Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Br J Haematol ; 189(5): 954-966, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32103498

RESUMO

A retrospective analysis of presentation clinical, laboratory and immunophenotypic features of 1 081 patients with paroxysmal nocturnal haemoglobinuria (PNH) clones [glycosylphosphatidylinositol (GPI)-deficient blood cells] identified at our hospital by flow cytometry over the past 25 years was undertaken. Three distinct clusters of patients were identified and significant correlations between presentation disease type and PNH clone sizes were evident. Smaller PNH clones predominate in cytopenic and myelodysplastic subtypes; large PNH clones were associated with haemolytic, thrombotic and haemolytic/thrombotic subtypes. Rare cases with an associated chronic myeloproliferative disorder had either large or small PNH clones. Cytopenia was a frequent finding, highlighting bone marrow failure as the major underlying feature associated with the detection of PNH clones in the peripheral blood. Red cell PNH clones showed significant correlations between the presence of type II (partial GPI deficiency) red cells and thrombotic disease. Haemolytic PNH was associated with type III (complete GPI deficiency) red cell populations of >20%. Those with both haemolytic and thrombotic features had major type II and type III red cell populations. Distinct patterns of presentation age decade were evident for clinical subtypes with a peak incidence of haemolytic PNH in the 30-49 year age group and a biphasic age distribution for the cytopenia group.


Assuntos
Glicosilfosfatidilinositóis/deficiência , Hemoglobinúria Paroxística/sangue , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Anemia Aplástica/etiologia , Anemia Hemolítica/etiologia , Antígenos CD55/deficiência , Antígenos CD59/deficiência , Criança , Pré-Escolar , Evolução Clonal , Células Clonais/patologia , Progressão da Doença , Feminino , Citometria de Fluxo , Hemoglobinúria Paroxística/complicações , Hemoglobinúria Paroxística/genética , Hemoglobinúria Paroxística/patologia , Humanos , Imunofenotipagem , Lactente , Linfócitos/patologia , Masculino , Pessoa de Meia-Idade , Transtornos Mieloproliferativos/etiologia , Neutrófilos/patologia , Receptores da Transferrina/sangue , Estudos Retrospectivos , Trombose/etiologia , Adulto Jovem
2.
Am J Pathol ; 189(1): 147-161, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30339839

RESUMO

The terminal complement complex (TCC) is formed on activation of the complement system, a crucial arm of innate immunity. TCC formation on cell membranes results in a transmembrane pore leading to cell lysis. In addition, sublytic TCC concentrations can modulate various cellular functions. TCC-induced effects may play a role in the pathomechanisms of inflammatory disorders of the bone, including rheumatoid arthritis and osteoarthritis. In this study, we investigated the effect of the TCC on bone turnover and repair. Mice deficient for complement component 6 (C6), an essential component for TCC assembly, and mice with a knockout of CD59, which is a negative regulator of TCC formation, were used in this study. The bone phenotype was analyzed in vivo, and bone cell behavior was analyzed ex vivo. In addition, the mice were subjected to a femur osteotomy. Under homeostatic conditions, C6-deficient mice displayed a reduced bone mass, mainly because of increased osteoclast activity. After femur fracture, the inflammatory response was altered and bone formation was disturbed, which negatively affected the healing outcome. By contrast, CD59-knockout mice only displayed minor skeletal alterations and uneventful bone healing, although the early inflammatory reaction to femur fracture was marginally enhanced. These results demonstrate that TCC-mediated effects regulate bone turnover and promote an adequate response to fracture, contributing to an uneventful healing outcome.


