienda Policlinico – San Marco, 95123 Catania, Italy
| | - Chiara Scuderi
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, 95123 Catania, Italy
| | - Daniele Filippo Condorelli
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, 95123 Catania, Italy
| |
Collapse
|
4
|
Recombinant Alpha-1 Antitrypsin as Dry Powder for Pulmonary Administration: A Formulative Proof of Concept. Pharmaceutics 2022; 14:pharmaceutics14122754. [PMID: 36559248 PMCID: PMC9784676 DOI: 10.3390/pharmaceutics14122754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Alpha-1 antitrypsin (AAT) deficiency is a genetic disorder associated with pulmonary emphysema and bronchiectasis. Its management currently consists of weekly infusions of plasma-purified human AAT, which poses several issues regarding plasma supplies, possible pathogen transmission, purification costs, and parenteral administration. Here, we investigated an alternative administration strategy for augmentation therapy by combining recombinant expression of AAT in bacteria and the production of a respirable powder by spray drying. The same formulation approach was then applied to plasma-derived AAT for comparison. Purified, active, and endotoxin-free recombinant AAT was produced at high yields and formulated using L-leucine and mannitol as excipients after identifying compromise conditions for protein activity and good aerodynamic performances. An oxygen-free atmosphere, both during formulation and powder storage, slowed down methionine-specific oxidation and AAT inactivation. This work is the first peer-reviewed report of AAT formulated as a dry powder, which could represent an alternative to current treatments.
Collapse
|
5
|
Zhao J, Feng C, Wang W, Su L, Jiao J. Human SERPINA3 induces neocortical folding and improves cognitive ability in mice. Cell Discov 2022; 8:124. [DOI: 10.1038/s41421-022-00469-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022] Open
Abstract
AbstractNeocortex expansion and folding are related to human intelligence and cognition, but the molecular and cellular mechanisms underlying cortical folding remain poorly understood. Here, we report that the human gene SERPINA3 is linked to gyrification. Specifically, the overexpression of SERPINA3 induced neocortical folding, increased the abundance of neurons, and improved cognitive abilities. Further, SERPINA3 promoted proliferation of the outer radial glia (oRG, also referred to as the basal radial glia) and increased the number of upper-layer neurons. The downstream target Glo1 was determined to be involved in SERPINA3-induced gyrification. Moreover, SERPINA3 increased the proliferation of oRG by binding to the Glo1 promoter. Assessment of behavior performance showed enhanced cognitive abilities in SERPINA3 knock-in mice. Our findings will enrich the understanding of neocortical expansion and gyrification and provide insights into possible treatments for intellectual disability and lissencephaly syndrome.
Collapse
|
6
|
Isojima T, Walker EC, Poulton IJ, McGregor NE, Wicks IP, Gooi JH, Martin TJ, Sims NA. G-CSF Receptor Deletion Amplifies Cortical Bone Dysfunction in Mice With STAT3 Hyperactivation in Osteocytes. J Bone Miner Res 2022; 37:1876-1890. [PMID: 35856245 DOI: 10.1002/jbmr.4654] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/27/2022] [Accepted: 07/16/2022] [Indexed: 11/09/2022]
Abstract
Bone strength is determined by the structure and composition of its thickened outer shell (cortical bone), yet the mechanisms controlling cortical consolidation are poorly understood. Cortical bone maturation depends on SOCS3-mediated suppression of IL-6 cytokine-induced STAT3 phosphorylation in osteocytes, the cellular network embedded in bone matrix. Because SOCS3 also suppresses granulocyte-colony-stimulating factor receptor (G-CSFR) signaling, we here tested whether global G-CSFR (Csf3r) ablation altereed bone structure in male and female mice lacking SOCS3 in osteocytes, (Dmp1Cre :Socs3f/f mice). Dmp1Cre :Socs3f/f :Csf3r-/- mice were generated by crossing Dmp1Cre :Socs3f/f mice with Csf3r-/- mice. Although G-CSFR is not expressed in osteocytes, Csf3r deletion further delayed cortical consolidation in Dmp1Cre :Socs3f/f mice. Micro-CT images revealed extensive, highly porous low-density bone, with little true cortex in the diaphysis, even at 26 weeks of age; including more low-density bone and less high-density bone in Dmp1Cre :Socs3f/f :Csf3r-/- mice than controls. By histology, the area where cortical bone would normally be found contained immature compressed trabecular bone in Dmp1Cre :Socs3f/f :Csf3r-/- mice and greater than normal levels of intracortical osteoclasts, extensive new woven bone formation, and the presence of more intracortical blood vessels than the already high levels observed in Dmp1Cre :Socs3f/f controls. qRT-PCR of cortical bone from Dmp1Cre :Socs3f/f :Csf3r-/- mice also showed more than a doubling of mRNA levels for osteoclasts, osteoblasts, RANKL, and angiogenesis markers. The further delay in cortical bone maturation was associated with significantly more phospho-STAT1 and phospho-STAT3-positive osteocytes, and a threefold increase in STAT1 and STAT3 target gene mRNA levels, suggesting G-CSFR deletion further increases STAT signaling beyond that of Dmp1Cre :Socs3f/f bone. G-CSFR deficiency therefore promotes STAT1/3 signaling in osteocytes, and when SOCS3 negative feedback is absent, elevated local angiogenesis, bone resorption, and bone formation delays cortical bone consolidation. This points to a critical role of G-CSF in replacing condensed trabecular bone with lamellar bone during cortical bone formation. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Tsuyoshi Isojima
- St. Vincent's Institute of Medical Research, Fitzroy, Australia.,Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan
| | - Emma C Walker
- St. Vincent's Institute of Medical Research, Fitzroy, Australia
| | | | | | - Ian P Wicks
- Walter and Eliza Hall Institute, Parkville, Australia
| | - Jonathan H Gooi
- St. Vincent's Institute of Medical Research, Fitzroy, Australia.,Bio21 Molecular Science and Biotechnology Institute, Parkville, Australia
| | - T John Martin
- St. Vincent's Institute of Medical Research, Fitzroy, Australia.,The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Australia
| | - Natalie A Sims
- St. Vincent's Institute of Medical Research, Fitzroy, Australia.,The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Australia
| |
Collapse
|
7
|
Quezado ZMN, Kamimura S, Smith M, Wang X, Heaven MR, Jana S, Vogel S, Zerfas P, Combs CA, Almeida LEF, Li Q, Quezado M, Horkayne-Szakaly I, Kosinski PA, Yu S, Kapadnis U, Kung C, Dang L, Wakim P, Eaton WA, Alayash AI, Thein SL. Mitapivat increases ATP and decreases oxidative stress and erythrocyte mitochondria retention in a SCD mouse model. Blood Cells Mol Dis 2022; 95:102660. [PMID: 35366607 DOI: 10.1016/j.bcmd.2022.102660] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 11/25/2022]
Abstract
Polymerization of deoxygenated sickle hemoglobin (HbS) leads to erythrocyte sickling. Enhancing activity of the erythrocyte glycolytic pathway has anti-sickling potential as this reduces 2,3-diphosphoglycerate (2,3-DPG) and increases ATP, factors that decrease HbS polymerization and improve erythrocyte membrane integrity. These factors can be modulated by mitapivat, which activates erythrocyte pyruvate kinase (PKR) and improves sickling kinetics in SCD patients. We investigated mechanisms by which mitapivat may impact SCD by examining its effects in the Townes SCD mouse model. Control (HbAA) and sickle (HbSS) mice were treated with mitapivat or vehicle. Surprisingly, HbSS had higher PKR protein, higher ATP, and lower 2,3-DPG levels, compared to HbAA mice, in contrast with humans with SCD, in whom 2,3-DPG is elevated compared to healthy subjects. Despite our inability to investigate 2,3-DPG-mediated sickling and hemoglobin effects, mitapivat yielded potential benefits in HbSS mice. Mitapivat further increased ATP without significantly changing 2,3-DPG or hemoglobin levels, and decreased levels of leukocytosis, erythrocyte oxidative stress, and the percentage of erythrocytes that retained mitochondria in HbSS mice. These data suggest that, even though Townes HbSS mice have increased PKR activity, further activation of PKR with mitapivat yields potentially beneficial effects that are independent of changes in sickling or hemoglobin levels.
Collapse
Affiliation(s)
- Zenaide M N Quezado
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA; Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Sayuri Kamimura
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Meghann Smith
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xunde Wang
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael R Heaven
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, MD 20993, USA
| | - Sirsendu Jana
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, MD 20993, USA
| | - Sebastian Vogel
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Patricia Zerfas
- Office of Research Services, Office of the Director, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christian A Combs
- Light Microscopy Core, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Luis E F Almeida
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Quan Li
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Martha Quezado
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Iren Horkayne-Szakaly
- Neuropathology and Ophthalmic Pathology, Joint Pathology Center, Defense Health Agency, Silver Spring, MD 20910, USA
| | | | - Shaoxia Yu
- Agios Pharmaceuticals Inc, Cambridge, MA 02139, USA
| | | | - Charles Kung
- Agios Pharmaceuticals Inc, Cambridge, MA 02139, USA
| | - Lenny Dang
- Agios Pharmaceuticals Inc, Cambridge, MA 02139, USA
| | - Paul Wakim
- Biostatistics and Clinical Epidemiology Service, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - William A Eaton
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Abdu I Alayash
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, MD 20993, USA
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
8
|
Plasma Proteomic Analysis Identified Proteins Associated with Faulty Neutrophils Functionality in Decompensated Cirrhosis Patients with Sepsis. Cells 2022; 11:cells11111745. [PMID: 35681439 PMCID: PMC9179303 DOI: 10.3390/cells11111745] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/17/2022] [Accepted: 04/20/2022] [Indexed: 02/04/2023] Open
Abstract
Decompensated cirrhosis (DC) is susceptible to infections and sepsis. Neutrophils and monocytes provide the first line of defense to encounter infection. We aimed to evaluate proteins related to neutrophils functionality in sepsis. 70 (DC), 40 with sepsis, 30 without (w/o) sepsis and 15 healthy controls (HC) plasma was analyzed for proteomic analysis, cytokine bead array, endotoxin, cell free DNA and whole blood cells were analyzed for nCD64-mHLADR index, neutrophils-monocytes, functionality and QRT-PCR. nCD64-mHLADR index was significantly increased (p < 0.0001) with decreased HLA-DR expression on total monocytes in sepsis (p = 0.045). Phagocytic activity of both neutrophils and monocytes were significantly decreased in sepsis (p = 0.002 and p = 0.0003). Sepsis plasma stimulated healthy neutrophils, showed significant increase in NETs (neutrophil extracellular traps) and cell free DNA (p = 0.049 and p = 0.04) compared to w/o sepsis and HC. Proteomic analysis revealed upregulated- DNAJC13, TMSB4X, GPI, GSTP1, PNP, ANPEP, COTL1, GCA, APOA1 and PGAM1 while downregulated- AHSG, DEFA1,SERPINA3, MPO, MMRN1and PROS1 proteins (FC > 1.5; p < 0.05) associated to neutrophil activation and autophagy in sepsis. Proteins such as DNAJC13, GPI, GSTP1, PNP, ANPEP, COTL1, PGAM1, PROS1, MPO, SERPINA3 and MMRN1 showed positive correlation with neutrophils activity and number, oxidative burst activity and clinical parameters such as MELD, MELD Na and Bilirubin. Proteomic analysis revealed that faulty functionality of neutrophils may be due to the autophagy proteins i.e., DNAJC13, AHSG, TMSB4X, PROS1 and SERPINA3, which can be used as therapeutic targets in decompensated cirrhosis patients with sepsis.
