1
|
Müller GA, Müller TD. (Patho)Physiology of Glycosylphosphatidylinositol-Anchored Proteins II: Intercellular Transfer of Matter (Inheritance?) That Matters. Biomolecules 2023; 13:994. [PMID: 37371574 DOI: 10.3390/biom13060994] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Glycosylphosphatidylinositol (GPI)-anchored proteins (APs) are anchored at the outer leaflet of the plasma membrane (PM) bilayer by covalent linkage to a typical glycolipid and expressed in all eukaryotic organisms so far studied. Lipolytic release from PMs into extracellular compartments and intercellular transfer are regarded as the main (patho)physiological roles exerted by GPI-APs. The intercellular transfer of GPI-APs relies on the complete GPI anchor and is mediated by extracellular vesicles such as microvesicles and exosomes and lipid-free homo- or heteromeric aggregates, and lipoprotein-like particles such as prostasomes and surfactant-like particles, or lipid-containing micelle-like complexes. In mammalian organisms, non-vesicular transfer is controlled by the distance between donor and acceptor cells/tissues; intrinsic conditions such as age, metabolic state, and stress; extrinsic factors such as GPI-binding proteins; hormones such as insulin; and drugs such as anti-diabetic sulfonylureas. It proceeds either "directly" upon close neighborhood or contact of donor and acceptor cells or "indirectly" as a consequence of the induced lipolytic release of GPI-APs from PMs. Those displace from the serum GPI-binding proteins GPI-APs, which have retained the complete anchor, and become assembled in aggregates or micelle-like complexes. Importantly, intercellular transfer of GPI-APs has been shown to induce specific phenotypes such as stimulation of lipid and glycogen synthesis, in cultured human adipocytes, blood cells, and induced pluripotent stem cells. As a consequence, intercellular transfer of GPI-APs should be regarded as non-genetic inheritance of (acquired) features between somatic cells which is based on the biogenesis and transmission of matter such as GPI-APs and "membrane landscapes", rather than the replication and transmission of information such as DNA. Its operation in mammalian organisms remains to be clarified.
Collapse
Affiliation(s)
- Günter A Müller
- Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) and German Center for Diabetes Research (DZD) at the Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) and German Center for Diabetes Research (DZD) at the Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| |
Collapse
|
2
|
Müller GA. Membrane insertion and intercellular transfer of glycosylphosphatidylinositol-anchored proteins: potential therapeutic applications. Arch Physiol Biochem 2020; 126:139-156. [PMID: 30445857 DOI: 10.1080/13813455.2018.1498904] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Anchorage of a subset of cell surface proteins in eukaryotic cells is mediated by a glycosylphosphatidylinositol (GPI) moiety covalently attached to the carboxy-terminus of the protein moiety. Experimental evidence for the potential of GPI-anchored proteins (GPI-AP) of being released from cells into the extracellular environment has been accumulating, which involves either the loss or retention of the GPI anchor. Release of GPI-AP from donor cells may occur spontaneously or in response to endogenous or environmental signals. The experimental evidence for direct insertion of exogenous GPI-AP equipped with the complete anchor structure into the outer plasma membrane bilayer leaflets of acceptor cells is reviewed as well as the potential underlying molecular mechanisms. Furthermore, promiscuous transfer of certain GPI-AP between plasma membranes of different cells in vivo under certain (patho)physiological conditions has been reported. Engineering of target cell surfaces using chimeric GPI-AP with complete GPI anchor may be useful for therapeutic applications.
Collapse
Affiliation(s)
- Günter A Müller
- Helmholtz Diabetes Center (HDC) at the Helmholtz Center München, Institute for Diabetes and Obesity, Oberschleissheim, Germany
- Department Biology I, Genetics, Ludwig-Maximilians-University München, Planegg-Martinsried, Germany
| |
Collapse
|
3
|
Müller GA. The release of glycosylphosphatidylinositol-anchored proteins from the cell surface. Arch Biochem Biophys 2018; 656:1-18. [DOI: 10.1016/j.abb.2018.08.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/07/2018] [Accepted: 08/14/2018] [Indexed: 12/15/2022]
|
4
|
The dysfunction of platelets in paroxysmal nocturnal hemoglobinuria. Thromb Res 2016; 148:50-55. [PMID: 27780113 DOI: 10.1016/j.thromres.2016.07.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 06/06/2016] [Accepted: 07/21/2016] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Thrombosis is a dangerous complication of paroxysmal nocturnal hemoglobinuria (PNH) and has a high mortality rate. However, the mechanism underlying the development of thrombosis in PNH remains unclear. To explore this, platelet function and serum complement activity were investigated in 14 patients with classical PNH, 11 with PNH aplastic anemia (AA) and 30 healthy controls. MATERIAL AND METHODS Serum concentrations of the terminal complement complex (sC5b-9) were determined by enzyme-linked immunofluorescence assay (ELISA), and the levels of C5b-9, CD61 and CD62p on platelet membranes were determined by flow cytometry. Clinical parameters were assessed, including D-dimer and platelet aggregation induced by adenosine diphosphate (ADP) and arachidonic acid (ARA). RESULTS Serum sC5b-9 concentrations were significantly lower in the PNH/PNH-AA than in the control group (P<0.01). C5b-9 deposition was significantly higher on CD59- platelets than on CD59+ platelets in PNH/PNH-AA patients and healthy controls (P<0.01 for each). D-dimer concentration was significantly higher in PNH/PNH-AA patients - especially those with lactate dehydrogenase (LDH) concentrations>1000U/L - than in controls (P<0.05). CD61 (P<0.05) expression was lower on CD59+ platelets in PNH than in controls and CD5- platelets in PNH. Expression of CD62p (P<0.01) was lower on CD59- and CD59+ platelets (P<0.01) in PNH cases than in controls. Platelet aggregation stimulated by the agonists ADP and ARA in the PNH/PNH-AA patients was significantly lower than that in controls (P<0.05). CONCLUSIONS The adhesion and aggregation of platelets, especially of CD59+ platelets, were compensatively decreased in PNH/PNH-AA patients without active thrombosis.