Assuntos
Regeneração Óssea , Complexo de Ataque à Membrana do Sistema Complemento , Fraturas do Fêmur , Consolidação da Fratura , Osteoclastos , Animais , Regeneração Óssea/genética , Regeneração Óssea/imunologia , Antígenos CD59/deficiência , Técnicas de Cultura de Células , Complemento C6/deficiência , Complexo de Ataque à Membrana do Sistema Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Eritrócitos/imunologia , Eritrócitos/metabolismo , Eritrócitos/patologia , Fraturas do Fêmur/genética , Fraturas do Fêmur/imunologia , Fraturas do Fêmur/metabolismo , Fraturas do Fêmur/patologia , Consolidação da Fratura/genética , Consolidação da Fratura/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Knockout , Osteoclastos/imunologia , Osteoclastos/metabolismo , Osteoclastos/patologia , Ovinos
3.
Front Immunol ; 9: 1749, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30116241

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired syndrome characterized by intravascular hemolysis, thrombosis, and bone marrow failure. The disease is caused by a mutation in the PIG-A gene that leads to the lack of glycosylphosphatidylinositol-anchored complement regulatory molecules CD55 and CD59 on affected blood cell surfaces. In previous studies, spontaneous clinical remissions have been described. The disease manifestations are very heterogeneous, and we wanted to examine if true remissions and disappearance of the clone occur. In a follow-up of a nation-wide cohort of 106 Finnish patients with a PNH clone, we found six cases, where the clone disappeared or was clearly diminished. Two of the patients subsequently developed leukemia, while the other four are healthy and in clinical remission. According to our data, spontaneous remissions are not as frequent as described earlier. Since the disappearance of the PNH cell clone may indicate either a favorable or a poor outcome-remission or malignancy-careful clinical monitoring in PNH is mandatory. Nevertheless, true remissions occur, and further studies are needed to understand the immunological background of this phenomenon and to obtain a better understanding of the natural history of the disease.


Assuntos
Hemoglobinúria Paroxística/complicações , Hemoglobinúria Paroxística/fisiopatologia , Leucemia/etiologia , Adulto , Anemia Aplástica/complicações , Medula Óssea/fisiopatologia , Antígenos CD59/deficiência , Células Clonais/metabolismo , Estudos de Coortes , Evolução Fatal , Feminino , Finlândia , Citometria de Fluxo , Seguimentos , Hemoglobinúria Paroxística/imunologia , Hospitais Universitários , Humanos , Masculino , Pessoa de Meia-Idade , Remissão Espontânea , Adulto Jovem
4.
Eur J Paediatr Neurol ; 22(5): 870-877, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29843966

RESUMO

CD59 is involved in lymphocyte signal transduction and regulates complement-mediated cell lysis by inhibiting the membrane attack complex. In the cases reported so far, congenital isolated CD59 deficiency was associated with recurrent episodes of hemolytic anemia, peripheral neuropathy, and strokes. Here, we report on a patient from a consanguineous Turkish family, who had a first episode of hemolytic anemia at one month of age and presented at 14 months with acute Guillain-Barré syndrome (GBS). The child suffered repeated infection-triggered relapses leading to the diagnosis of chronic inflammatory demyelinating polyneuropathy (CIDP). Although partly steroid-responsive, the polyneuropathy failed to be stabilized by a number of immunosuppressive agents. At the age of 6 years, he developed acute hemiparesis and showed progressive stenosis of proximal cerebral arteries, evolving into Moyamoya syndrome (MMS) with recurrent infarctions leading to death at 8 years of age. Post-mortem genetic analysis revealed a pathogenic p.(Asp49Valfs*31) mutation in CD59. Re-analysis of brain biopsy specimens showed absent CD59 expression and severe endothelial damage. Whereas strokes are a known feature of CD59 deficiency, MMS has not previously been described in this condition. Therefore, we conclude that in MMS combined with hemolysis or neuropathy CD59 deficiency should be considered. Establishing the diagnosis and targeted therapy with eculizumab might have prevented the lethal course in our patient.