Collapse
|
9
|
Lévesque JP, Summers KM, Millard SM, Bisht K, Winkler IG, Pettit AR. Role of macrophages and phagocytes in orchestrating normal and pathologic hematopoietic niches. Exp Hematol 2021; 100:12-31.e1. [PMID: 34298116 DOI: 10.1016/j.exphem.2021.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 12/13/2022]
Abstract
The bone marrow (BM) contains a mosaic of niches specialized in supporting different maturity stages of hematopoietic stem and progenitor cells such as hematopoietic stem cells and myeloid, lymphoid, and erythroid progenitors. Recent advances in BM imaging and conditional gene knockout mice have revealed that niches are a complex network of cells of mesenchymal, endothelial, neuronal, and hematopoietic origins, together with local physicochemical parameters. Within these complex structures, phagocytes, such as neutrophils, macrophages, and dendritic cells, all of which are of hematopoietic origin, have been found to be important in regulating several niches in the BM, including hematopoietic stem cell niches, erythropoietic niches, and niches involved in endosteal bone formation. There is also increasing evidence that these macrophages have an important role in adapting hematopoiesis, erythropoiesis, and bone formation in response to inflammatory stressors and play a key part in maintaining the integrity and function of these. Likewise, there is also accumulating evidence that subsets of monocytes, macrophages, and other phagocytes contribute to the progression and response to treatment of several lymphoid malignancies such as multiple myeloma, Hodgkin lymphoma, and non-Hodgkin lymphoma, as well as lymphoblastic leukemia, and may also play a role in myelodysplastic syndrome and myeloproliferative neoplasms associated with Noonan syndrome and aplastic anemia. In this review, the potential functions of macrophages and other phagocytes in normal and pathologic niches are discussed, as are the challenges in studying BM and other tissue-resident macrophages at the molecular level.
Collapse
Affiliation(s)
- Jean-Pierre Lévesque
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia.
| | - Kim M Summers
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| | - Susan M Millard
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| | - Kavita Bisht
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| | - Ingrid G Winkler
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| | - Allison R Pettit
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| |
Collapse
|
10
|
Skerrett-Byrne DA, Trigg NA, Bromfield EG, Dun MD, Bernstein IR, Anderson AL, Stanger SJ, MacDougall LA, Lord T, Aitken RJ, Roman SD, Robertson SA, Nixon B, Schjenken JE. Proteomic Dissection of the Impact of Environmental Exposures on Mouse Seminal Vesicle Function. Mol Cell Proteomics 2021; 20:100107. [PMID: 34089863 PMCID: PMC8250459 DOI: 10.1016/j.mcpro.2021.100107] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/19/2021] [Accepted: 05/28/2021] [Indexed: 12/15/2022] Open
Abstract
Seminal vesicles are an integral part of the male reproductive accessory gland system. They produce a complex array of secretions containing bioactive constituents that support gamete function and promote reproductive success, with emerging evidence suggesting these secretions are influenced by our environment. Despite their significance, the biology of seminal vesicles remains poorly defined. Here, we complete the first proteomic assessment of mouse seminal vesicles and assess the impact of the reproductive toxicant acrylamide. Mice were administered acrylamide (25 mg/kg bw/day) or control daily for five consecutive days prior to collecting seminal vesicle tissue. A total of 5013 proteins were identified in the seminal vesicle proteome with bioinformatic analyses identifying cell proliferation, protein synthesis, cellular death, and survival pathways as prominent biological processes. Secreted proteins were among the most abundant, and several proteins are linked with seminal vesicle phenotypes. Analysis of the effect of acrylamide on the seminal vesicle proteome revealed 311 differentially regulated (FC ± 1.5, p ≤ 0.05, 205 up-regulated, 106 downregulated) proteins, orthogonally validated via immunoblotting and immunohistochemistry. Pathways that initiate protein synthesis to promote cellular survival were prominent among the dysregulated pathways, and rapamycin-insensitive companion of mTOR (RICTOR, p = 6.69E-07) was a top-ranked upstream driver. Oxidative stress was implicated as contributing to protein changes, with acrylamide causing an increase in 8-OHdG in seminal vesicle epithelial cells (fivefold increase, p = 0.016) and the surrounding smooth muscle layer (twofold increase, p = 0.043). Additionally, acrylamide treatment caused a reduction in seminal vesicle secretion weight (36% reduction, p = 0.009) and total protein content (25% reduction, p = 0.017). Together these findings support the interpretation that toxicant exposure influences male accessory gland physiology and highlights the need to consider the response of all male reproductive tract tissues when interpreting the impact of environmental stressors on male reproductive function.
Collapse
Affiliation(s)
- David A Skerrett-Byrne
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Natalie A Trigg
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Elizabeth G Bromfield
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Matthew D Dun
- Cancer Signalling Research Group, Faculty of Health and Medicine, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Priority Research Centre for Cancer Research Innovation and Translation, Hunter Medical Research Institute, Lambton, NSW, Australia
| | - Ilana R Bernstein
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Amanda L Anderson
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Simone J Stanger
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Lily A MacDougall
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Tessa Lord
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - R John Aitken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Shaun D Roman
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Sarah A Robertson
- The Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - John E Schjenken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia; Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| |
Collapse
|
11
|
Ratajczak MZ, Adamiak M, Ratajczak J, Kucia M. Heme Oxygenase 1 (HO-1) as an Inhibitor of Trafficking of Normal and Malignant Hematopoietic Stem Cells - Clinical and Translational Implications. Stem Cell Rev Rep 2021; 17:821-828. [PMID: 33196976 PMCID: PMC8166705 DOI: 10.1007/s12015-020-10083-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2020] [Indexed: 12/12/2022]
Abstract
Evidence indicates that bone marrow (BM)-residing hematopoietic stem/progenitor cells (HSPCs) are released into peripheral blood (PB) after administration of pro-mobilizing drugs, which induce a state of sterile inflammation in the BM microenvironment. In the reverse process, as seen after hematopoietic transplantation, intravenously injected HSPCs home and engraft into BM niches. Here again, conditioning for transplantation by myeloablative chemo- or radiotherapy induces a state of sterile inflammation that promotes HSPC seeding to BM stem cell niches. Therefore, the trafficking of HSPCs and their progeny, including granulocytes and monocytes/macrophages, is regulated by a response to pro-inflammatory stimuli. This responsiveness to inflammatory cues is also preserved after malignant transformation of hematopoietic cells. Results from our laboratory indicate that the responsiveness of hematopoietic cells to pro-inflammatory stimuli is orchestrated by Nlrp3 inflammasome. As reported, HO-1 effectively attenuates intracellular activation of Nlrp3 inflammasome as well as the pro-inflammatory effects of several humoral mediators, including complement cascade (ComC) cleavage fragments that promote migration of hematopoietic cells. Based on this finding, inhibition of HO-1 activity may become a practical strategy to enhance the mobilization and homing of normal HSPCs, and, alternatively, its activation may prevent unwanted spread and in vivo expansion of leukemic cells. Graphical Abstract.
Collapse
Affiliation(s)
- Mariusz Z. Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Mateusz Adamiak
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
| | - Magda Kucia
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
12
|
Bianchera A, Alomari E, Bruno S. Augmentation therapy with alpha 1-antitrypsin: present and future of production, formulation, and delivery. Curr Med Chem 2021; 29:385-410. [PMID: 34036902 DOI: 10.2174/0929867328666210525161942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/24/2021] [Accepted: 04/12/2021] [Indexed: 11/22/2022]
Abstract
Alpha 1-antitrypsin is one of the first protein therapeutics introduced on the market - more than 30 years ago - and, to date, it is indicated only for the treatment of the severe forms of a genetic condition known as alpha-1 antitrypsin deficiency. The only approved preparations are derived from plasma, posing potential problems associated with its limited supply and high processing costs. Moreover, augmentation therapy with alpha 1-antitrypsin is still limited to intravenous infusions, a cumbersome regimen for patients. Here, we review the recent literature on its possible future developments, focusing on i) the recombinant alternatives to the plasma-derived protein, ii) novel formulations, and iii) novel administration routes. Regulatory issues and the still unclear noncanonical functions of alpha 1-antitrypsin - possibly associated with the glycosylation pattern found only in the plasma-derived protein - have hindered the introduction of new products. However, potentially new therapeutic indications other than the treatment of alpha-1 antitrypsin deficiency might open the way to new sources and new formulations.
Collapse
Affiliation(s)
- Annalisa Bianchera
- Dipartimento di Scienze degli Alimenti e del Farmaco, University of Parma, Parma, Italy
| | - Esraa Alomari
- Dipartimento di Scienze degli Alimenti e del Farmaco, University of Parma, Parma, Italy
| | - Stefano Bruno
- Dipartimento di Scienze degli Alimenti e del Farmaco, University of Parma, Parma, Italy
| |
Collapse
|
13
|
Kandarakov OF, Kravatsky YV, Polyakova NS, Bruter AV, Gordeeva EG, Belyavsky AV. Mitomycin C Treatment of Stromal Layers Enhances the Support of In Vitro Hematopoiesis in Co-Culture Systems. Mol Biol 2021. [DOI: 10.1134/s0026893321010088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
14
|
Chlis NK, Rausch L, Brocker T, Kranich J, Theis FJ. Predicting single-cell gene expression profiles of imaging flow cytometry data with machine learning. Nucleic Acids Res 2020; 48:11335-11346. [PMID: 33119742 PMCID: PMC7672460 DOI: 10.1093/nar/gkaa926] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 08/24/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023] Open
Abstract
High-content imaging and single-cell genomics are two of the most prominent high-throughput technologies for studying cellular properties and functions at scale. Recent studies have demonstrated that information in large imaging datasets can be used to estimate gene mutations and to predict the cell-cycle state and the cellular decision making directly from cellular morphology. Thus, high-throughput imaging methodologies, such as imaging flow cytometry can potentially aim beyond simple sorting of cell-populations. We introduce IFC-seq, a machine learning methodology for predicting the expression profile of every cell in an imaging flow cytometry experiment. Since it is to-date unfeasible to observe single-cell gene expression and morphology in flow, we integrate uncoupled imaging data with an independent transcriptomics dataset by leveraging common surface markers. We demonstrate that IFC-seq successfully models gene expression of a moderate number of key gene-markers for two independent imaging flow cytometry datasets: (i) human blood mononuclear cells and (ii) mouse myeloid progenitor cells. In the case of mouse myeloid progenitor cells IFC-seq can predict gene expression directly from brightfield images in a label-free manner, using a convolutional neural network. The proposed method promises to add gene expression information to existing and new imaging flow cytometry datasets, at no additional cost.