Collapse
|
5
|
Fibach E, Dana M. Oxidative stress in paroxysmal nocturnal hemoglobinuria and other conditions of complement-mediated hemolysis. Free Radic Biol Med 2015; 88:63-9. [PMID: 25937178 DOI: 10.1016/j.freeradbiomed.2015.04.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/23/2015] [Accepted: 04/21/2015] [Indexed: 12/11/2022]
Abstract
The complement (C') system and redox status play important roles in the physiological functioning of the body, such as the defense system, but they are also involved in various pathological conditions, including hemolytic anemia. Herein, we review the interaction between the C' and the redox systems in C'-mediated hemolytic anemias, paroxysmal nocturnal hemoglobinuria (PNH) and autoimmune hemolytic anemia, including acute hemolytic transfusion reaction. Blood cells in these diseases have been shown to have increased oxidative status, which was further elevated by interaction with activated C'. The results suggest that oxidative stress, in conjunction with activated C', may cause the underlying symptoms of these diseases, such as intra- and extravascular hemolysis and thrombotic complications. Antioxidants ameliorate oxidative stress by preventing generation of free radicals, by scavenging and preventing their accumulation, and by correcting their cellular damage. Antioxidants have been shown to reduce the oxidative stress and inhibit hemolysis as well as platelet activation mediated by activated C'. This raises the possibility that treatment with antioxidants might be considered as a potential therapeutic modality for C'-mediated hemolytic anemias. Currently, eculizumab, a humanized monoclonal antibody that specifically targets the C' protein C5, is the main treatment modality for PNH. However, because antioxidants are well tolerated and relatively inexpensive, they might be considered as potential adjuvants or an alternative therapeutic modality for PNH and other C'-mediated hemolytic anemias.
Collapse
Affiliation(s)
- Eitan Fibach
- Department of Hematology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel.
| | - Mutaz Dana
- Department of Hematology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| |
Collapse
|
6
|
Meng Y, Fu R, Liu H, Wang Y, Li L, Liu C, Zhang T, Ding S, Li L, Ruan E, Qu W, Wang H, Wang X, Wang G, Liu H, Wu Y, Song J, Xing L, Guan J, Shao Z. [Study on C5b-9 deposited on the membrane of platelets and its dysfunction in patients with paroxysmal nocturnal hemoglobinuria]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2015; 36:516-9. [PMID: 26134020 PMCID: PMC7343058 DOI: 10.3760/cma.j.issn.0253-2727.2015.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVE To explore the expression levels of terminal complement complex (C5b-9) and CD62p on platelets and the soluble C5b-9 (sC5b-9) level in serum in patients with PNH or PNH-aplastic anemia (AA). METHODS Serum levels of sC5b-9, complement C3 and C4 were detected by using ELISA in 25 patients with PNH/PNH-AA. The quantities of C5b-9 and CD62p on the membrane of platelets were detected by flow cytometry. RESULTS ①In PNH/PNH-AA group, the serum sC5b-9 level [390.27(265.73-676.87) μg/L] was lower than that in control group [540.39(344.20-1 576.78) μg/L] (P<0.01). ②The platelet PNH clone (CD59⁻CD61⁺/CD61⁺) size [50.58(23.29-81.60)%] was bigger in the PNH/PNH-AA group than that [23.57(15.58-29.02)%] in control group (P<0.01). The percentages of C5b-9 deposition (C5b-9⁺CD61⁺/CD61⁺) were higher on the PNH clone platelets (CD59⁻CD61⁺) in the PNH/PNH-AA group [(17.53 ± 6.27)%] than those on the normal platelets (CD59⁺CD61⁺) in PNH patients 11.33±5.03)%] and control [(10.88±3.58)%] group (P<0.01). ③ The expression of CD62p (CD62p⁺CD61⁺/CD61⁺) on PNH clone platelets in PNH patients [(61.98 ± 11.71)%] was higher than that on the normal platelets in PNH patients [(43.76±11.30)%] and control group [(38.23±18.07)%] (P<0.01). In addition, the expression of CD62p on normal platelets was higher in PNH patients than control (P<0.05). ④The deposition of C5b-9 positively correlated with the expression of CD62p on the platelets (r=0.559, P=0.002). CONCLUSION Deficiency of CD59 antigen on platelets in PNH patients may lead to the deposition of C5b-9 on its membrane and its dysfunction, which may contribute to thrombosis events in PNH.
Collapse
Affiliation(s)
- Yinping Meng
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Rong Fu
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Hui Liu
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Yihao Wang
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Lijuan Li
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Chunyan Liu
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Tian Zhang
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Shaoxue Ding
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Liyan Li
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Erbao Ruan
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Wen Qu
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Huaquan Wang
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Xiaoming Wang
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Guojin Wang
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Hong Liu
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Yuhong Wu
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Jia Song
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Limin Xing
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Jing Guan
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Zonghong Shao
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| |
Collapse
|
7
|
Canalejo K, Riera Cervantes N, Felippo M, Sarandría C, Aixalá M. Paroxysmal nocturnal haemoglobinuria. Experience over a 10 years period. Int J Lab Hematol 2013; 36:213-21. [PMID: 24188430 DOI: 10.1111/ijlh.12156] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 09/09/2013] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Paroxysmal nocturnal hemoglobinuria (PNH) is a hemolytic, clonal and acquired disorder of the hematopoietic stem cell with a deficiency of all glycophosphatidyl-inositol (GPI) linked proteins. The aim of this retrospective study was to analyse haematological and biochemical data from 152 patients referred to our laboratory for diagnosis of PNH by flow cytometry (FC). METHODS Patients and healthy donor (152 and 99 respectively) were studied. Ham, sucrose, lactate dehydrogenase (LDH), Iron, haptoglobin (Hp), blood cell morphology and Kaplow cytochemical stain for leukocyte alkaline phosphatase (LAP) were carried out. GPI-proteins anti-CD55 and CD59 in erythrocytes and the former, plus anti CD16b and CD66b on neutrophils were evaluated by FC. RESULTS Anemia and/or leukopenia and/or thrombocytopenia, increased reticulocyte count and LDH were observed in patients with PNH clone. Some of them had dacriocytes, schistocytes. LAP was low. On average, we detected 50% CD59 (-) erythrocytes and 29, 83, 78% CD55/59 (-), CD16b (-), CD66b (-) neutrophils, respectively. CONCLUSION Paroxysmal nocturnal hemoglobinuria clone was detected in 20/152 patients. Negative population's percentages were high in patients with classic PNH, Hematimetry, LAP and adequate use of CF contribute to PNH clone detection in the laboratory.