Assuntos
Anemia Hemolítica/complicações , Encéfalo/patologia , Hemoglobinúria/complicações , Doença de Moyamoya/genética , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/genética , Acidente Vascular Cerebral/genética , Anemia Hemolítica/genética , Antígenos CD59/deficiência , Antígenos CD59/genética , Criança , Pré-Escolar , Evolução Fatal , Feminino , Síndrome de Guillain-Barré/genética , Hemoglobinúria/genética , Humanos , Lactente , Recém-Nascido , Masculino , Mutação , Turquia
5.
J Immunol Methods ; 456: 15-22, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29447841

RESUMO

BACKGROUND: To prevent unwanted complement activation and subsequent damage, complement activation must be tightly regulated on healthy host cells. Dysregulation of the complement system contributes to the pathology of diseases like Paroxysmal Nocturnal Hemoglobinuria and atypical Hemolytic Uremic Syndrome. To investigate complement regulator deficiencies, primary patient cells may be used, but access to patient cells may be limited and cells are heterogeneous between different patients. To inhibit regulator function on healthy host cells, blocking antibodies can be used, though it may be difficult to exclude antibody-mediated effects. To circumvent these issues, we created single and combined complement regulator human knockout cells to be able to in vitro investigate complement activation and regulation on human cells. METHODS: CRISPR/Cas9 was used to knockout (KO) complement regulatory proteins CD46, CD55 and/or CD59 in human HAP1 cells. Single cell derived cell lines were profiled by Sanger sequencing and flow cytometry. To confirm the lack of complement regulatory function, the cells were exposed to complement in normal human serum and subsequently C3 and C4 deposition on the cell surface were detected by using flow cytometry. RESULTS: We created single KO cell lines that completely lacked CD46, CD55 or CD59. We additionally generated double CD46/CD55, CD46/CD59 and CD55/CD59 KOs and triple CD46/CD55/CD59 KOs. Upon classical pathway activation, deletion of CD46 resulted in increased C3 and C4 deposition, while deleting CD55 mainly resulted to increased C3 deposition, confirming their reported function in complement regulation. Upon alternative pathway activation, C3 deposition was only observed on the triple CD46/CD55/CD59 KO cells and not on any of the other cell lines, suggesting that human cells are resistant to spontaneous complement activation and suggesting a role for CD59 in C3 regulation. CONCLUSIONS: The generation of complement regulator KO cell lines provides a relevant tool for future in vitro investigations of complement activation and regulation on human cells. Furthermore, these cell lines may also be helpful to evaluate therapeutic complement inhibitors and may shed light on novel roles of complement regulatory proteins as we here observed for CD59.


Assuntos
Antígenos CD55/deficiência , Antígenos CD59/deficiência , Sistemas CRISPR-Cas/genética , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/imunologia , Edição de Genes , Técnicas de Inativação de Genes , Proteína Cofatora de Membrana/deficiência , Linhagem Celular , Voluntários Saudáveis , Humanos , Masculino
6.
Acta Neuropathol Commun ; 5(1): 57, 2017 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-28750658

RESUMO

Pathogenesis in aquaporin-4 immunoglobulin G (AQP4-IgG) seropositive neuromyelitis optica spectrum disorders (herein called NMO) involves complement-dependent cytotoxicity initiated by AQP4-IgG binding to astrocyte AQP4. We recently reported that rats lacking complement inhibitor protein CD59 were highly susceptible to development of NMO pathology in brain and spinal cord following direct AQP4-IgG administration (Yao and Verkman, Acta Neuropath Commun 2017, 5:15). Here, we report evidence that CD59 is responsible for protection of peripheral, AQP4-expressing tissues in seropositive NMO. Rats made seropositive by intraperitoneal injection of AQP4-IgG developed marked weakness by 24 h and died soon thereafter. Serum creatine phosphokinase at 24 h was >900-fold greater in seropositive CD59-/- rats than in seropositive CD59+/+ (or control) rats. AQP4-expressing cells in skeletal muscle and kidney, but not in stomach, of seropositive CD59-/- rats showed injury with deposition of AQP4-IgG and activated complement C5b-9, and inflammation. Organ injury in seropositive CD59-/- rats was prevented by a complement inhibitor. Significant pathological changes in seropositive CD59-/- rats were not seen in optic nerve, spinal cord or brain, including circumventricular tissue. These results implicate a major protective role of CD59 outside of the central nervous system in seropositive NMO, and hence offer an explanation as to why peripheral, AQP4-expressing cells are largely unaffected in NMO.