Collapse
Affiliation(s)
- Nikolaos-Kosmas Chlis
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg 85764, Germany.,Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg 82377, Germany
| | - Lisa Rausch
- Institute for Immunology, Medical Faculty, Ludwig Maximilian University of Munich, 82152 Planegg-Martinsried, Germany
| | - Thomas Brocker
- Institute for Immunology, Medical Faculty, Ludwig Maximilian University of Munich, 82152 Planegg-Martinsried, Germany
| | - Jan Kranich
- Institute for Immunology, Medical Faculty, Ludwig Maximilian University of Munich, 82152 Planegg-Martinsried, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg 85764, Germany.,Department of Mathematics, Technical University of Munich, Garching 85748, Germany
| |
Collapse
|
15
|
Walker EC, Truong K, McGregor NE, Poulton IJ, Isojima T, Gooi JH, Martin TJ, Sims NA. Cortical bone maturation in mice requires SOCS3 suppression of gp130/STAT3 signalling in osteocytes. eLife 2020; 9:e56666. [PMID: 32458800 PMCID: PMC7253175 DOI: 10.7554/elife.56666] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/01/2020] [Indexed: 12/23/2022] Open
Abstract
Bone strength is determined by its dense cortical shell, generated by unknown mechanisms. Here we use the Dmp1Cre:Socs3f/f mouse, with delayed cortical bone consolidation, to characterise cortical maturation and identify control signals. We show that cortical maturation requires a reduction in cortical porosity, and a transition from low to high density bone, which continues even after cortical shape is established. Both processes were delayed in Dmp1Cre:Socs3f/f mice. SOCS3 (suppressor of cytokine signalling 3) inhibits signalling by leptin, G-CSF, and IL-6 family cytokines (gp130). In Dmp1Cre:Socs3f/f bone, STAT3 phosphorylation was prolonged in response to gp130-signalling cytokines, but not G-CSF or leptin. Deletion of gp130 in Dmp1Cre:Socs3f/f mice suppressed STAT3 phosphorylation in osteocytes and osteoclastic resorption within cortical bone, leading to rescue of the corticalisation defect, and restoration of compromised bone strength. We conclude that cortical bone development includes both pore closure and accumulation of high density bone, and that these processes require suppression of gp130-STAT3 signalling in osteocytes.
Collapse
Affiliation(s)
- Emma C Walker
- St. Vincent’s Institute of Medical ResearchFitzroyAustralia
| | - Kim Truong
- St. Vincent’s Institute of Medical ResearchFitzroyAustralia
- University of Melbourne, Department of Medicine at St. Vincent’s HospitalFitzroyAustralia
| | | | | | - Tsuyoshi Isojima
- St. Vincent’s Institute of Medical ResearchFitzroyAustralia
- Department of Pediatrics, Teikyo University School of MedicineTokyoJapan
| | - Jonathan H Gooi
- St. Vincent’s Institute of Medical ResearchFitzroyAustralia
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of MelbourneParkvilleAustralia
| | - T John Martin
- St. Vincent’s Institute of Medical ResearchFitzroyAustralia
- University of Melbourne, Department of Medicine at St. Vincent’s HospitalFitzroyAustralia
| | - Natalie A Sims
- St. Vincent’s Institute of Medical ResearchFitzroyAustralia
- University of Melbourne, Department of Medicine at St. Vincent’s HospitalFitzroyAustralia
| |
Collapse
|
16
|
Mai B, Friscia M, Wahed A, Nguyen A, Chen L. The novel coexistence of SERPINA1 and JAK2 V617F mutations in alpha-1 antitrypsin deficiency. Pathology 2020; 52:382-385. [PMID: 32115143 DOI: 10.1016/j.pathol.2019.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/25/2019] [Accepted: 12/01/2019] [Indexed: 11/30/2022]
Affiliation(s)
- Brenda Mai
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Michaelangelo Friscia
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Amer Wahed
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Andy Nguyen
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Lei Chen
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, TX, USA.
| |
Collapse
|
17
|
Starodubtseva N, Nizyaeva N, Baev O, Bugrova A, Gapaeva M, Muminova K, Kononikhin A, Frankevich V, Nikolaev E, Sukhikh G. SERPINA1 Peptides in Urine as A Potential Marker of Preeclampsia Severity. Int J Mol Sci 2020; 21:E914. [PMID: 32019243 PMCID: PMC7037458 DOI: 10.3390/ijms21030914] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/18/2022] Open
Abstract
Preeclampsia (PE) is a multisystem disorder associated with pregnancy and its frequency varies from 5 to 20 percent of pregnancies. Although a number of preeclampsia studies have been carried out, there is no consensus about disease etiology and pathogenesis so far. Peptides of SERPINA1 (α1-antitrypsin) in urine remain one of the most promising peptide markers of PE. In this study the diagnostic potential of urinary α1-antitrypsin peptides in PE was evaluated. The urinary peptidome composition of 79 pregnant women with preeclampsia (PE), chronic arterial hypertension (CAH), and a control group was investigated. Mann-Whitney U-test (p < 0.05) revealed seven PE specific SERPINA1 peptides demonstrating 52% sensitivity and 100% specificity. SERPINA1 in urine has been associated with the most severe forms of preeclampsia (p = 0.014), in terms of systolic hypertension (p = 0.01) and proteinuria (p = 0.006). According to Spearman correlation analysis, the normalized intensity of SERPINA1 urinary peptides has a similar diagnostic pattern with known diagnostic PE markers, such as sFLT/PLGF. SERPINA1 peptides were not urinary excreted in superimposed PE (PE with CAH), which is a milder form of PE. An increase in expression of SERPINA1 in the structural elements of the placenta during preeclampsia reflects a protective mechanism against hypoxia. Increased synthesis of SERPINA1 in the trophoblast leads to protein accumulation in fibrinoid deposits. It may block syncytial knots and placenta villi, decreasing trophoblast invasion. Excretion of PE specific SERPINA1 peptides is associated with syncytiotrophoblast membrane destruction degradation and increased SERPINA1 staining. It confirms that the placenta could be the origin of SERPINA1 peptides in urine. Significant correlation (p < 0.05) of SERPINA1 expression in syncytiotrophoblast membrane and cytoplasm with the main clinical parameters of severe PE proves the role of SERPINA1 in PE pathogenesis. Estimation of SERPINA1 peptides in urine can be used as a diagnostic test of the severity of the condition to determine further treatment, particularly the need for urgent surgical delivery.
Collapse
Affiliation(s)
- Natalia Starodubtseva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (N.S.); (N.N.); (O.B.); (A.B.); (M.G.); (K.M.); (A.K.); (G.S.)
- Moscow Institute of Physics and Technology, 141701 Moscow, Russia
| | - Natalia Nizyaeva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (N.S.); (N.N.); (O.B.); (A.B.); (M.G.); (K.M.); (A.K.); (G.S.)
| | - Oleg Baev
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (N.S.); (N.N.); (O.B.); (A.B.); (M.G.); (K.M.); (A.K.); (G.S.)
| | - Anna Bugrova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (N.S.); (N.N.); (O.B.); (A.B.); (M.G.); (K.M.); (A.K.); (G.S.)
- Emanuel Institute for Biochemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Masara Gapaeva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (N.S.); (N.N.); (O.B.); (A.B.); (M.G.); (K.M.); (A.K.); (G.S.)
| | - Kamilla Muminova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (N.S.); (N.N.); (O.B.); (A.B.); (M.G.); (K.M.); (A.K.); (G.S.)
| | - Alexey Kononikhin
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (N.S.); (N.N.); (O.B.); (A.B.); (M.G.); (K.M.); (A.K.); (G.S.)
- Skolkovo Institute of Science and Technology, Skolkovo, 121205 Moscow, Russia
| | - Vladimir Frankevich
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (N.S.); (N.N.); (O.B.); (A.B.); (M.G.); (K.M.); (A.K.); (G.S.)
| | - Eugene Nikolaev
- Skolkovo Institute of Science and Technology, Skolkovo, 121205 Moscow, Russia
- V.L. Talrose Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Gennady Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (N.S.); (N.N.); (O.B.); (A.B.); (M.G.); (K.M.); (A.K.); (G.S.)
- First Moscow State Medical University Named after I.M. Sechenov, 119146 Moscow, Russia
| |
Collapse
|
18
|
Abstract
Enforced egress of hematopoietic stem cells (HSCs) out of the bone marrow (BM) into the peripheral circulation, termed mobilization, has come a long way since its discovery over four decades ago. Mobilization research continues to be driven by the need to optimize the regimen currently available in the clinic with regard to pharmacokinetic and pharmacodynamic profile, costs, and donor convenience. In this review, we describe the most recent findings in the field and how we anticipate them to affect the development of mobilization strategies in the future. Furthermore, the significance of mobilization beyond HSC collection, i.e. for chemosensitization, conditioning, and gene therapy as well as a means to study the interactions between HSCs and their BM microenvironment, is reviewed. Open questions, controversies, and the potential impact of recent technical progress on mobilization research are also highlighted.
Collapse
Affiliation(s)
- Darja Karpova
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, 69120, Germany
| | - Michael P Rettig
- Division of Oncology, Department of Medicine, Washington University School of Medicine,, St. Louis, Missouri, 63110, USA
| | - John F DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine,, St. Louis, Missouri, 63110, USA
| |
Collapse
|
19
|
Serum Proteomic Analysis Reveals Vitamin D-Binding Protein (VDBP) as a Potential Biomarker for Low Bone Mineral Density in Mexican Postmenopausal Women. Nutrients 2019; 11:nu11122853. [PMID: 31766436 PMCID: PMC6950314 DOI: 10.3390/nu11122853] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/15/2019] [Accepted: 11/16/2019] [Indexed: 12/16/2022] Open
Abstract
Osteoporosis is a skeletal disease mainly affecting women over 50 years old and it represents a serious public health problem because of the high socioeconomic burden. This disease is characterized by deterioration of bone microarchitecture, low bone mineral density (BMD), and increased risk of fragility fractures. This study aimed to identify serum useful proteins as biomarkers for the diagnosis and/or prognosis of osteoporosis and fracture risk. We collected 446 serum samples from postmenopausal women aged ≥45 years old. Based on the BMD measurement, we classified the participants into three groups: osteoporotic, osteopenic, and normal. In an initial discovery stage, we conducted a proteomic approach using two-dimensional differential gel electrophoresis (2D-DIGE). The peptides into the spots of interest were identified through matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF/TOF). Enzyme-linked immunosorbent assay (ELISA) was performed to validate the proteins of interest. We identified 27 spots of interest when comparing low BMD versus normal BMD postmenopausal women. Based on their relevance in bone metabolism, we analyzed three proteins: ceruloplasmin (CP), gelsolin (GSN), and vitamin D-binding protein (VDBP). Our results demonstrated that low serum VDBP levels correlate with low BMD (osteopenic and osteoporotic). Therefore, VDBP could be considered as a novel, potential, and non-invasive biomarker for the early detection of osteoporosis.
Collapse
|
20
|
Zhang L, Zhang J, Ye Z, Manevich Y, Townsend DM, Marshall DT, Tew KD. S-Glutathionylated Serine Proteinase Inhibitors as Biomarkers for Radiation Exposure in Prostate Cancer Patients. Sci Rep 2019; 9:13792. [PMID: 31551460 PMCID: PMC6760651 DOI: 10.1038/s41598-019-50288-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 08/30/2019] [Indexed: 12/27/2022] Open
Abstract
In biological tissues, radiation causes the formation of reactive oxygen species (ROS), some of which lead to sequential oxidation of certain protein cysteine residues. Resultant cysteinyl radicals are subject to post-translational modification through S-glutathionylation. The present clinical trial was designed to determine if S-glutathionylated serine protease inhibitors (serpins) in blood could be used as biomarkers of exposure to radiation. 56 male prostate cancer patients treated with radiotherapy were enrolled in the trial and levels of S-glutathionylated serpins A1 and A3 were assessed by immunoblotting. Patients were classified into three groups: (1) external beam radiation therapy (EBRT); (2) brachytherapy (BT); (3) both EBRT and BT. Prior to treatment, baseline plasma levels of both unmodified and S-glutathionylated serpins were similar in each group. We identified elevated plasma levels of S-glutathionylated serpin A1 monomer, trimer and serpin A3 monomer in patient blood following radiation. Maximal increased levels of these S-glutathionylated serpins were correlated with increased duration of radiotherapy treatments. We conclude that it is practical to quantify patient plasma S-glutathionylated serpins and that these post-translationally modified proteins are candidate biomarkers for measuring radiation exposure. This provides a platform for use of such biomarkers in trials with the range of drugs that, like radiation, produce ROS.