Collapse
Affiliation(s)
- K Canalejo
- Apoyo Médico, Instituto de Investigaciones Hematológicas, Academia Nacional de Medicina, Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
8
|
Abstract
The most frequent and feared complication of paroxysmal nocturnal hemoglobinuria (PNH) is thrombosis. Recent research has demonstrated that the complement and coagulation systems are closely integrated with each influencing the activity of the other to the extent that thrombin itself has recently been shown to activate the alternative pathway of complement. This may explain some of the complexity of the thrombosis in PNH. In this review, the recent changes in our understanding of the pathophysiology of thrombosis in PNH, as well as the treatment of thrombosis, will be discussed. Mechanisms explored include platelet activation, toxicity of free hemoglobin, nitric oxide depletion, absence of other glycosylphosphatidylinositol-linked proteins such as urokinase-type plasminogen activator receptor and endothelial dysfunction. Complement inhibition with eculizumab has a dramatic effect in PNH and has a major impact in the prevention of thrombosis as well as its management in this disease.
Collapse
|
9
|
Ferreira VP, Pangburn MK, Cortés C. Complement control protein factor H: the good, the bad, and the inadequate. Mol Immunol 2010; 47:2187-97. [PMID: 20580090 DOI: 10.1016/j.molimm.2010.05.007] [Citation(s) in RCA: 308] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The complement system is an essential component of the innate immune system that participates in elimination of pathogens and altered host cells and comprises an essential link between the innate and adaptive immune system. Soluble and membrane-bound complement regulators protect cells and tissues from unintended complement-mediated injury. Complement factor H is a soluble complement regulator essential for controlling the alternative pathway in blood and on cell surfaces. Normal recognition of self-cell markers (i.e. polyanions) and C3b/C3d fragments is necessary for factor H function. Inadequate recognition of host cell surfaces by factor H due to mutations and polymorphisms have been associated with complement-mediated tissue damage and disease. On the other hand, unwanted recognition of pathogens and altered self-cells (i.e. cancer) by factor H is used as an immune evasion strategy. This review will focus on the current knowledge related to these versatile recognition properties of factor H.
Collapse
Affiliation(s)
- Viviana P Ferreira
- Department of Medical Microbiology and Immunology, College of Medicine, University of Toledo, Toledo, OH 43614, United States.
| | | | | |
Collapse
|
10
|
Xiao J, Han B, Wu YJ, Zhong YP, Sun WL. Ex vivo expansion and long-term hematopoietic reconstitution ability of sorted CD34+CD59+ cells from patients with paroxysmal nocturnal hemoglobinuria. Int J Hematol 2010; 92:58-67. [PMID: 20577837 DOI: 10.1007/s12185-010-0628-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2009] [Revised: 06/02/2010] [Accepted: 06/04/2010] [Indexed: 11/27/2022]
Abstract
Autologous bone marrow transplantation (ABMT) for paroxysmal nocturnal hemoglobinuria (PNH) remains difficult so far. To expand residual normal CD34(+)CD59(+) cells isolated from patients with PNH and observe the long-term hematopoietic reconstruction ability of the expanded cells both ex vivo and in vivo, CD34(+)CD59(+) cells from 13 PNH patients and CD34(+) cells from 11 normal controls were separated from bone marrow mononuclear cells first by immunomagnetic microbeads and then by flow cytometry autoclone sorting. The cells were then cultivated under different conditions. The long-term hematopoietic supporting ability of expanded CD34(+)CD59(+) cells was evaluated by long-term culture in semi-solid medium in vitro and long-term engraftment in irradiated severe combined immunodeficiency (SCID) mice in vivo. The best combination of hematopoietic growth factors for ex vivo expansion was SCF + IL-3 + IL-6 + FL + Tpo + Epo. The most suitable time for harvest was on day 7. CD34(+)CD59(+) PNH cells retained strong colony-forming capacity even after expansion. The survival rate, complete blood cell count recovery on day 90, and human CD45 expression in different organs were similar between the irradiated SCID mice transplanted with expanded CD34(+)CD59(+) PNH cells and those with normal CD34(+) cells (P > 0.05) both in primary and secondary transplantation. These data provided a new potential way of managing PNH with ABMT.
Collapse
Affiliation(s)
- Juan Xiao
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academe of Medical Science, Beijing, China
| | | | | | | | | |
Collapse
|
11
|
Abstract
Pathogen-specific complement activation requires direct recognition of pathogens and/or the absence of complement control mechanisms on their surfaces. Antibodies direct complement activation to potential pathogens recognized by the cellular innate and adaptive immune systems. Similarly, the plasma proteins MBL and ficolins direct activation to microorganisms expressing common carbohydrate structures. The absence of complement control proteins permits amplification of complement by the alternative pathway on any unprotected surface. The importance of complement recognition molecules (MBL, ficolins, factor H, C3, C1q, properdin, and others) to human disease are becoming clear as analysis of genetic data and knock out animals reveals links between complement proteins and specific diseases.