Assuntos
Antígenos CD59/deficiência , Neuromielite Óptica/metabolismo , Animais , Aquaporina 4/imunologia , Aquaporina 4/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Antígenos CD59/genética , Creatina Quinase/sangue , Modelos Animais de Doenças , Mucosa Gástrica/metabolismo , Humanos , Imunoglobulina G/imunologia , Rim/metabolismo , Rim/patologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Neuromielite Óptica/patologia , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Ratos Sprague-Dawley , Ratos Transgênicos , Proteínas Recombinantes/imunologia , Medula Espinal/metabolismo , Medula Espinal/patologia , Estômago/patologia
7.
J Ocul Pharmacol Ther ; 33(5): 400-411, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28333572

RESUMO

PURPOSE: The membrane attack complex (MAC) in choriocapillaris (CC) and retinal pigment epithelium (RPE) increase with age and disease (age-related macular degeneration). MAC assembly can be inhibited by CD59, a membrane-bound regulator. Here we further investigated the role of CD59 in murine choroidal neovascularization (CNV), a model involving both CC and RPE, and tested whether CR2-CD59, a soluble targeted form of CD59, provides protection. METHODS: Laser-induced CNV was generated in wild type and CD59a-deficient mice (CD59-/-). CNV size was measured by optical coherence tomography, and CR2-CD59 was injected intraperitoneally. Endogenous CD59 localization and MAC deposition were identified by immunohistochemistry and quantified by confocal microscopy. Cell-type-specific responses to MAC were examined in retinal pigment epithelial cells (ARPE-19) and microvascular endothelial cells (HMEC-1). RESULTS: CD59 levels were severely reduced and protein was mislocalized in the RPE surrounding the lesion. CNV lesion size and subretinal fluid accumulation were exacerbated in CD59-/- when compared with those in WT mice, and an increase in MAC deposition was noted. In contrast, CR2-CD59 significantly reduced both structural features of CNV severity. In vitro, MAC inhibition in ARPE-19 cells prevented barrier function loss and accelerated wound healing and cell adhesion, whereas in HMEC-1 cells, CR2-CD59 decelerated wound healing and cell adhesion. CONCLUSION: These data further support the importance of CD59 in controlling ocular injury responses and indicate that pharmacological inhibition of the MAC with CR2-CD59 may be a viable therapeutic approach for reducing complement-mediated ocular pathology.


Assuntos
Antígenos CD59/metabolismo , Neovascularização de Coroide/tratamento farmacológico , Inativadores do Complemento/farmacologia , Células Endoteliais/patologia , Lasers , Receptores de Complemento 3d/metabolismo , Animais , Antígenos CD59/administração & dosagem , Antígenos CD59/deficiência , Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/patologia , Inativadores do Complemento/administração & dosagem , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Complemento 3d/administração & dosagem , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia
8.
Acta Neuropathol Commun ; 5(1): 15, 2017 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-28212662

RESUMO

Neuromyelitis optica spectrum disorders (herein called NMO) is an inflammatory demyelinating disease of the central nervous system in which pathogenesis involves complement-dependent cytotoxicity (CDC) produced by immunoglobulin G autoantibodies targeting aquaporin-4 (AQP4-IgG) on astrocytes. We reported evidence previously, using CD59-/- mice, that the membrane-associated complement inhibitor CD59 modulates CDC in NMO (Zhang and Verkman, J. Autoimmun. 53:67-77, 2014). Motivated by the observation that rats, unlike mice, have human-like complement activity, here we generated CD59-/- rats to investigate the role of CD59 in NMO and to create NMO pathology by passive transfer of AQP4-IgG under conditions in which minimal pathology is produced in normal rats. CD59-/- rats generated by CRISPR/Cas9 technology showed no overt phenotype at baseline except for mild hemolysis. CDC assays in astrocyte cultures and cerebellar slices from CD59-/- rats showed much greater sensitivity to AQP4-IgG and complement than those from CD59+/+ rats. Intracerebral administration of AQP4-IgG in CD59-/- rats produced marked NMO pathology, with astrocytopathy, inflammation, deposition of activated complement, and demyelination, whereas identically treated CD59+/+ rats showed minimal pathology. A single, intracisternal injection of AQP4-IgG in CD59-/- rats produced hindlimb paralysis by 3 days, with inflammation and deposition of activated complement in spinal cord, optic nerves and brain periventricular and surface matter, with most marked astrocyte injury in cervical spinal cord. These results implicate an important role of CD59 in modulating NMO pathology in rats and demonstrate amplification of AQP4-IgG-induced NMO disease with CD59 knockout.