Collapse
Affiliation(s)
- Leilei Zhang
- Departments of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jie Zhang
- Departments of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Zhiwei Ye
- Departments of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Yefim Manevich
- Departments of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Danyelle M Townsend
- Departments of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - David T Marshall
- Departments of Radiation Oncology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kenneth D Tew
- Departments of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA.
| |
Collapse
|
21
|
de Kruijf EJFM, Fibbe WE, van Pel M. Cytokine-induced hematopoietic stem and progenitor cell mobilization: unraveling interactions between stem cells and their niche. Ann N Y Acad Sci 2019; 1466:24-38. [PMID: 31006885 PMCID: PMC7217176 DOI: 10.1111/nyas.14059] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/15/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023]
Abstract
Peripheral blood hematopoietic stem and progenitor cells (HSPCs), mobilized by granulocyte colony‐stimulating factor, are widely used as a source for both autologous and allogeneic stem cell transplantation. The use of mobilized HSPCs has several advantages over traditional bone marrow–derived HSPCs, including a less invasive harvesting process for the donor, higher HSPC yields, and faster hematopoietic reconstitution in the recipient. For years, the mechanisms by which cytokines and other agents mobilize HSPCs from the bone marrow were not fully understood. The field of stem cell mobilization research has advanced significantly over the past decade, with major breakthroughs in the elucidation of the complex mechanisms that underlie stem cell mobilization. In this review, we provide an overview of the events that underlie HSPC mobilization and address the relevant cellular and molecular components of the bone marrow niche. Furthermore, current and future mobilizing agents will be discussed.
Collapse
Affiliation(s)
- Evert-Jan F M de Kruijf
- Section of Stem Cell Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Willem E Fibbe
- Section of Stem Cell Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Melissa van Pel
- Section of Stem Cell Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
22
|
Periasamy P, Tran V, O’Neill HC. Identification of genes which regulate stroma-dependent in vitro hematopoiesis. PLoS One 2018; 13:e0205583. [PMID: 30308055 PMCID: PMC6181386 DOI: 10.1371/journal.pone.0205583] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 09/27/2018] [Indexed: 12/22/2022] Open
Abstract
Cultured splenic stroma has been shown to support in vitro hematopoiesis in overlaid bone marrow and spleen progenitors. These co-cultures support longterm production of a novel dendritic-like cell type along with transient production of myeloid cells. They also maintain a progenitor cell population. The splenic stromal lines 5G3 and 3B5 have been identified as a supporter and a non-supporter of hematopoiesis. Based on their gene expression profile, both 5G3 and 3B5 express genes related to hematopoiesis, while 5G3 cells express several unique genes, and show upregulation of some genes over 3B5. Based on gene expression studies, specific inhibitors were tested for capacity to inhibit hematopoiesis in co-cultures. Addition of specific antibodies and small molecule inhibitors identified VCAM1, CXCL12, CSF1 and SPP1 as potential regulators of hematopoiesis, although both are expressed by 5G3 and 3B5. Through inhibition of function, SVEP1 and ALDH1 are also shown here to be deterministic of 5G3 hematopoietic support capacity, since these are uniquely expressed by 5G3 and not 3B5. The achievement of inhibition is notable given the dynamic, longterm nature of co-cultures which involve only small numbers of cells. The alternate plan, to add recombinant soluble factors produced by 5G3 back into 3B5 co-cultures in order to recover in vitro hematopoiesis, proved ineffective. Out of 6 different factors added to 3B5, only IGF2 showed any effect on cell production. The identification of differentially expressed or upregulated genes in 5G3 has provided an insight into potential pathways involved in in vitro hematopoiesis leading to production of dendritic-like cells.
Collapse
Affiliation(s)
- Pravin Periasamy
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Vinson Tran
- Division of Biomedical Science, Research School of Biology, The Australian National University, Canberra, Australia
| | - Helen C. O’Neill
- Clem Jones Centre for Regenerative Medicine, Faculty of Health Sciences and Medicine, Bond University, Gold Coast, Queensland, Australia
- * E-mail:
| |
Collapse
|
23
|
Shamji MH, Temblay JN, Cheng W, Byrne SM, Macfarlane E, Switzer AR, Francisco NDC, Olexandra F, Jacubczik F, Durham SR, Ashton-Rickardt PG. Antiapoptotic serine protease inhibitors contribute to survival of allergenic T H2 cells. J Allergy Clin Immunol 2018; 142:569-581.e5. [PMID: 29106998 PMCID: PMC5920800 DOI: 10.1016/j.jaci.2017.07.055] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 07/07/2017] [Accepted: 07/20/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND The mechanisms that regulate maintenance of persistent TH2 cells and potentiate allergic inflammation are not well understood. OBJECTIVE The function of serine protease inhibitor 2A (Spi2A) was studied in mouse TH2 cells, and the serine protease inhibitor B3 (SERPINB3) and SERPINB4 genes were studied in TH2 cells from patients with grass pollen allergy. METHODS Spi2A-deficient TH2 cells were studied in in vitro culture or in vivo after challenge of Spi2A knockout mice with ovalbumin in alum. Expression of SERPINB3 and SERPINB4 mRNA was measured in in vitro-cultured TH2 cells and in ex vivo CD27-CD4+ cells and innate lymphoid cell (ILC) 2 from patients with grass pollen allergy by using quantitative PCR. SERPINB3 and SERPINB4 mRNA levels were knocked down in cultured CD27-CD4+ cells with small hairpin RNA. RESULTS There were lower levels of in vitro-polarized TH2 cells from Spi2A knockout mice (P < .005) and in vivo after ovalbumin challenge (P < .05), higher levels of apoptosis (Annexin V positivity, P < .005), and less lung allergic inflammation (number of lung eosinophils, P < .005). In vitro-polarized TH2 cells from patients with grass pollen allergy expressed higher levels of both SERPINB3 and SERPINB4 mRNA (both P < .05) compared with unpolarized CD4 T cells. CD27-CD4+ from patients with grass pollen allergy expressed higher levels of both SERPINB3 and SERPINB4 mRNA (both P < .0005) compared with CD27+CD4+ cells. ILC2 expressed higher levels of both SERPINB3 and SERPINB4 mRNA (both P < .0005) compared with ILC1. Knockdown of either SERPINB3 or SERPINB4 mRNA (both P < .005) levels resulted in decreased viability of CD27-CD4+ compared with control transduced cells. CONCLUSION The Serpins Spi2A in mice and SERPINB3 and SERPINB4 in allergic patients control the viability of TH2 cells. This provides proof of principle for a therapeutic approach for allergic disease through ablation of allergic memory TH2 cells through SERPINB3 and SERPINB4 mRNA downregulation.
Collapse
Affiliation(s)
- Mohamed H Shamji
- Immunomodulation and Tolerance Group, London, United Kingdom; Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, and the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Jeff N Temblay
- Section of Immunobiology, Division of Inflammation and Immunology, Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Wei Cheng
- Section of Immunobiology, Division of Inflammation and Immunology, Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Susan M Byrne
- Section of Immunobiology, Division of Inflammation and Immunology, Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Ellen Macfarlane
- Immunomodulation and Tolerance Group, London, United Kingdom; Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, and the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Amy R Switzer
- Immunomodulation and Tolerance Group, London, United Kingdom; Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, and the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Natalia D C Francisco
- Immunomodulation and Tolerance Group, London, United Kingdom; Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, and the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | | | - Fabian Jacubczik
- Section of Immunobiology, Division of Inflammation and Immunology, Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Stephen R Durham
- Immunomodulation and Tolerance Group, London, United Kingdom; Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, and the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Philip G Ashton-Rickardt
- Section of Immunobiology, Division of Inflammation and Immunology, Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom.
| |
Collapse
|
24
|
de Kruijf EJF, Zuijderduijn R, Stip MC, Fibbe WE, van Pel M. Mesenchymal stromal cells induce a permissive state in the bone marrow that enhances G-CSF-induced hematopoietic stem cell mobilization in mice. Exp Hematol 2018; 64:59-70.e2. [DOI: 10.1016/j.exphem.2018.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/17/2018] [Accepted: 05/08/2018] [Indexed: 01/05/2023]
|
25
|
Shin EK, Kim DY, Rah WJ, Koh H, Suh JY, Chang M, Nam E, Lee YH. Differences in Mobilization Efficiency between Small Children and Adults with Healthy Marrows. CLINICAL PEDIATRIC HEMATOLOGY-ONCOLOGY 2017. [DOI: 10.15264/cpho.2017.24.2.130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Eun-kyung Shin
- Department of Pediatrics, Hanyang University College of Medicine, Hanyang University Medical Center, Seoul, Korea
| | - Do-Yeon Kim
- Department of Pediatrics, Hanyang University College of Medicine, Hanyang University Medical Center, Seoul, Korea
| | - Wee-Jin Rah
- Department of Pediatrics, Hanyang University College of Medicine, Hanyang University Medical Center, Seoul, Korea
| | - Hani Koh
- Department of Pediatrics, Hanyang University College of Medicine, Hanyang University Medical Center, Seoul, Korea
- Blood and Marrow Transplantation Center, Hanyang University College of Medicine, Hanyang University Medical Center, Seoul, Korea
| | - Jin Young Suh
- Blood and Marrow Transplantation Center, Hanyang University College of Medicine, Hanyang University Medical Center, Seoul, Korea
| | - Misoo Chang
- Biostatistical Consulting and Research Lab, Hanyang University College of Medicine, Hanyang University Medical Center, Seoul, Korea
| | - Eunwoo Nam
- Biostatistical Consulting and Research Lab, Hanyang University College of Medicine, Hanyang University Medical Center, Seoul, Korea
| | - Young-Ho Lee
- Department of Pediatrics, Hanyang University College of Medicine, Hanyang University Medical Center, Seoul, Korea
- Blood and Marrow Transplantation Center, Hanyang University College of Medicine, Hanyang University Medical Center, Seoul, Korea
| |
Collapse
|
26
|
Richter R, Forssmann W, Henschler R. Current Developments in Mobilization of Hematopoietic Stem and Progenitor Cells and Their Interaction with Niches in Bone Marrow. Transfus Med Hemother 2017. [PMID: 28626366 DOI: 10.1159/000477262] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The clinical application of hematopoietic stem and progenitor cells (HSPCs) has evolved from a highly experimental stage in the 1980s to a currently clinically established treatment for more than 20,000 patients annually who suffer from hematological malignancies and other severe diseases. Studies in numerous murine models have demonstrated that HSPCs reside in distinct niches within the bone marrow environment. Whereas transplanted HSPCs travel through the bloodstream and home to sites of hematopoiesis, HSPCs can be mobilized from these niches into the blood either physiologically or induced by pharmaceutical drugs. Firstly, this review aims to give a synopsis of milestones defining niches and mobilization pathways for HSPCs, including the identification of several cell types involved such as osteoblasts, adventitial reticular cells, endothelial cells, monocytic cells, and granulocytic cells. The main factors that anchor HSPCs in the niche, and/or induce their quiescence are vascular cell adhesion molecule(VCAM)-1, CD44, hematopoietic growth factors, e.g. stem cell factor (SCF) and FLT3 Ligand, chemokines including CXCL12, growth-regulated protein beta and IL-8, proteases, peptides, and other chemical transmitters such as nucleotides. In the second part of the review, we revise the current understanding of HSPC mobilization. Here, we discuss which mechanisms found to be active in HSPC mobilization correspond to the mechanisms relevant for HSPC interaction with niche cells, but also deal with other mediators and signals that target individual cell types and receptors to mobilize HSPCs. A multitude of questions remain to be addressed for a better understanding of HSPC biology and its implications for therapy, including more comprehensive concepts for regulatory circuits such as calcium homeostasis and parathormone, metabolic regulation such as by leptin, the significance of autonomic nervous system, the consequences of alteration of niches in aged patients, or the identification of more easily accessible markers to better predict the efficiency of HSPC mobilization.
Collapse
Affiliation(s)
- Rudolf Richter
- Department of Internal Medicine, Clinic of Immunology, Hanover Medical School, Hanover, Germany.,MVZ Labor PD Dr. Volkmann & Kollegen, Karlsruhe, Germany
| | - Wolfgang Forssmann
- Department of Internal Medicine, Clinic of Immunology, Hanover Medical School, Hanover, Germany
| | - Reinhard Henschler
- Swiss Red Cross Blood Transfusion Services Zurich and Chur, Zurich, Switzerland
| |
Collapse
|
27
|
Zhang Y, He J, Zhao J, Xu M, Lou D, Tso P, Li Z, Li X. Effect of ApoA4 on SERPINA3 mediated by nuclear receptors NR4A1 and NR1D1 in hepatocytes. Biochem Biophys Res Commun 2017; 487:327-332. [PMID: 28412351 DOI: 10.1016/j.bbrc.2017.04.058] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 04/12/2017] [Indexed: 12/29/2022]
Abstract
ApoA4 exerts anti-inflammatory effects, but the mechanism remains unclear. SERPINA3 is a member of the serine proteinase inhibitor gene family, and has been shown to be involved in anti-inflammation and associated with a number of human diseases. In this study, we revealed that ApoA4 stimulates the gene expression of SERPINA3 in mouse hepatocytes both in vivo and in vitro, in a dose- and time-dependent manner. The transcriptional response of SERPINA3 to ApoA4 is regulated through the binding of ApoA4 with nuclear receptors NR4A1 and NR1D1 on the SERPINA3 promoter, which was verified with ChIP, Luciferase activity assay and RNA interference-mediated NR4A1 or NR1D1 gene knockdown. These data suggests that ApoA4 transcriptionally induced SERPINA3 expression via NR1D1 and NR4A1. Our findings may throw light on the function of ApoA4 in inflammatory responses and acute-phase reactions, as well as the development of SERPINA3 relative diseases.
Collapse
Affiliation(s)
- Yupeng Zhang
- National Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Jing He
- College of Medicine and Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Jing Zhao
- College of Medicine and Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Min Xu
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, USA
| | - Danwen Lou
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, USA
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, USA
| | - Zongfang Li
- National Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoming Li
- National Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
28
|
Cellular players of hematopoietic stem cell mobilization in the bone marrow niche. Int J Hematol 2016; 105:129-140. [PMID: 27943116 DOI: 10.1007/s12185-016-2162-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 11/21/2016] [Accepted: 11/29/2016] [Indexed: 12/23/2022]
Abstract
Hematopoietic stem cells (HSC) reside in perivascular regions of the bone marrow (BM) embedded within a complex regulatory unit called the niche. Cellular components of HSC niches include vascular endothelial cells, mesenchymal stromal progenitor cells and a variety of mature hematopoietic cells such as macrophages, neutrophils, and megakaryocytes-further regulated by sympathetic nerves and complement components as described in this review. Three decades ago the discovery that cytokines induce a large number of HSC to mobilize from the BM into the blood where they are easily harvested, revolutionised the field of HSC transplantation-curative for immune-deficiencies and some malignancies. However, despite now routine use of granulocyte-colony stimulating factor (G-CSF) to mobilise HSC for transplant, only in last 15 years has research on the mechanisms behind why and how HSC can be induced to move into the blood began. These studies have revealed the complexity of the niche that retains HSC in the BM. This review describes how BM niches and HSC themselves change during administration of G-CSF-or in the recovery phase of chemotherapy-to facilitate movement of HSC into the blood, and research now leading to development of novel therapeutics to further boost HSC mobilization and transplant success.
Collapse
|
29
|
Bendall L. Extracellular molecules in hematopoietic stem cell mobilisation. Int J Hematol 2016; 105:118-128. [PMID: 27826715 DOI: 10.1007/s12185-016-2123-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 11/01/2016] [Indexed: 01/11/2023]
Abstract
Hematopoietic stem cells are a remarkable resource currently used for the life saving treatment, hematopoietic stem cell transplantation. Today, hematopoietic stem cells are primarily obtained from mobilized peripheral blood following treatment of the donor with the cytokine G-CSF, and in some settings, chemotherapy and/or the CXCR4 antagonist plerixafor. The collection of hematopoietic stem cells is contingent on adequate and timely mobilization of these cells into the peripheral blood. The use of healthy donors, particularly when unrelated to the patient, requires mobilization strategies be safe for the donor. While current mobilization strategies are largely successful, adequate mobilization fails to occur in a significant portion of donors. Understanding the mechanisms involved in the egress of stem cells from the bone marrow provides opportunities to further improve the process of collecting hematopoietic stem cells. Here, the role extracellular components of the blood and bone marrow in the mobilization process are discussed.
Collapse
Affiliation(s)
- Linda Bendall
- Centre for Cancer Research, Westmead Institute for Medical Research, University of Sydney, 176 Hawkesbury Rd, Westmead, Sydney, NSW, 2145, Australia.
| |
Collapse
|
30
|
Walker EC, Johnson RW, Hu Y, Brennan HJ, Poulton IJ, Zhang JG, Jenkins BJ, Smyth GK, Nicola NA, Sims NA. Murine Oncostatin M Acts via Leukemia Inhibitory Factor Receptor to Phosphorylate Signal Transducer and Activator of Transcription 3 (STAT3) but Not STAT1, an Effect That Protects Bone Mass. J Biol Chem 2016; 291:21703-21716. [PMID: 27539849 DOI: 10.1074/jbc.m116.748483] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/15/2016] [Indexed: 12/31/2022] Open
Abstract
Oncostatin M (OSM) and leukemia inhibitory factor (LIF) are IL-6 family members with a wide range of biological functions. Human OSM (hOSM) and murine LIF (mLIF) act in mouse cells via a LIF receptor (LIFR)-glycoprotein 130 (gp130) heterodimer. In contrast, murine OSM (mOSM) signals mainly via an OSM receptor (OSMR)-gp130 heterodimer and binds with only very low affinity to mLIFR. hOSM and mLIF stimulate bone remodeling by both reducing osteocytic sclerostin and up-regulating the pro-osteoclastic factor receptor activator of NF-κB ligand (RANKL) in osteoblasts. In the absence of OSMR, mOSM still strongly suppressed sclerostin and stimulated bone formation but did not induce RANKL, suggesting that intracellular signaling activated by the low affinity interaction of mOSM with mLIFR is different from the downstream effects when mLIF or hOSM interacts with the same receptor. Both STAT1 and STAT3 were activated by mOSM in wild type cells or by mLIF/hOSM in wild type and Osmr-/- cells. In contrast, in Osmr-/- primary osteocyte-like cells stimulated with mOSM (therefore acting through mLIFR), microarray expression profiling and Western blotting analysis identified preferential phosphorylation of STAT3 and induction of its target genes but not of STAT1 and its target genes; this correlated with reduced phosphorylation of both gp130 and LIFR. In a mouse model of spontaneous osteopenia caused by hyperactivation of STAT1/3 signaling downstream of gp130 (gp130Y757F/Y757F), STAT1 deletion rescued the osteopenic phenotype, indicating a beneficial effect of promoting STAT3 signaling over STAT1 downstream of gp130 in this low bone mass condition, and this may have therapeutic value.
Collapse
Affiliation(s)
- Emma C Walker
- From the St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Rachelle W Johnson
- From the St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Yifang Hu
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Holly J Brennan
- From the St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Ingrid J Poulton
- From the St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Jian-Guo Zhang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Medical Biology, and
| | - Brendan J Jenkins
- Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton 3168, Victoria, Australia, and
| | - Gordon K Smyth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Departments of Mathematics and Statistics
| | - Nicos A Nicola
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Medical Biology, and
| | - Natalie A Sims
- From the St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia, .,Medicine at St. Vincent's Hospital, The University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
31
|
Dennie D, Louboutin JP, Strayer DS. Migration of bone marrow progenitor cells in the adult brain of rats and rabbits. World J Stem Cells 2016; 8:136-157. [PMID: 27114746 PMCID: PMC4835673 DOI: 10.4252/wjsc.v8.i4.136] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 09/11/2015] [Accepted: 02/16/2016] [Indexed: 02/06/2023] Open
Abstract
Neurogenesis takes place in the adult mammalian brain in three areas: Subgranular zone of the dentate gyrus (DG); subventricular zone of the lateral ventricle; olfactory bulb. Different molecular markers can be used to characterize the cells involved in adult neurogenesis. It has been recently suggested that a population of bone marrow (BM) progenitor cells may migrate to the brain and differentiate into neuronal lineage. To explore this hypothesis, we injected recombinant SV40-derived vectors into the BM and followed the potential migration of the transduced cells. Long-term BM-directed gene transfer using recombinant SV40-derived vectors leads to expression of the genes delivered to the BM firstly in circulating cells, then after several months in mature neurons and microglial cells, and thus without central nervous system (CNS) lesion. Most of transgene-expressing cells expressed NeuN, a marker of mature neurons. Thus, BM-derived cells may function as progenitors of CNS cells in adult animals. The mechanism by which the cells from the BM come to be neurons remains to be determined. Although the observed gradual increase in transgene-expressing neurons over 16 mo suggests that the pathway involved differentiation of BM-resident cells into neurons, cell fusion as the principal route cannot be totally ruled out. Additional studies using similar viral vectors showed that BM-derived progenitor cells migrating in the CNS express markers of neuronal precursors or immature neurons. Transgene-positive cells were found in the subgranular zone of the DG of the hippocampus 16 mo after intramarrow injection of the vector. In addition to cells expressing markers of mature neurons, transgene-positive cells were also positive for nestin and doublecortin, molecules expressed by developing neuronal cells. These cells were actively proliferating, as shown by short term BrdU incorporation studies. Inducing seizures by using kainic acid increased the number of BM progenitor cells transduced by SV40 vectors migrating to the hippocampus, and these cells were seen at earlier time points in the DG. We show that the cell membrane chemokine receptor, CCR5, and its ligands, enhance CNS inflammation and seizure activity in a model of neuronal excitotoxicity. SV40-based gene delivery of RNAi targeting CCR5 to the BM results in downregulating CCR5 in circulating cells, suggesting that CCR5 plays an important role in regulating traffic of BM-derived cells into the CNS, both in the basal state and in response to injury. Furthermore, reduction in CCR5 expression in circulating cells provides profound neuroprotection from excitotoxic neuronal injury, reduces neuroinflammation, and increases neuronal regeneration following this type of insult. These results suggest that BM-derived, transgene-expressing, cells can migrate to the brain and that they become neurons, at least in part, by differentiating into neuron precursors and subsequently developing into mature neurons.
Collapse
|
32
|
De Grandis M, Lhoumeau AC, Mancini SJC, Aurrand-Lions M. Adhesion receptors involved in HSC and early-B cell interactions with bone marrow microenvironment. Cell Mol Life Sci 2016; 73:687-703. [PMID: 26495446 PMCID: PMC11108274 DOI: 10.1007/s00018-015-2064-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 09/16/2015] [Accepted: 10/08/2015] [Indexed: 02/06/2023]
Abstract
Hematopoiesis takes place in the bone marrow of adult mammals and is the process by which blood cells are replenished every day throughout life. Differentiation of hematopoietic cells occurs in a stepwise manner through intermediates of differentiation that could be phenotypically identified. This has allowed establishing hematopoietic cell classification with hematopoietic stem cells (HSCs) at the top of the hierarchy. HSCs are mostly quiescent and serve as a reservoir for maintenance of lifelong hematopoiesis. Over recent years, it has become increasingly clear that HSC quiescence is not only due to intrinsic properties, but is also mediated by cognate interactions between HSCs and surrounding cells within micro-anatomical sites called “niches”. This hematopoietic/stromal crosstalk model also applies to more mature progenitors such as B cell progenitors, which are thought to reside in distinct “niches”. This prompted many research teams to search for specific molecular mechanisms supporting leuko-stromal crosstalk in the bone marrow and acting at specific stage of differentiation to regulate hematopoietic homeostasis. Here, we review recent data on adhesion mechanisms involved in HSCs and B cell progenitors interactions with surrounding bone marrow stromal cells.
Collapse
Affiliation(s)
- Maria De Grandis
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm U1068, CNRS UMR7258, Aix-Marseille Université UM105, Marseille, France
| | - Anne-Catherine Lhoumeau
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm U1068, CNRS UMR7258, Aix-Marseille Université UM105, Marseille, France
| | - Stéphane J. C. Mancini
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm U1068, CNRS UMR7258, Aix-Marseille Université UM105, Marseille, France
| | - Michel Aurrand-Lions
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm U1068, CNRS UMR7258, Aix-Marseille Université UM105, Marseille, France
| |
Collapse
|
33
|
Li L, Byrne SM, Rainville N, Su S, Jachimowicz E, Aucher A, Davis DM, Ashton-Rickardt PG, Wojchowski DM. Brief report: serpin Spi2A as a novel modulator of hematopoietic progenitor cell formation. Stem Cells 2015; 32:2550-6. [PMID: 24964278 DOI: 10.1002/stem.1778] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 05/05/2014] [Accepted: 05/30/2014] [Indexed: 01/15/2023]
Abstract
Prime regulation over hematopoietic progenitor cell (HPC) production is exerted by hematopoietins (HPs) and their Janus kinase-coupled receptors (HP-Rs). For HP/HP-R studies, one central challenge in determining specific effects involves the delineation of nonredundant signal transduction factors and their lineage restricted actions. Via loss-of-function studies, we define roles for an HP-regulated Serpina3g/Spi2A intracellular serpin during granulomyelocytic, B-cell, and hematopoietic stem cell (HSC) formation. In granulomyelocytic progenitors, granulocyte macrophage colony stimulating factor (GMCSF) strongly induced Serpina3g expression with Stat5 dependency. Spi2A-knockout (KO) led to 20-fold decreased CFU-GM formation, limited GMCSF-dependent granulocyte formation, and compromised neutrophil survival upon tumor necrosis factor alpha (TNF-α) exposure. In B-cell progenitors, Serpina3g was an interleukin-7 (IL7) target. Spi2A-KO elevated CFU-preB greater than sixfold and altered B-cell formation in competitive bone marrow transplant (BMT), and CpG challenge experiments. In HSCs, Serpina3g/Spi2A expression was also elevated. Spi2A-KO compromised LT-HSC proliferation (as well as lineage(neg) Sca1(pos) Kit(pos) (LSK) cell lysosomal integrity), and skewed LSK recovery post 5-FU. Spi2A therefore functions to modulate HP-regulated immune cell and HSC formation post-5-FU challenge.
Collapse
Affiliation(s)
- Lei Li
- COBRE Center of Excellence in Stem Cell Biology and Regenerative Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA; Department of Pediatrics, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Paul F, Arkin Y, Giladi A, Jaitin DA, Kenigsberg E, Keren-Shaul H, Winter D, Lara-Astiaso D, Gury M, Weiner A, David E, Cohen N, Lauridsen FKB, Haas S, Schlitzer A, Mildner A, Ginhoux F, Jung S, Trumpp A, Porse BT, Tanay A, Amit I. Transcriptional Heterogeneity and Lineage Commitment in Myeloid Progenitors. Cell 2015; 163:1663-77. [PMID: 26627738 DOI: 10.1016/j.cell.2015.11.013] [Citation(s) in RCA: 700] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/30/2015] [Accepted: 11/05/2015] [Indexed: 11/29/2022]
Abstract
Within the bone marrow, stem cells differentiate and give rise to diverse blood cell types and functions. Currently, hematopoietic progenitors are defined using surface markers combined with functional assays that are not directly linked with in vivo differentiation potential or gene regulatory mechanisms. Here, we comprehensively map myeloid progenitor subpopulations by transcriptional sorting of single cells from the bone marrow. We describe multiple progenitor subgroups, showing unexpected transcriptional priming toward seven differentiation fates but no progenitors with a mixed state. Transcriptional differentiation is correlated with combinations of known and previously undefined transcription factors, suggesting that the process is tightly regulated. Histone maps and knockout assays are consistent with early transcriptional priming, while traditional transplantation experiments suggest that in vivo priming may still allow for plasticity given strong perturbations. These data establish a reference model and general framework for studying hematopoiesis at single-cell resolution.
Collapse
Affiliation(s)
- Franziska Paul
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ya'ara Arkin
- Department of Computer Science and Applied Mathematics and Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Amir Giladi
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Ephraim Kenigsberg
- Department of Computer Science and Applied Mathematics and Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Hadas Keren-Shaul
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Deborah Winter
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - David Lara-Astiaso
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Meital Gury
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Assaf Weiner
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Nadav Cohen
- Department of Computer Science and Applied Mathematics and Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Felicia Kathrine Bratt Lauridsen
- The Finsen Laboratory, Rigshospitalet, University of Copenhagen, Copenhagen 2200, Denmark; Biotech Research and Innovation Centre (BRIC), Copenhagen 2200, Denmark; Danish Stem Cell Centre (DanStem) Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Simon Haas
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ), 69120 Heidelberg, Germany
| | - Andreas Schlitzer
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), BIOPOLIS 138648, Singapore; Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| | - Alexander Mildner
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), BIOPOLIS 138648, Singapore
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ), 69120 Heidelberg, Germany
| | - Bo Torben Porse
- The Finsen Laboratory, Rigshospitalet, University of Copenhagen, Copenhagen 2200, Denmark; Biotech Research and Innovation Centre (BRIC), Copenhagen 2200, Denmark; Danish Stem Cell Centre (DanStem) Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Amos Tanay
- Department of Computer Science and Applied Mathematics and Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
35
|
Wysoczynski M, Ratajczak J, Pedziwiatr D, Rokosh G, Bolli R, Ratajczak MZ. Identification of heme oxygenase 1 (HO-1) as a novel negative regulator of mobilization of hematopoietic stem/progenitor cells. Stem Cell Rev Rep 2015; 11:110-118. [PMID: 25086571 PMCID: PMC4333311 DOI: 10.1007/s12015-014-9547-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Activation of complement cascade (ComC) play and important role in mobilization of hematopoietic stem/progenitor cells (HSPCs) from bone marrow (BM) into peripheral blood (PB). While there are vast experimental data on the mechanisms and factors that induce or promote mobilization of HSPCs, there is relatively less data on negative regulators of this process. We demonstrate for the first time that heme oxygenase-1 (HO-1) that has a well-documented anti-inflammatory potential plays an important and heretofore unrecognized role in retention of HSPCs in BM niches by i) modulating negatively activation of mobilization promoting ComC, ii) maintaining stromal derived factor-1 (SDF-1) level in the BM microenvironment and iii) attenuating chemotactic responsiveness of HSPCs to SDF-1 and sphingosine-1 phosphate (S1P) gradients in PB. Furthermore, our data showing a positive mobilizing effect by a non-toxic small-molecule inhibitor of HO-1 (SnPP) suggest that blockade of HO-1 would be a promising strategy to facilitate mobilization of HSPCs. Further studies are also needed to evaluate better the molecular mechanisms responsible for the potential effect of HO-1 in homing of HSPCs after transplantation.
Collapse
Affiliation(s)
- Marcin Wysoczynski
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY USA
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
| | - Daniel Pedziwiatr
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
| | - Gregg Rokosh
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
| | - Roberto Bolli
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
| | - Mariusz Z. Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY 40202 USA
- Department of Physiology, Pomeranian University of Medicine, 70-111 Szczecin, Poland
| |
Collapse
|
36
|
Klein G, Schmal O, Aicher WK. Matrix metalloproteinases in stem cell mobilization. Matrix Biol 2015; 44-46:175-83. [PMID: 25617493 DOI: 10.1016/j.matbio.2015.01.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/15/2015] [Accepted: 01/15/2015] [Indexed: 01/05/2023]
Abstract
Hematopoietic stem cells (HSCs) have the capability to migrate back and forth between their preferred microenvironment in bone marrow niches and the peripheral blood, but under steady-state conditions only a marginal number of stem cells can be found in the circulation. Different mobilizing agents, however, which create a highly proteolytic milieu in the bone marrow, can drastically increase the number of circulating HSCs. Among other proteases secreted and membrane-bound matrix metalloproteinases (MMPs) are known to be involved in the induced mobilization process and can digest niche-specific extracellular matrix components and cytokines responsible for stem cell retention to the niches. Iatrogenic stem cell mobilization and stem cell homing to their niches are clinically employed on a routine basis, although the exact mechanisms of both processes are still not fully understood. In this review we provide an overview on the various roles of MMPs in the induced release of HSCs from the bone marrow.
Collapse
Affiliation(s)
- Gerd Klein
- Center for Medical Research, Department of Internal Medicine, Section for Transplantation Immunology and Immunohematology, University of Tübingen, Germany.
| | - Olga Schmal
- Center for Medical Research, Department of Internal Medicine, Section for Transplantation Immunology and Immunohematology, University of Tübingen, Germany
| | | |
Collapse
|
37
|
Chen H, Fang X, Zhu H, Li S, He J, Gu P, Fan D, Han F, Zeng Y, Yu X, Luo B, Xu H, Yi X. Gene expression profile analysis for different idiopathic interstitial pneumonias subtypes. Exp Lung Res 2014; 40:367-79. [PMID: 25058599 DOI: 10.3109/01902148.2014.933985] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Idiopathic interstitial pneumonias (IIPs) are a group of diffuse parenchymal lung diseases of unknown etiology characterized by the presence of various degrees of inflammation and fibrosis. We aimed to screen the differences among IIPs subtypes in the gene level by using the microarray expression profiles of normal lung tissue and IIPs tissue for the key genes associated with early diagnosis and treatment of IIPs. METHODS The gene expression profile of six kinds of IIPs (GSE 32537) subtypes tissue and normal lung tissues were downloaded. The differentially expressed genes (DEGs) in different IIPs subtypes were selected by using the expression profiling. In addition, the screened DEGs were further analyzed by function annotation, pathway analysis, and interaction network analysis to reveal the differences among these subtypes. RESULTS The gene expression analysis showed that nine genes including SERPINA3, IL1R2, CBS, MGAM, SLCO4A1, S100A12, FPR1, SDR16C5, and MT1X in six subtypes of IIPs were significantly increased. There were significant differences in DEGs among six subtypes of IIPs, and the DEGs of some IIPs subtypes involved in immune, inflammatory response and cell adhesion processes. Moreover, the PPI network analysis indicated that SERPINA3 played an important role in the molecular mechanisms of IIPs. CONCLUSION This comprehensive description of altered gene expression in different subtypes of IIPs underscores the complex biological processes characteristic of different subtypes of IIPs and may provide a foundation for future research into this devastating disease.
Collapse
Affiliation(s)
- Hanzhang Chen
- 1Department of Pathology, Central Hospital of Shanghai Zhabei District , Shanghai , China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Salazar-Olivo LA, Mejia-Elizondo R, Alonso-Castro AJ, Ponce-Noyola P, Maldonado-Lagunas V, Melendez-Zajgla J, Saavedra-Alanis VM. SerpinA3g participates in the antiadipogenesis and insulin-resistance induced by tumor necrosis factor-α in 3T3-F442A cells. Cytokine 2014; 69:180-8. [PMID: 24973688 DOI: 10.1016/j.cyto.2014.05.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 05/09/2014] [Accepted: 05/30/2014] [Indexed: 01/20/2023]
Abstract
Tumor necrosis factor alpha (TNF-α) is a proven modulator of adipose metabolism, but the mechanisms by which this cytokine affects the development and function of adipose tissue have not been fully elucidated to date. Using differential display analysis, in this study, we demonstrate that gene expression of the serine protease inhibitor A3g (SerpinA3g) is specifically induced in 3T3-F442A preadipocytes by TNF-α but not by other adipogenic inhibitors, such as retinoic acid (RA) or transforming growth factor type beta (TGF-β). The specific induction of SerpinA3g by TNF-α was confirmed by RT-PCR in both preadipose and terminally differentiated 3T3-F442A cells. The knockdown of SerpinA3g using small interfering RNA prevented the antiadipogenesis elicited by TNF-α in 3T3-F442A cells but not the antiadipogenesis induced by RA or TGF-β. SerpinA3g-silenced 3T3-F442A cells also did not display TNF-α-induced insulin resistance. Our results demonstrate that SerpinA3g is specifically induced by TNF-α in 3T3-F442A cells, regardless of their stage of differentiation, and participates in the antiadipogenesis and insulin resistance induced by this cytokine. Our results suggest that SerpinA3g plays a role in the TNF-α modulation of adipose tissue development and metabolism. Additional studies are warranted regarding the mechanisms mediating adipose SerpinA3g effects.
Collapse
Affiliation(s)
- Luis A Salazar-Olivo
- Instituto Potosino de Investigación Científica y Tecnológica, Molecular Biology Division, San Luis Potosí, México.
| | - Rebeca Mejia-Elizondo
- Instituto Potosino de Investigación Científica y Tecnológica, Molecular Biology Division, San Luis Potosí, México
| | - Angel Josabad Alonso-Castro
- Instituto Potosino de Investigación Científica y Tecnológica, Molecular Biology Division, San Luis Potosí, México
| | - Patricia Ponce-Noyola
- Universidad de Guanajuato, Department of Biology, Division of Natural and Exact Sciences, Guanajuato, México
| | | | | | | |
Collapse
|
39
|
Hou A, Lan W, Law KP, Khoo SCJ, Tin MQ, Lim YP, Tong L. Evaluation of global differential gene and protein expression in primary Pterygium: S100A8 and S100A9 as possible drivers of a signaling network. PLoS One 2014; 9:e97402. [PMID: 24825356 PMCID: PMC4019582 DOI: 10.1371/journal.pone.0097402] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 04/16/2014] [Indexed: 01/12/2023] Open
Abstract
Purpose Pterygium is a wing shaped fibrovascular growth on the ocular surface, characterized by fibrosis, angiogenesis, extracellular matrix remodeling, and inflammatory infiltrates. Epidemiologic studies have linked pterygium formation to various chronic inflammatory conditions, such as ultraviolet radiation, sawdust exposure, and dry eye disease. The purpose of this study is to identify proteins that are differentially expressed in primary pterygium by using a combination of gene microarray and proteomic platforms. Methods Paired pterygium and uninvolved conjunctiva tissues of four patients were evaluated for differences in global gene transcript levels using a genechip microarray. Proteins extracted from another four pairs of tissues were quantified by iTRAQ approach. Western blot and immunofluorescent staining on additional patients were used to validate dysregulated protein expression obtained from microarray and proteomics data. In addition, primary conjunctival fibroblasts were treated with recombinant S100A8, S100A9 or both. Transcript level changes of a panel of potential target genes were evaluated by real time-PCR. Results The following were up-regulated at both protein and transcript levels S100 A8 and A9, aldehyde dehydrogenase 3 family, member1 (ALDH3A1) and vimentin (VIM). Conversely, serpin peptidase inhibitor clade A member 1 (SERPINA1) and transferrin (TF) were down-regulated. Upon adding S100A8, S100A9 or both, the inflammatory chemokine CXCL1, matrix proteins vimentin, biglycan, and gelsolin, as well as annexin-A2, thymosin-β4, chymase (CMA1), member of Ras oncogene family RAB10 and SERPINA1 were found to be up-regulated. Conclusions We identified 3 up-regulated and 2 down-regulated proteins by using a stringent approach comparing microarray and proteomic data. On stimulating cells with S100A8/9, a repertoire of key genes found to be up-regulated in pterygium tissue, were induced in these cells. S100A8/9 may be an upstream trigger for inflammation and other disease pathways in pterygium.
Collapse
Affiliation(s)
- Aihua Hou
- Ocular Surface Research Group, Singapore Eye Research Institute, Singapore, Singapore
- * E-mail: (AH); (LT)
| | - Wanwen Lan
- Ocular Surface Research Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Kai Pong Law
- Department of Biochemistry, National University of Singapore, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ser Chin Jasmine Khoo
- Ocular Surface Research Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Min Qi Tin
- Ocular Surface Research Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Yoon Pin Lim
- Department of Biochemistry, National University of Singapore, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Louis Tong
- Ocular Surface Research Group, Singapore Eye Research Institute, Singapore, Singapore
- Department of Biochemistry, National University of Singapore, Singapore, Singapore
- Duke-NUS Graduate Medical School, Singapore, Singapore, Singapore
- Singapore National Eye Center, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- * E-mail: (AH); (LT)
| |
Collapse
|
40
|
Peripheral blood hematopoietic stem and progenitor cell frequency is unchanged in patients with alpha-1-antitrypsin deficiency. Int J Hematol 2014; 99:714-20. [PMID: 24752699 DOI: 10.1007/s12185-014-1581-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 12/17/2022]
Abstract
Granulocyte-colony-stimulating factor (G-CSF)-induced hematopoietic stem and progenitor cell (HSPC) mobilization is associated with the release of neutrophil-derived proteases. Previously, we have shown that alpha-1-antitrypsin (AAT) inhibits these proteases in mice, resulting in inhibition of HSPC mobilization. Here, we studied the relationship between AAT and HSPC in steady state and cytokine-induced mobilization in humans. Patients with alpha-1-antitrypsin deficiency (AATD) have an 85-90 % decrease of AAT in the peripheral blood (PB). We hypothesized that this leads to increased proteolytic activity in the bone marrow and increased steady-state PB HSPC numbers. Using flow cytometry and semi-solid cell culture, we found no significant difference in PB HSPC in AATD patients (n = 18) as compared to controls (n = 22). Healthy stem cell donors (n = 43) were mobilized with G-CSF for 5 days and the number of CD45(+)/CD34(+) HSPC were determined in PB. We found that, during mobilization, PB AAT levels increased significantly, positively correlating with PB CD45(+)/CD34(+) cells (r = 0.31, p = 0.005). In conclusion, although serum AAT levels and HSPC mobilization in healthy stem cell donors are positively correlated, AAT is not an indispensable protease-inhibitor in the constitutive circulation of HSPC. These findings suggest a model in which both protease-dependent and -independent pathways contribute to HSPC mobilization.
Collapse
|
41
|
Nakayama R, Kuroda J, Taniyama N, Yamamoto-Sugitani M, Wada S, Kiyota M, Mizutani S, Chinen Y, Matsumoto Y, Nagoshi H, Shimura Y, Kobayashi T, Horiike S, Sato K, Taniwaki M. Suppression of SERPINA1-albumin complex formation by galectin-3 overexpression leads to paracrine growth promotion of chronic myelogenous leukemia cells. Leuk Res 2013; 38:103-8. [PMID: 23953881 DOI: 10.1016/j.leukres.2013.07.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 07/12/2013] [Indexed: 01/08/2023]
Abstract
Galectin-3 is induced in chronic myelogenous leukemia (CML) cells by co-culture with bone marrow stromal cells, making paracrine growth promotion of CML cells in conditioned medium (CM) from galectin-3 overexpressing CML cells more potent. We used gel filtration chromatography to demonstrate that the bovine SERPINA1-fetal bovine serum albumin (BSA) complex was specifically suppressed in CM from galectin-3 overexpressing cells. The SERPINA1-BSA complex as well as human plasma SERPINA1 inhibited the growth of CML cells, while exogenous galectin-3 partly offset this effect. These findings suggest that galectin-3 overexpression promotes paracrine growth of CML cells by interfering with the action of the growth inhibitory SERPINA1-albumin complex.
Collapse
Affiliation(s)
- Ryuko Nakayama
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Junya Kuroda
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Nobuko Taniyama
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Mio Yamamoto-Sugitani
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Sayori Wada
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Miki Kiyota
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shinsuke Mizutani
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshiaki Chinen
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yosuke Matsumoto
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hisao Nagoshi
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuji Shimura
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tsutomu Kobayashi
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigeo Horiike
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenji Sato
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Masafumi Taniwaki
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
42
|
Grek CL, Zhang J, Manevich Y, Townsend DM, Tew KD. Causes and consequences of cysteine S-glutathionylation. J Biol Chem 2013; 288:26497-504. [PMID: 23861399 DOI: 10.1074/jbc.r113.461368] [Citation(s) in RCA: 251] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Post-translational S-glutathionylation occurs through the reversible addition of a proximal donor of glutathione to thiolate anions of cysteines in target proteins, where the modification alters molecular mass, charge, and structure/function and/or prevents degradation from sulfhydryl overoxidation or proteolysis. Catalysis of both the forward (glutathione S-transferase P) and reverse (glutaredoxin) reactions creates a functional cycle that can also regulate certain protein functional clusters, including those involved in redox-dependent cell signaling events. For translational application, S-glutathionylated serum proteins may be useful as biomarkers in individuals (who may also have polymorphic expression of glutathione S-transferase P) exposed to agents that cause oxidative or nitrosative stress.
Collapse
Affiliation(s)
- Christina L Grek
- From the Departments of Cell and Molecular Pharmacology and Experimental Therapeutics and
| | | | | | | | | |
Collapse
|
43
|
Pharmacologic stabilization of HIF-1α increases hematopoietic stem cell quiescence in vivo and accelerates blood recovery after severe irradiation. Blood 2013; 121:759-69. [DOI: 10.1182/blood-2012-02-408419] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Key Points
HIF-1α protein stabilization increases HSC quiescence in vivo. HIF-1α protein stabilization increases HSC resistance to irradiation and accelerates recovery.
Collapse
|
44
|
Hosing C. Hematopoietic stem cell mobilization with G-CSF. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2012; 904:37-47. [PMID: 22890920 DOI: 10.1007/978-1-61779-943-3_3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
Cytokine mobilized peripheral blood stem cells are the preferred source of stem cells in autologous stem cell transplantation and have virtually replaced bone marrow as the stem cell source. In recent years, a dramatic increase has been reported in the use of peripheral blood stem cells for allogeneic transplantation as well. The reason for this rise is that peripheral blood stem cell transplants when compared to bone marrow transplants are associated with a more rapid recovery of granulocytes and platelets after transplantation and a lower regimen-related and transplant-related mortality. Peripheral blood stem cells can be easily harvested on an outpatient basis without the need for general anesthesia. In most cases peripheral blood stem cells are collected after G-CSF administration. In this chapter we describe peripheral blood stem cell mobilization in autologous transplant patients and in allogeneic donors using G-CSF.
Collapse
Affiliation(s)
- Chitra Hosing
- Department of Stem Cell Transplantation and Cell Therapy, M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
45
|
Winkler IG, Barbier V, Nowlan B, Jacobsen RN, Forristal CE, Patton JT, Magnani JL, Lévesque JP. Vascular niche E-selectin regulates hematopoietic stem cell dormancy, self renewal and chemoresistance. Nat Med 2012; 18:1651-7. [DOI: 10.1038/nm.2969] [Citation(s) in RCA: 314] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 09/12/2012] [Indexed: 01/16/2023]
|
46
|
Byrne SM, Aucher A, Alyahya S, Elder M, Olson ST, Davis DM, Ashton-Rickardt PG. Cathepsin B controls the persistence of memory CD8+ T lymphocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2012; 189:1133-43. [PMID: 22745374 PMCID: PMC3401340 DOI: 10.4049/jimmunol.1003406] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The persistence of memory T lymphocytes confers lifelong protection from pathogens. Memory T cells survive and undergo homeostatic proliferation (HSP) in the absence of Ag, although the cell-intrinsic mechanisms by which cytokines drive the HSP of memory T cells are not well understood. In this study we report that lysosome stability limits the long-term maintenance of memory CD8(+) T cell populations. Serine protease inhibitor (Spi) 2A, an anti-apoptotic cytosolic cathepsin inhibitor, is induced by both IL-15 and IL-7. Mice deficient in Spi2A developed fewer memory phenotype CD44(hi)CD8(+) T cells with age, which underwent reduced HSP in the bone marrow. Spi2A was also required for the maintenance of central memory CD8(+) T cell populations after acute infection with lymphocytic choriomeningitis virus. Spi2A-deficient Ag-specific CD8(+) T cell populations declined more than wild-type competitors after viral infection, and they were eroded further after successive infections. Spi2A protected memory cells from lysosomal breakdown by inhibiting cathepsin B. The impaired maintenance of Spi2A-deficient memory CD8(+) T cells was rescued by concomitant cathepsin B deficiency, demonstrating that cathepsin B was a physiological target of Spi2A in memory CD8(+) T cell survival. Our findings support a model in which protection from lysosomal rupture through cytokine-induced expression of Spi2A determines the long-term persistence of memory CD8(+) T cells.
Collapse
Affiliation(s)
- Susan M. Byrne
- Section of Immunobiology, Division of Immunology and Inflammation, Department of Medicine, Faculty of Medicine, Imperial College London, London W12 0NN, UK
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Anne Aucher
- Section of Immunology and Infection, Division of Cell and Molecular Biology, Imperial College London, London SW7 2AZ
| | - Syarifah Alyahya
- Section of Immunobiology, Division of Immunology and Inflammation, Department of Medicine, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Matthew Elder
- Section of Immunobiology, Division of Immunology and Inflammation, Department of Medicine, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Steven T. Olson
- Center for Molecular Biology of Oral Diseases, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Daniel M. Davis
- Section of Immunology and Infection, Division of Cell and Molecular Biology, Imperial College London, London SW7 2AZ
| | - Philip G. Ashton-Rickardt
- Section of Immunobiology, Division of Immunology and Inflammation, Department of Medicine, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| |
Collapse
|
47
|
Grek CL, Townsend DM, Uys JD, Manevich Y, Coker WJ, Pazoles CJ, Tew KD. S-glutathionylated serine proteinase inhibitors as plasma biomarkers in assessing response to redox-modulating drugs. Cancer Res 2012; 72:2383-93. [PMID: 22406622 DOI: 10.1158/0008-5472.can-11-4088] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Many cancer drugs impact cancer cell redox regulatory mechanisms and disrupt redox homeostasis. Pharmacodynamic biomarkers that measure therapeutic efficacy or toxicity could improve patient management. Using immunoblot analyses and mass spectrometry, we identified that serpins A1 and A3 were S-glutathionylated in a dose- and time-dependent manner following treatment of mice with drugs that alter reactive oxygen or nitrogen species. Tandem mass spectrometry analyses identified Cys(256) of serpin A1 and Cys(263) of serpin A3 as the S-glutathionylated residues. In human plasma from cancer patients, there were higher levels of unmodified serpin A1 and A3, but following treatments with redox active drugs, relative S-glutathionylation of these serpins was higher in plasma from normal individuals. There is potential for S-glutathionylated serpins A1 and A3 to act as pharmacodynamic biomarkers for evaluation of patient response to drugs that target redox pathways.
Collapse
Affiliation(s)
- Christina L Grek
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Bristow CL, Babayeva MA, LaBrunda M, Mullen MP, Winston R. α1Proteinase inhibitor regulates CD4+ lymphocyte levels and is rate limiting in HIV-1 disease. PLoS One 2012; 7:e31383. [PMID: 22363634 PMCID: PMC3281957 DOI: 10.1371/journal.pone.0031383] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 01/06/2012] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The regulation of adult stem cell migration through human hematopoietic tissue involves the chemokine CXCL12 (SDF-1) and its receptor CXCR4 (CD184). In addition, human leukocyte elastase (HLE) plays a key role. When HLE is located on the cell surface (HLE(CS)), it acts not as a proteinase, but as a receptor for α(1)proteinase inhibitor (α(1)PI, α(1)antitrypsin, SerpinA1). Binding of α(1)PI to HLE(CS) forms a motogenic complex. We previously demonstrated that α(1)PI deficiency attends HIV-1 disease and that α(1)PI augmentation produces increased numbers of immunocompetent circulating CD4(+) lymphocytes. Herein we investigated the mechanism underlying the α(1)PI deficiency that attends HIV-1 infection. METHODS AND FINDINGS Active α(1)PI in HIV-1 subjects (median 17 µM, n = 35) was significantly below normal (median 36 µM, p<0.001, n = 30). In HIV-1 uninfected subjects, CD4(+) lymphocytes were correlated with the combined factors α(1)PI, HLE(CS) (+) lymphocytes, and CXCR4(+) lymphocytes (r(2) = 0.91, p<0.001, n = 30), but not CXCL12. In contrast, in HIV-1 subjects with >220 CD4 cells/µl, CD4(+) lymphocytes were correlated solely with active α(1)PI (r(2) = 0.93, p<0.0001, n = 26). The monoclonal anti-HIV-1 gp120 antibody 3F5 present in HIV-1 patient blood is shown to bind and inactivate human α(1)PI. Chimpanzee α(1)PI differs from human α(1)PI by a single amino acid within the 3F5-binding epitope. Unlike human α(1)PI, chimpanzee α(1)PI did not bind 3F5 or become depleted following HIV-1 challenge, consistent with the normal CD4(+) lymphocyte levels and benign syndrome of HIV-1 infected chimpanzees. The presence of IgG-α(1)PI immune complexes correlated with decreased CD4(+) lymphocytes in HIV-1 subjects. CONCLUSIONS This report identifies an autoimmune component of HIV-1 disease that can be overcome therapeutically. Importantly, results identify an achievable vaccine modification with the novel objective to protect against AIDS as opposed to the current objective to protect against HIV-1 infection.
Collapse
Affiliation(s)
- Cynthia L Bristow
- Weill Cornell Medical College, New York, New York, United States of America.
| | | | | | | | | |
Collapse
|
49
|
Xiong Y, Uys JD, Tew KD, Townsend DM. S-glutathionylation: from molecular mechanisms to health outcomes. Antioxid Redox Signal 2011; 15:233-70. [PMID: 21235352 PMCID: PMC3110090 DOI: 10.1089/ars.2010.3540] [Citation(s) in RCA: 244] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Redox homeostasis governs a number of critical cellular processes. In turn, imbalances in pathways that control oxidative and reductive conditions have been linked to a number of human disease pathologies, particularly those associated with aging. Reduced glutathione is the most prevalent biological thiol and plays a crucial role in maintaining a reduced intracellular environment. Exposure to reactive oxygen or nitrogen species is causatively linked to the disease pathologies associated with redox imbalance. In particular, reactive oxygen species can differentially oxidize certain cysteine residues in target proteins and the reversible process of S-glutathionylation may mitigate or mediate the damage. This post-translational modification adds a tripeptide and a net negative charge that can lead to distinct structural and functional changes in the target protein. Because it is reversible, S-glutathionylation has the potential to act as a biological switch and to be integral in a number of critical oxidative signaling events. The present review provides a comprehensive account of how the S-glutathionylation cycle influences protein structure/function and cellular regulatory events, and how these may impact on human diseases. By understanding the components of this cycle, there should be opportunities to intervene in stress- and aging-related pathologies, perhaps through prevention and diagnostic and therapeutic platforms.
Collapse
Affiliation(s)
- Ying Xiong
- Department of Pharmaceutical Sciences, Medical University of South Carolina, Charleston, 29425, USA
| | | | | | | |
Collapse
|
50
|
Wang D, Wang W, Dawkins P, Paterson T, Kalsheker N, Sallenave JM, Houghton AM. Deletion of Serpina1a, a murine α1-antitrypsin ortholog, results in embryonic lethality. Exp Lung Res 2011; 37:291-300. [PMID: 21574874 DOI: 10.3109/01902148.2011.554599] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is the fourth leading cause of death in the United States Approximately 1% to 2% of COPD patients suffer from α(1)-antitrypsin (A1AT) deficiency, the major inheritable predisposition to COPD/emphysema. To further study the role of A1AT deficiency in the pathogenesis of COPD/emphysema, the authors attempted to generate null-mutant mice for Serpina1a, 1 of 2 A1AT orthologs in mice. Here the authors show that targeted deletion of Serpina1a results in embryonic lethality prior to 8.5 days post conception (dpc). The results are surprising given that A1AT-null humans exist and therefore do not require this gene product for normal development. The Serpina1 gene cluster is substantially different between mouse and man. Through gene duplication, mice have 3 to 5 (depending on the strain) highly homologous proteinase inhibiting (Pi) genes, 2 of which inhibit neutrophil elastase. Despite the abundance of Pi genes in mice, Serpina1a serves a critical, nonredundant function during early mouse development. A1AT-deficient mice have been highly sought after to study emphysema, cancer, and liver disease, and as a model to perfect gene replacement therapy. These results highlight important differences between human and murine serpins and point to the difficulty inherent to using gene-targeted mice to study this common human genetic disease.
Collapse
Affiliation(s)
- Dongmei Wang
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | | | | | | | | | | |
Collapse
|