Collapse
|
12
|
Wang SA, Pozdnyakova O, Jorgensen JL, Medeiros LJ, Stachurski D, Anderson M, Raza A, Woda BA. Detection of paroxysmal nocturnal hemoglobinuria clones in patients with myelodysplastic syndromes and related bone marrow diseases, with emphasis on diagnostic pitfalls and caveats. Haematologica 2008; 94:29-37. [PMID: 19001281 DOI: 10.3324/haematol.13601] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The presence of paroxysmal nocturnal hemoglobinuria clones in the setting of aplastic anemia or myelodysplastic syndrome has been shown to have prognostic and therapeutic implications. However, the status of paroxysmal nocturnal hemoglobinuria clones in various categories of myelodysplastic syndrome and in other bone marrow disorders is not well-studied. DESIGN AND METHODS By using multiparameter flow cytometry immunophenotypic analysis with antibodies specific for four glycosylphosphatidylinositol-anchored proteins (CD55, CD59, CD16, CD66b) and performing an aerolysin lysis confirmatory test in representative cases, we assessed the paroxysmal nocturnal hemoglobinuria-phenotype granulocytes in 110 patients with myelodysplastic syndrome, 15 with myelodysplastic/myeloproliferative disease, 5 with idiopathic myelofibrosis and 6 with acute myeloid leukemia. RESULTS Paroxysmal nocturnal hemoglobinuria-phenotype granulocytes were detected in nine patients with low grade myelodysplastic syndrome who showed clinicopathological features of bone marrow failure, similar to aplastic anemia. All paroxysmal nocturnal hemoglobinuria-positive cases demonstrated loss of the four glycosylphosphatidylinositol-anchored proteins, with CD16(-)CD66b(-) clones being larger than those of CD55(-)CD59(-) (p<0.05). Altered glycosylphosphatidylinositol-anchored protein expression secondary to granulocytic hypogranulation, immaturity, and/or immunophenotypic abnormalities was present in a substantial number of cases and diagnostically challenging. CONCLUSIONS These results show that routine screening for paroxysmal nocturnal hemoglobinuria clones in patients with an intrinsic bone marrow disease who show no clinical evidence of hemolysis has an appreciable yield in patients with low grade myelodysplastic syndromes. The recognition of diagnostic caveats and pitfalls associated with the underlying intrinsic bone marrow disease is essential in interpreting paroxysmal nocturnal hemoglobinuria testing correctly. In our experience, the CD16/CD66b antibody combination is superior to CD55/CD59 in screening for subclinical paroxysmal nocturnal hemoglobinuria because it detects a large clone size and is less subject to analytical interference.
Collapse
Affiliation(s)
- Sa A Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Hernández-Campo PM, Almeida J, Orfao A. Hemoglobinuria paroxística nocturna. Med Clin (Barc) 2008; 131:617-30. [DOI: 10.1157/13127921] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
14
|
Oxidative status of red blood cells, neutrophils, and platelets in paroxysmal nocturnal hemoglobinuria. Exp Hematol 2008; 36:369-77. [DOI: 10.1016/j.exphem.2007.12.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2007] [Revised: 11/27/2007] [Accepted: 12/05/2007] [Indexed: 02/01/2023]
|
15
|
Noji H, Shichishima T, Okamoto M, Shichishima-Nakamura A, Matsumoto H, Tajima H, Ogawa K, Maruyama Y. Microvascular thrombosis in the hepatic vein of a patient with paroxysmal nocturnal hemoglobinuria. Int J Hematol 2007; 86:216-21. [PMID: 17988986 DOI: 10.1532/ijh97.07064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is characterized by complement-mediated hemolysis, venous thrombosis, and bone marrow failure. In May 2003, a 33-year-old man was admitted to a hospital with right hypochondralgia and fever. He had a history of aplastic anemia. The patient's diagnosis of diffuse microvessel thrombosis in the hepatic vein due to an unknown cause was derived from the findings of a contrast-enhanced computed tomography examination of the abdominal region, angiographic evaluation of abdominal vessels, and pathohistologic examination of a liver biopsy sample. The patient was subsequently treated with warfarin. The abdominal pain and fever continued, however, and anemia gradually appeared. In April 2004, the patient was referred to our hospital to examine the cause of the thrombosis. On admission, slight anemia and a low serum haptoglobin level were observed. A flow cytometry evaluation of CD55 and/or CD59, CD59, and CD48 expression in erythrocytes, granulocytes, and monocytes, respectively, showed that the respective proportions of negative populations were 5.6%, 97.1%, and 96.2%. The patient then received a diagnosis of aplastic anemia/PNH syndrome, which had caused the hemolytic anemia and thrombosis, although no hemoglobinuria had been observed during his clinical course. This patient is, to our knowledge, the first reported case of a PNH patient with thrombosis present only in hepatic microvessels and not in hepatic large vessels, in spite of the presence of few hemolytic events.
Collapse
Affiliation(s)
- Hideyoshi Noji
- First Department of Internal Medicine, Fukushima Medical University, Fukushima, Japan
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Ferreira VP, Pangburn MK. Factor H mediated cell surface protection from complement is critical for the survival of PNH erythrocytes. Blood 2007; 110:2190-2. [PMID: 17554058 PMCID: PMC1976366 DOI: 10.1182/blood-2007-04-083170] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) cells are partially (type II) or completely (type III) deficient in GPI-linked complement regulatory proteins CD59 and CD55. PNH III erythrocytes circulate 6 to 60 days in vivo. Why these cells are not lysed as rapidly by complement as unprotected foreign cells, which normally lyse within minutes, remains undetermined. Factor H plays a key role in the homeostasis of complement in fluid phase and on cell surfaces. We have recently shown that a recombinant protein encompassing the C-terminus of factor H (rH19-20) specifically blocks cell-surface complement regulatory functions of factor H without affecting fluid-phase control of complement. Here we show that PNH II and III cells become highly susceptible to complement-mediated lysis by nonacidified normal human serum in vitro, when the cell surface complement-regulatory functions of factor H are blocked. The results indicate that cells deficient in surface-bound regulators are protected for extended periods of time by factor H.
Collapse
Affiliation(s)
- Viviana P Ferreira
- Department of Biochemistry, Center for Biomedical Research, University of Texas Health Science Center, Tyler, TX 75708, USA.
| | | |
Collapse
|
17
|
Microvascular thrombosis in the hepatic vein of a patient with paroxysmal nocturnal hemoglobinuria. Int J Hematol 2007. [DOI: 10.1007/bf03006923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
18
|
Hill A, Richards SJ, Hillmen P. Recent developments in the understanding and management of paroxysmal nocturnal haemoglobinuria. Br J Haematol 2007; 137:181-92. [PMID: 17408457 DOI: 10.1111/j.1365-2141.2007.06554.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Paroxysmal nocturnal haemoglobinuria (PNH) has been recognised as a discrete disease entity since 1882. Approximately a half of patients will eventually die as a result of having PNH. Many of the symptoms of PNH, including recurrent abdominal pain, dysphagia, severe lethargy and erectile dysfunction, result from intravascular haemolysis with absorption of nitric oxide by free haemoglobin from the plasma. These symptoms, as well as the occurrence of thrombosis and aplasia, significantly affect patients' quality of life; thrombosis is the leading cause of premature mortality. The syndrome of haemolytic-anaemia-associated pulmonary hypertension has been further identified in PNH patients. There is currently an air of excitement surrounding therapies for PNH as recent therapeutic developments, particularly the use of the complement inhibitor eculizumab, promise to radically alter the symptomatology and natural history of haemolytic PNH.
Collapse
Affiliation(s)
- Anita Hill
- Department of Haematology, Leeds Teaching Hospitals NHS Trust, Great George Street, Leeds, UK.
| | | | | |
Collapse
|
19
|
Abstract
Cell surface proteins containing covalently linked lipids associate with specialized membrane domains. Morphogens like Hedgehog and Wnt use their lipid anchors to bind to lipoprotein particles and employ lipoproteins to travel through tissues. Removal of their lipid anchors or decreasing lipoprotein levels give rise to adverse Hedgehog and Wnt signaling. Some parasites can also transfer their glycosylphosphatidylinositol-anchored surface proteins to host lipoprotein particles. These antigen-loaded lipoproteins spread throughout the circulation, and probably hamper an adequate immune response by killing neutrophils. Together, these findings imply a widespread role for lipoproteins in intercellular transfer of lipid-anchored surface proteins, and may have various physiological consequences. Here, we discuss how lipid-modified proteins may be transferred to and from lipoproteins at the cellular level.
Collapse
Affiliation(s)
- Sylvia Neumann
- Department of Membrane Enzymology, Bijvoet Center and Institute of Biomembranes, Utrecht University, Utrecht, The Netherlands
| | | | | |
Collapse
|
20
|
Hill A, Ridley SH, Esser D, Oldroyd RG, Cullen MJ, Kareclas P, Gallagher S, Smith GP, Richards SJ, White J, Smith RAG, Hillmen P. Protection of erythrocytes from human complement–mediated lysis by membrane-targeted recombinant soluble CD59: a new approach to PNH therapy. Blood 2006; 107:2131-7. [PMID: 16322479 DOI: 10.1182/blood-2005-02-0782] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) results from the expansion of a hematopoietic clone that is deficient in glycosylphosphatidylinositol-anchored molecules. PNH is characterized by chronic hemolysis with acute exacerbations due to the uncontrolled activity of complement on PNH cells, which lack the inhibitor of homologous complement, CD59. Symptoms include severe fatigue, hemoglobinuria, esophageal spasm, erectile dysfunction, and thrombosis. We report the use of a novel synthetically modified recombinant human CD59, rhCD59-P, a soluble protein that attaches to cell membranes. In vitro treatment of PNH erythrocytes with rhCD59-P resulted in levels of CD59 equivalent to normal erythrocytes and effectively protected erythrocytes from complement-mediated hemolysis. The administration of rhCD59-P to CD1 mice resulted in levels of CD59 on erythrocytes, which protected them from complement-mediated lysis. Thus, rhCD59-P corrects the CD59 deficiency in vitro and can bind to erythrocytes in an in vivo murine model, protecting the cells from the activity of human complement, and represents a potential therapeutic strategy in PNH.
Collapse
Affiliation(s)
- Anita Hill
- Haematological Malignancy Diagnostic Service, Leeds Teaching Hospitals National Health Service Trust, Algernon Firth Building, Great George Street, Leeds, United Kingdom, LS1 3EX.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Hernández-Campo PM, Almeida J, Sánchez ML, Malvezzi M, Orfao A. Normal patterns of expression of glycosylphosphatidylinositol-anchored proteins on different subsets of peripheral blood cells: A frame of reference for the diagnosis of paroxysmal nocturnal hemoglobinuria. CYTOMETRY PART B-CLINICAL CYTOMETRY 2006; 70:71-81. [PMID: 16493662 DOI: 10.1002/cyto.b.20087] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Evaluation of the expression of glycosylphosphatidylinositol-anchored membrane proteins (GPI-AP) is currently used for the diagnosis of paroxysmal nocturnal hemoglobinuria (PNH). In this study, we analyzed the amount of expression of a wide variety of GPI-AP in different subsets of hematopoietic cells present in normal peripheral blood (PB), to establish their normal patterns of expression and provide a frame of reference for the definition of the best combination of GPI-AP and PB cell subsets to be applied in the diagnosis and monitoring of PNH. RESULTS Our results show variable patterns of expression of different GPI-AP in distinct subsets of normal PB cells. Combined use of CD55 and CD59 represented the most useful dual-marker combination; however, its utility remained suboptimal for several subsets of leukocytes and for platelets. CONCLUSIONS For some cell subsets such as the neutrophils additional useful markers could be selected from a relatively broad panel (CD16/CD24/CD55/CD59/CD66b/CD157), whereas for other cell subsets the number of useful antigens was either restricted (monocytes: CD14/CD55/CD157; B cells: CD24/CD48/CD52/CD55; CD4+ T cells: CD48/CD52/CD55; eosinophils: CD55/CD59; CD8+ T cells: CD48/CD55) or limited to a single marker (CD48 on CD56low NK cells, CD55 on BDCA3- dendritic cells and CD56high NK cells, and CD59 for red cells), from all antigens analyzed.
Collapse
Affiliation(s)
- Pilar María Hernández-Campo
- Servicio General de Citometría, Centro de Investigación del Cáncer, Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | | | | | | | | |
Collapse
|
22
|
Shaheen SP, Talwalkar SS, Simons R, Yam L. Acute Lymphoblastic Leukemic Transformation in a Patient With Chronic Idiopathic Myelofibrosis and Paroxysmal Nocturnal Hemoglobinuria: A Case Report and Review of the Literature. Arch Pathol Lab Med 2005; 129:96-9. [DOI: 10.5858/2005-129-96-alltia] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Abstract
Leukemic transformation of chronic idiopathic myelofibrosis (CIMF) to acute lymphoblastic leukemia (ALL) is rare. We report a case of a patient with CIMF who developed paroxysmal nocturnal hemoglobinuria (PNH) 2 years after initial presentation. His disease eventually transformed to ALL of precursor B-cell type. In that CIMF and PNH are clonal stem cell disorders with different pathogeneses, there may be an association between them. However, leukemic transformation is a rare sequel of both disorders. Coexistence of CIMF and PNH and subsequent transformation to ALL have, to our knowledge, never been previously reported in the world literature. The simultaneous presentation of CIMF and PNH, complicated by the rare sequela of leukemic transformation, raises important issues with regard to diagnosis and treatment.
Collapse
Affiliation(s)
- Saad Paul Shaheen
- From the Departments of Pathology and Laboratory Medicine (Drs Shaheen and Talwalkar), Medicine (Dr Simons), and Medicine/Hematology (Dr Yam), University of Louisville, Louisville, Ky
| | - Sameer S. Talwalkar
- From the Departments of Pathology and Laboratory Medicine (Drs Shaheen and Talwalkar), Medicine (Dr Simons), and Medicine/Hematology (Dr Yam), University of Louisville, Louisville, Ky
| | - Ruth Simons
- From the Departments of Pathology and Laboratory Medicine (Drs Shaheen and Talwalkar), Medicine (Dr Simons), and Medicine/Hematology (Dr Yam), University of Louisville, Louisville, Ky
| | - Lung Yam
- From the Departments of Pathology and Laboratory Medicine (Drs Shaheen and Talwalkar), Medicine (Dr Simons), and Medicine/Hematology (Dr Yam), University of Louisville, Louisville, Ky
| |
Collapse
|
23
|
Hevessy Z, Nagy B, Kiss F, Kiss A, Kappelmayer J. Mean fluorescence intensity rate is a useful marker in the detection of paroxysmal nocturnal hemoglobinuria clones. Clin Chem Lab Med 2005; 43:919-23. [PMID: 16176170 DOI: 10.1515/cclm.2005.157] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractParoxysmal nocturnal hemoglobinuria (PNH) is an acquired disorder of the pluripotent stem cell resulting from the somatic mutation of the X-linked
Collapse
Affiliation(s)
- Zsuzsa Hevessy
- Department of Clinical Biochemistry and Molecular Pathology, Medical and Health Science Center, University of Debrecen, Nagyerdei krt 98, Debrecen, Hungary
| | | | | | | | | |
Collapse
|
24
|
Sobkeng Goufack E, Mammou S, Scotto B, De Muret A, Maakaroun A, Socie G, Bacq Y. Thrombose des veines hépatiques au cours d’un traitement par infliximab (Remicade®) révélant une hémoglobinurie paroxystique nocturne. ACTA ACUST UNITED AC 2004; 28:596-9. [PMID: 15243394 DOI: 10.1016/s0399-8320(04)95017-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We report the case of a 41-Year-old man presenting with hepatic vein thrombosis (Budd-Chiari syndrome) during Infliximab therapy for ankylosing spondylitis. The systematic work-up revealed paroxysmal nocturnal hemoglobinuria. One Year later the patient was receiving anticoagulation therapy and was in good condition. The role of Infliximab in the development of thrombosis in this patient with rare underlying thrombophilia is discussed.
Collapse
Affiliation(s)
- Enam Sobkeng Goufack
- Service d'Hépatogastroentérologie, Hôpital Trousseau, CHRU de Tours, 37044 Tours Cedex
| | | | | | | | | | | | | |
Collapse
|
25
|
Karadimitris A, Araten DJ, Luzzatto L, Notaro R. Severe telomere shortening in patients with paroxysmal nocturnal hemoglobinuria affects both GPI- and GPI+ hematopoiesis. Blood 2003; 102:514-6. [PMID: 12623835 DOI: 10.1182/blood-2003-01-0128] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A most distinctive feature of paroxysmal nocturnal hemoglobinuria (PNH) is that in each patient glycosylphosphatidylinositol-negative (GPI-) and GPI+ hematopoietic stem cells (HSCs) coexist, and both contribute to hematopoiesis. Telomere size correlates inversely with the cell division history of HSCs. In 10 patients with hemolytic PNH the telomeres in sorted GPI- granulocytes were shorter than in sorted GPI+ granulocytes in 4 cases, comparable in 2 cases, and longer in the remaining 4 cases. Furthermore, the telomeres of both GPI- and GPI+ hematopoietic cells were markedly shortened compared with age-matched controls. The short telomeres in the GPI- cells probably reflect the large number of cell divisions required for the progeny of a single cell to contribute a large proportion of hematopoiesis. The short telomeres of the GPI+ cells indicate that the residual hematopoiesis contributed by these cells is not normal. This epigenetic change is an additional feature shared by PNH and aplastic anemia.
Collapse
|
26
|
Rosenfeld C, Cheever MA, Gaiger A. WT1 in acute leukemia, chronic myelogenous leukemia and myelodysplastic syndrome: therapeutic potential of WT1 targeted therapies. Leukemia 2003; 17:1301-12. [PMID: 12835718 DOI: 10.1038/sj.leu.2402988] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Among clinicians, initial awareness of the Wilms' tumor gene was limited mostly to pediatric oncologists. Almost a decade ago, overexpression of Wilms' tumor 1 (WT1) was observed in adult acute leukemia. Subsequent studies indicated that WT1 overexpression occurs in most cases of acute myelogenous leukemia, acute lymphoblastic leukemia, chronic myelogenous leukemia (CML), and myelodysplastic syndrome (MDS). Limited tissue expression of WT1 in adults suggests that WT1 can be a target for leukemia/MDS therapy. WT1 expression in stem/progenitor cells remains unsettled. However, lack of progenitor cell suppression by WT1 antisense or WT1-specific cytotoxic T cells provide some assurance that WT1 expression in progenitor cells is minimal or absent. Immunotherapy-based WT1 approaches are furthest along in preclinical development. WT1-specific cytotoxic lymphocytes can be generated from normals and leukemic patients. In mice, WT1 vaccines elicit specific immune responses without evidence of tissue damage. In this paper, we review studies validating the immunogenicity of WT1 and propose that leukemia and MDS may be a good clinical model to test the efficacy of a WT1 vaccine.
Collapse
|
27
|
Inoue N, Murakami Y, Kinoshita T. Molecular genetics of paroxysmal nocturnal hemoglobinuria. Int J Hematol 2003; 77:107-12. [PMID: 12627844 DOI: 10.1007/bf02983208] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired hematopoietic stem cell disorder characterized by the clonal expansion of glycosylphosphatidylinositol (GPI)-deficient cells that leads to complement-mediated hemolysis. A somatic mutation in the PIG-A gene involved in GPI biosynthesis causes a deficiency of GPI-anchored proteins. However, it is evident that the clonal expansion of GPI-deficient cells is not caused by only the PIG-A mutation and that other changes should be involved in the development of PNH. Some patients with aplastic anemia (AA) develop PNH. Furthermore, it has been reported that most patients with AA and refractory anemia (RA) who carry HLA-DRB1*15 and show a good response to immunosuppressive therapies have an expanded population of GPI-deficient clones. This finding, together with recent data showing resistance of GPI-deficient cells to cytotoxic cells, suggests that GPI-deficient cells escape immunologic attack and are positively selected in the autoimmune environment. However, GPI-deficient clones found in AA and RA are generally small and do not increase to near-complete dominance. Therefore, 1 or more additional genetic abnormalities that confer the growth phenotype on GPI-deficient cells are probably required for fully developed PNH or so-called florid PNH. The next 10 years should witness the discovery of the molecular mechanisms of immunologic selection and the identification of abnormalities involved in the further clonal expansion of PNH cells.
Collapse
Affiliation(s)
- Norimitsu Inoue
- Department of Molecular Genetics, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka University, Suita, Osaka, Japan
| | | | | |
Collapse
|
28
|
Abstract
Mutations seem to be only one of the mechanisms involved in carcinogenesis; selection of mutated clones is a second crucial mechanism. An evolutionary (darwinian) theory of carcinogenesis can be useful to explain some contradictory observations of epidemiology, and to provide a common theoretical framework for carcinogenesis. In both the selection of species and in carcinogenesis (selection of mutated cells), mutation and selection can be interpreted as necessary and insufficient causes. Selection presupposes competition among clones-that is, survival advantage of the mutated species; without selective forces a mutation is mute, while the lack of mutations makes selective advantage impossible. The identification of carcinogen related fingerprints is ambiguous: it can suggest both a genuine mutational hotspot left by the carcinogenic stimulus (like in tobacco related p53 mutations), and selective advantage of clones whose mutations seem to be not exposure specific (like in the case of aflatoxin). We present several examples of exposures that can increase the risk of cancer in humans not via mutations but through a putative mechanism of clone selection.
Collapse
Affiliation(s)
- P Vineis
- CPO-Piemonte and Università di Torino, Italy.
| | | | | |
Collapse
|
29
|
Vineis P, Miligi L, Crosignani P, Davico L, Fontana A, Masala G, Nanni O, Ramazzotti V, Rodella S, Stagnaro E, Tumino R, Viganò C, Vindigni C, Costantini AS. Delayed infection, late tonsillectomy or adenoidectomy and adult leukaemia: a case-control study. Br J Cancer 2003; 88:47-9. [PMID: 12556958 PMCID: PMC2376795 DOI: 10.1038/sj.bjc.6600689] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
In a population-based case-control study among adults in Italy, of 261 lymphoid and 313 myeloid leukaemias and 1718 controls, a later age at adenoidectomy and tonsillectomy (after age 10 years) increased considerably the risk of lymphocytic (but not myeloid) leukaemia (odds ratio 4.2, 95% confidence interval 1.1-16.2). We propose that late infection is a proliferative stimulus for B-cells.
Collapse
Affiliation(s)
- P Vineis
- Servizio di Epidemiologia dei Tumori, Ospedale S. Giovanni Battista e Università di Torino, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Meletis J, Terpos E, Samarkos M, Meletis C, Apostolidou E, Komninaka V, Anargyrou K, Korovesis K, Mavrogianni D, Variami E, Viniou N, Konstantopoulos K. Red cells with paroxysmal nocturnal hemoglobinuria-phenotype in patients with acute leukemia. Hematology 2002; 7:69-74. [PMID: 12186694 DOI: 10.1080/10245330290028560] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
CD55 and CD59 are complement regulatory proteins that are linked to the cell membrane via a glycosyl-phosphatidylinositol anchor. They are reduced mainly in paroxysmal nocturnal hemoglobinuria (PNH) and in other hematological disorders. However, there are very few reports in the literature concerning their expression in patients with acute leukemias (AL). We studied the CD55 and CD59 expression in 88 newly diagnosed patients with AL [65 with acute non-lymphoblastic leukemia (ANLL) and 23 with acute lymphoblastic leukemia (ALL)] using the sephacryl gel test, the Ham and sucrose lysis tests and we compared the results with patients' clinical data and disease course. Eight patients with PNH were also studied as controls. Red cell populations deficient in both CD55 and CD59 were detected in 23% of ANLL patients (especially of M(0), M(2) and M(6) FAB subtypes), 13% of ALL and in all PNH patients. CD55-deficient erythrocytes were found in 6 ANLL patients while the expression of CD59 was decreased in only 3 patients with ANLL. No ALL patient had an isolated deficiency of these antigens. There was no correlation between the existence of CD55 and/or CD59 deficiency and the percentage of bone marrow infiltration, karyotype or response to treatment. However no patient with M(3), M(5), M(7) subtype of ANLL and mature B- or T-cell ALL showed a reduced expression of both antigens. The deficient populations showed no alteration after chemotherapy treatment or during disease course. This study provides evidence about the lower expression of CD55 and CD59 in some AL patients and the correlation with their clinical data. The possible mechanisms and the significance of this phenotype are discussed.
Collapse
Affiliation(s)
- John Meletis
- First Department of Internal Medicine, University of Athens School of Medicine, Laiko General Hospital, Agiou Thoma 17, 11527, Athens, Greece.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kinoshita T, Inoue N. Relationship between aplastic anemia and paroxysmal nocturnal hemoglobinuria. Int J Hematol 2002; 75:117-22. [PMID: 11939256 DOI: 10.1007/bf02982015] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Since aplastic anemia-paroxysmal nocturnal hemoglobinuria syndrome was reported in 1967, the overlap of idiopathic aplastic anemia (AA) and paroxysmal nocturnal hemoglobinuria (PNH) has been well known. The link between the 2 diseases became even more evident when immunosuppressive therapy improved survival of patients with severe AA. More than 10% of patients with AA develop clinically evident PNH. Moreover, flow cytometric analysis demonstrates that the majority of patients with AA have a subclinical percentage of granulocytes with PNH phenotype. Some of them have clearly recognizable PNH clones. Granulocytes with a PNH phenotype are also often found in normal individuals, though at much smaller percentages of cells. This finding suggests that a PNH clone is expanded in AA. consistent with a hypothesis that blood cells from patients with PNH are more resistant to an autoimmune environment. Survival of PNH clones in pathologic bone marrow may account for limited expansion of PNH clones; however, additional genetic change(s) that confers cells with growth phenotype may be required for the full development of PNH.
Collapse
Affiliation(s)
- Taroh Kinoshita
- Department of Immunoregulation, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | | |
Collapse
|
32
|
De Nardo C, Fonsatti E, Sigalotti L, Calabrò L, Colizzi F, Cortini E, Coral S, Altomonte M, Maio M. Recombinant transmembrane CD59 (CD59-TM) confers complement resistance to GPI-anchored protein defective melanoma cells. J Cell Physiol 2002; 190:200-6. [PMID: 11807824 DOI: 10.1002/jcp.10050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Protectin (CD59) is a glycosylphosphatidylinositol (GPI)-anchored cell membrane glycoprotein, broadly expressed on melanocytic cells, that represents the main restriction factor of complement (C)-mediated lysis of human melanoma cells. Levels of CD59 expression may impair the clinical efficacy of C-activating monoclonal antibodies (mAb); thus, we investigated the molecular mechanisms underlying the lack of CD59 expression in selected melanoma cells. Serological and biochemical analyses showed that MeWo melanoma cells expressed CD59 neither at cell surface nor at cytoplasmic levels; however, no critical mutations were identified in their CD59 mRNA. Consistently, MeWo CD59 cDNA (MeWo-CD59) was appropriately translated when transfected into the CD59-positive Mel 100 melanoma cells, and into the CD59-negative Nalm-6 pre-B leukemia cells that acquired resistance to C. In contrast, transfection of MeWo cells with CD59 cDNA from Mel 275 melanoma cells did not induce CD59 expression; however, their transfection with the CD59-TM chimeric construct, obtained by replacing the GPI-anchoring signal of MeWo-CD59 with the transmembrane tail of the human low-density lipoprotein receptor, induced the expression of a C-protective transmembrane form of CD59. These data, together with the absent expression of additional GPI-anchored proteins (i.e., CD55), suggest that defects in the biosynthesis and/or processing of GPI-anchored proteins underlie the lack of CD59 expression in MeWo cells. Further unveiling of the molecular mechanism that turns off CD59 expression in human melanoma cells will help to set-up more effective therapeutic strategies utilizing C-activating mAb in melanoma patients.
Collapse
Affiliation(s)
- Chiara De Nardo
- Cancer Bioimmunotherapy Unit, Centro di Riferimento Oncologico, Istituto Nazionale di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|