Assuntos
Aquaporina 4/imunologia , Antígenos CD59/deficiência , Imunoglobulina G/imunologia , Neuromielite Óptica/imunologia , Neuromielite Óptica/patologia , Animais , Aquaporina 4/administração & dosagem , Encéfalo/imunologia , Encéfalo/patologia , Antígenos CD59/genética , Sistemas CRISPR-Cas , Células Cultivadas , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Humanos , Imunoglobulina G/administração & dosagem , Neurônios/imunologia , Neurônios/patologia , Nervo Óptico/imunologia , Nervo Óptico/patologia , Paralisia/imunologia , Paralisia/patologia , Ratos Sprague-Dawley , Ratos Transgênicos , Medula Espinal/imunologia , Medula Espinal/patologia , Técnicas de Cultura de Tecidos
9.
J Diabetes Complications ; 31(2): 311-317, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27729184

RESUMO

AIMS: Clinical and experimental evidence supports a strong link between the complement system, complement regulatory proteins and the pathogenesis of diabetes vascular complications. We previously reported that the complement regulatory protein CD59 is inactivated by glycation in humans with diabetes. Our objective for this study is to assess experimentally how the deficiency of CD59 impacts the development of diabetic atherosclerosis in vivo. METHODS: We crossed mCD59 sufficient and deficient mice into the ApoE-/- background to generate mCd59ab+/+/ApoE-/- and mCd59ab-/-/ApoE-/- mice, and induced diabetes by multiple low dose injections of streptozotocin. Atherosclerosis was detected by hematoxylin and eosin (H&E) and oil red-O staining. Membrane attack complex (MAC) deposition and macrophage infiltration were detected by immunostaining. RESULTS: Diabetic mCD59 deficient (mCD59ab-/-/ApoE-/-) mice developed nearly 100% larger atherosclerotic lesion areas in the aorta (7.5%±0.6 vs 3.6%±0.7; p<0.005) and in the aortic roots (H&E: 26.2%±1.9 vs. 14.3%±1.1; p<0.005), in both cases associated with increased lipid (Oil red-O: 14.9%±1.1 vs. 7.8%±1.1; p<0.05) and MAC deposition (6.8%±0.8 vs. 3.0%±0.7; p<0.005) and macrophage infiltration (31.5%±3.7 vs. 16.4%±3.0; p<0.05) in the aortic roots as compared to their diabetic mCD59 sufficient (mCD59ab+/+/ApoE-/-) counterpart. CONCLUSIONS: The deficiency of CD59 accelerates the development of diabetic atherosclerosis.


Assuntos
Apolipoproteínas E/metabolismo , Aterosclerose/metabolismo , Antígenos CD59/metabolismo , Diabetes Mellitus Tipo 1/complicações , Angiopatias Diabéticas/metabolismo , Endotélio Vascular/metabolismo , Animais , Aorta , Apolipoproteínas E/genética , Aterosclerose/complicações , Aterosclerose/imunologia , Aterosclerose/patologia , Glicemia/análise , Antígenos CD59/deficiência , Antígenos CD59/genética , Ativação do Complemento/efeitos dos fármacos , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Cruzamentos Genéticos , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/induzido quimicamente , Angiopatias Diabéticas/imunologia , Angiopatias Diabéticas/patologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Ativação de Macrófagos/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estreptozocina/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA