Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Immunol Methods ; 456: 15-22, 2018 05.
Article in English | MEDLINE | ID: mdl-29447841

ABSTRACT

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.


Subject(s)
CD55 Antigens/deficiency , CD59 Antigens/deficiency , CRISPR-Cas Systems/genetics , Complement System Proteins/genetics , Complement System Proteins/immunology , Gene Editing , Gene Knockout Techniques , Membrane Cofactor Protein/deficiency , Cell Line , Healthy Volunteers , Humans , Male
2.
Transplantation ; 102(5): e195-e204, 2018 05.
Article in English | MEDLINE | ID: mdl-29266033

ABSTRACT

BACKGROUND: We investigated in vitro whether HLA highly sensitized patients with end-stage renal disease will be disadvantaged immunologically after a genetically engineered pig kidney transplant. METHODS: Blood was drawn from patients with a calculated panel-reactive antibody (cPRA) 99% to 100% (Gp1, n = 10) or cPRA 0% (Gp2, n = 12), and from healthy volunteers (Gp3, n = 10). Serum IgM and IgG binding was measured (i) to galactose-α1-3 galactose and N-glycolylneuraminic acid glycans by enzyme-linked immunosorbent assay, and (ii) to pig red blood cell, pig aortic endothelial cells, and pig peripheral blood mononuclear cell from α1,3-galactosyltransferase gene-knockout (GTKO)/CD46 and GTKO/CD46/cytidine monophosphate-N-acetylneuraminic acid hydroxylase-knockout (CMAHKO) pigs by flow cytometry. (iii) T-cell and B-cell phenotypes were determined by flow cytometry, and (iv) proliferation of T-cell and B-cell carboxyfluorescein diacetate succinimidyl ester-mixed lymphocyte reaction. RESULTS: (i) By enzyme-linked immunosorbent assay, there was no difference in IgM or IgG binding to galactose-α1-3 galactose or N-glycolylneuraminic acid between Gps1 and 2, but binding was significantly reduced in both groups compared to Gp3. (ii) IgM and IgG binding in Gps1 and 2 was also significantly lower to GTKO/CD46 pig cells than in healthy controls, but there were no differences between the 3 groups in binding to GTKO/CD46/CMAHKO cells. (iii and iv) Gp1 patients had more memory T cells than Gp2, but there was no difference in T or B cell proliferation when stimulated by any pig cells. The proliferative responses in all 3 groups were weakest when stimulated by GTKO/CD46/CMAHKO pig peripheral blood mononuclear cell. CONCLUSIONS: (i) End-stage renal disease was associated with low antipig antibody levels. (ii) Xenoreactivity decreased with increased genetic engineering of pig cells. (iii) High cPRA status had no significant effect on antibody binding or T-cell and B-cell response.


Subject(s)
Galactosyltransferases/genetics , HLA Antigens/immunology , Kidney Failure, Chronic/surgery , Kidney Transplantation/methods , Membrane Cofactor Protein/genetics , Mixed Function Oxygenases/genetics , Animals , Animals, Genetically Modified , B-Lymphocytes/immunology , Case-Control Studies , Cells, Cultured , Galactosyltransferases/deficiency , Galactosyltransferases/immunology , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/prevention & control , HLA Antigens/blood , Heterografts , Humans , Immunologic Memory , Isoantibodies/blood , Isoantibodies/immunology , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/diagnosis , Kidney Failure, Chronic/immunology , Kidney Transplantation/adverse effects , Lymphocyte Activation , Membrane Cofactor Protein/deficiency , Membrane Cofactor Protein/immunology , Mixed Function Oxygenases/deficiency , Mixed Function Oxygenases/immunology , Sus scrofa , T-Lymphocytes/immunology
3.
Eur J Immunol ; 47(7): 1200-1210, 2017 07.
Article in English | MEDLINE | ID: mdl-28444759

ABSTRACT

IFN-γ-producing T helper 1 (Th1) cell responses mediate protection against infections but uncontrolled Th1 activity also contributes to a broad range of autoimmune diseases. Autocrine complement activation has recently emerged as key in the induction and contraction of human Th1 immunity: activation of the complement regulator CD46 and the C3aR expressed by CD4+ T cells via autocrine generated ligands C3b and C3a, respectively, are critical to IFN-γ production. Further, CD46-mediated signals also induce co-expression of immunosuppressive IL-10 in Th1 cells and transition into a (self)-regulating and contracting phase. In consequence, C3 or CD46-deficient patients suffer from recurrent infections while dysregulation of CD46 signaling contributes to Th1 hyperactivity in rheumatoid arthritis and multiple sclerosis. Here, we report a defect in CD46-regulated Th1 contraction in patients with systemic lupus erythematosus (SLE). We observed that MMP-9-mediated increased shedding of soluble CD46 by Th1 cells was associated with this defect and that inhibition of MMP-9 activity normalized release of soluble CD46 and restored Th1 contraction in patients' T cells. These data may deliver the first mechanistic explanation for the increased serum CD46 levels observed in SLE patients and indicate that targeting CD46-cleaving proteases could be a novel avenue to modulate Th1 responses.


Subject(s)
Lupus Erythematosus, Systemic/immunology , Membrane Cofactor Protein/immunology , Membrane Cofactor Protein/metabolism , Th1 Cells/immunology , Adult , Autoimmunity , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Complement Activation , Female , Humans , Immunity, Innate , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-10/pharmacology , Male , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Membrane Cofactor Protein/blood , Membrane Cofactor Protein/deficiency , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Th1 Cells/drug effects
4.
J Neuroimmunol ; 308: 25-29, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28187911

ABSTRACT

Central nervous system consequences of viral infections are rare, but when they do occur, they are often serious and clinically challenging to manage. Our awareness of the perils of neuroinvasion by viruses is growing: the recently appreciated impact of Ebola and Zika virus infections on CNS integrity, decreases in vaccination coverage for potentially neurotropic viruses such as measles, and increased neurovirulence of some influenza strains collectively highlight the need for a better understanding of the viral-neural interaction. Defining these interactions and how they result in neuropathogenesis is paramount for the development of better clinical strategies, especially given the limited treatment options that are available due to the unique physiology of the brain that limits migration of blood-borne molecules into the CNS parenchyma. In this perspective, we discuss some unique aspects of neuronal viral infections and immune-mediated control that impact the pathogenic outcomes of these infections. Further, we draw attention to an often overlooked aspect of neuropathogenesis research: that lack of overt disease, which is often equated with survival post-infection, likely only scratches the surface of the myriad ways by which neurotropic infections can impair CNS function.


Subject(s)
Central Nervous System Viral Diseases/mortality , Central Nervous System/pathology , Kaplan-Meier Estimate , Animals , Central Nervous System/virology , Central Nervous System Viral Diseases/genetics , Disease Models, Animal , Humans , Interferon-gamma/deficiency , Interferon-gamma/genetics , Membrane Cofactor Protein/deficiency , Membrane Cofactor Protein/genetics , Mice , Mice, Transgenic , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics
5.
Xenotransplantation ; 24(1)2017 01.
Article in English | MEDLINE | ID: mdl-28054735

ABSTRACT

BACKGROUND: To investigate graft survival after full-thickness corneal xenotransplantation from α1,3-galactosyltransferase gene-knockout (GTKO) pigs expressing a human complement regulatory protein (GTKO/CD46 pigs) in rhesus monkeys. METHODS: Rhesus monkeys (n=10) were transplanted with full-thickness corneas from wild-type (WT; n=4) and GTKO/CD46 (n=4) pigs or from monkeys (n=2). All recipient monkeys received post-transplant subconjunctival injections of betamethasone. Corneal grafts were evaluated by slit-lamp. Histopathology, immunohistochemistry of the grafts, and cytokine concentrations in the aqueous humor were tested 6 months after transplantation. Anti-pig IgM/IgG and anti-galactose-α1,3-galactose (Gal) antibodies were determined by flow cytometry and ELISA, respectively. RESULTS: The longest graft survival of WT and GTKO/CD46 xenografts was 157 and 171 days, respectively. There was no significant difference in graft survival between WT and GTKO pig corneas. Anterior synechiae occurred in two recipients of WT and all recipients of GTKO/CD46 grafts. All xenograft recipients developed a retrocorneal membrane, inflammatory cells (CD3+ T lymphocytes) infiltrated the corneal stroma, and in the aqueous humor, IL-6 was increased 6 months after transplantation. Induced antibody responses were documented against Gal and/or non-Gal pig antigens. In contrast, allografts survived >180 days without any rejection, with no increase in cytokines in the aqueous humor, and no elicited serum anti-pig antibodies were detected. CONCLUSIONS: α1,3-galactosyltransferase gene-knockout/CD46 pig corneas were not associated with prolonged graft survival or a reduced antibody response compared with WT pig corneas. The prevention of the development of anterior synechiae and a retrocorneal membrane after corneal xenotransplantation would appear to be important if prolonged corneal xenograft survival is to be achieved.


Subject(s)
Cornea/pathology , Galactosyltransferases/genetics , Graft Rejection/immunology , Heterografts/transplantation , Membrane Cofactor Protein/metabolism , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Cornea/surgery , Corneal Transplantation/methods , Galactosyltransferases/deficiency , Gene Knockout Techniques , Graft Rejection/genetics , Graft Survival/immunology , Macaca mulatta , Membrane Cofactor Protein/deficiency , Swine , Transplantation, Heterologous/methods
6.
Am J Pathol ; 186(8): 2088-2104, 2016 08.
Article in English | MEDLINE | ID: mdl-27295359

ABSTRACT

In the mouse, membrane cofactor protein (CD46), a key regulator of the alternative pathway of the complement system, is only expressed in the eye and on the inner acrosomal membrane of spermatozoa. We noted that although Cd46(-/-) mice have normal systemic alternative pathway activating ability, lack of CD46 leads to dysregulated complement activation in the eye, as evidenced by increased deposition of C5b-9 in the retinal pigment epithelium (RPE) and choroid. A knockout of CD46 induced the following cardinal features of human dry age-related macular degeneration (AMD) in 12-month-old male and female mice: accumulation of autofluorescent material in and hypertrophy of the RPE, dense deposits in and thickening of Bruch's membrane, loss of photoreceptors, cells in subretinal space, and a reduction of choroidal vessels. Collectively, our results demonstrate spontaneous age-related degenerative changes in the retina, RPE, and choroid of Cd46(-/-) mice that are consistent with human dry AMD. These findings provide the exciting possibility of using Cd46(-/-) mice as a convenient and reliable animal model for dry AMD. Having such a relatively straight-forward model for dry AMD should provide valuable insights into pathogenesis and a test model system for novel drug targets. More important, tissue-specific expression of CD46 gives the Cd46(-/-) mouse model of dry AMD a unique advantage over other mouse models using knockout strains.


Subject(s)
Disease Models, Animal , Geographic Atrophy/genetics , Macular Degeneration/genetics , Membrane Cofactor Protein/deficiency , Animals , Blotting, Western , Female , Geographic Atrophy/pathology , Macular Degeneration/pathology , Male , Membrane Cofactor Protein/genetics , Mice , Mice, Knockout
8.
Virus Res ; 155(1): 48-54, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20801174

ABSTRACT

Fiber-substituted Ad serotype 5 vectors containing the fiber protein from Ad serotype 35 (Ad5F35) exhibit properties that render them suitable as a platform for targeted Ad vectors. Ad5F35 vectors do not show apparent tropism in certain organs, including the liver, and they elicit less innate immunity than other vectors after intravenous administration. In order to develop a targeted Ad vector, we previously developed fiber-mutant Ad5F35 vectors containing the integrin binding Arg-Gly-Asn (RGD) motif in the FG or HI loop of the Ad35 fiber knob. Mutant Ad5F35 vectors containing the RGD peptide in the FG or HI loop transduced CD46-negative cells more efficiently in an RGD-dependent manner, as compared to the efficiency achieved with unmodified Ad5F35 vectors (Matsui et al., 2009. Gene Therapy 16, 1050-1057). However, the transduction efficiency of the mutant Ad5F35 vectors in CD46-negative cells remained lower than had been expected. Ad5F35 vectors containing the RGD peptide in the HI or FG loop enabled a 6-fold higher transduction efficiency than that achieved with unmodified Ad5F35 vectors in CD46-negative cells, although this cell type abundantly expresses α(v)-integrins. In the present study, we aimed to enhance the transduction efficiency of fiber-mutant Ad5F35 vectors. To this end, we developed an Ad5F35-vector system in which foreign peptides could be incorporated into regions of FG and HI loops of the Ad35 fiber knob by means of in vitro ligation. Using this Ad5F35-vector system, firefly luciferase-expressing mutant Ad5F35 vectors containing the RGD peptides in both loops (Ad5F35-2xRGD-L2) were constructed. In CD46-negative cells, Ad5F35-2xRGD-L2 showed 12-fold and 3-fold greater transduction efficiency than unmodified Ad5F35 vectors and mutant Ad5F35 vectors containing only one copy of the RGD peptide in the FG or the HI loop. In addition, transduction with Ad5F35-2xRGD-L2 in CD46-negative cells was RGD peptide-dependent. These results indicate that fiber-mutant Ad5F35 vectors, by which foreign peptides can be simultaneously incorporated into both the FG and the HI loops of the Ad35 fiber knob, could be a promising gene delivery vehicle for various gene therapies, and could facilitate basic research efforts such as analyses of gene function.


Subject(s)
Adenoviridae/genetics , Capsid Proteins/metabolism , Genetic Vectors , Oligopeptides/metabolism , Transduction, Genetic , Viral Tropism , Capsid Proteins/chemistry , Cell Line , Humans , Membrane Cofactor Protein/deficiency , Oligopeptides/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombination, Genetic
10.
J Reprod Immunol ; 81(1): 62-73, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19501411

ABSTRACT

The mammalian female reproductive tract has an abundance of complement components, which play a vital role in protection against genital pathogens. Sperm may be protected against complement-mediated damage by complement regulatory proteins, including membrane cofactor protein (CD46), decay accelerating factor (CD55) and CD59. However, sperm from Apodemus (field mice) do not express CD46 protein. The aim of the present study was to determine whether Apodemus sperm may be protected against complement-mediated damage by expression of CD55 and CD59 in the absence of CD46. We demonstrate here that, like Mus musculus mice (house mice), wild-caught Apodemus flavicollis, Apodemus microps and Apodemus sylvaticus mice express both glycosylphosphatidylinositol (GPI)- and transmembrane (TM)-anchored testicular CD55 mRNA transcripts. In Mus, testicular GPI- and TM-CD55 transcripts are generated by two distinct but closely related genes. We show that in contrast to Mus, CD55 isoforms in A. sylvaticus are generated by alternative splicing of a single copy gene. Testicular CD59 mRNA transcripts were also identified in A. flavicollis, A. microps, A. sylvaticus and M. musculus. CD55 and CD59 proteins are broadly distributed on epididymal sperm from wild-caught Apodemus and Mus mice as well as BALB/c mice, with expression on the acrosome, neck and tail. Thus, despite not expressing CD46 protein, Apodemus sperm may be protected against complement-mediated injury in the female genital tract by CD55 and CD59.


Subject(s)
Acrosome/metabolism , CD55 Antigens/metabolism , CD59 Antigens/metabolism , Complement System Proteins/immunology , Spermatozoa/metabolism , Acrosome/diagnostic imaging , Acrosome/immunology , Alternative Splicing , Animals , Base Sequence , CD55 Antigens/genetics , CD55 Antigens/immunology , CD59 Antigens/genetics , CD59 Antigens/immunology , Complement System Proteins/metabolism , Cytoprotection , Cytotoxicity, Immunologic , Glycosylphosphatidylinositols/metabolism , Immunohistochemistry , Male , Membrane Cofactor Protein/deficiency , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Murinae , Sequence Alignment , Spermatozoa/immunology , Spermatozoa/ultrastructure , Ultrasonography
11.
Nephrol Dial Transplant ; 24(9): 2752-4, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19376828

ABSTRACT

We report two cases of children who presented with haemolytic uraemic syndrome following varicella infection. One of them had a membrane cofactor protein mutation, and the other had anti-factor H antibodies. These observations show that infectious agents such as varicella-zoster virus may be the trigger of haemolytic uraemic syndrome in patients with complement dysregulation.


Subject(s)
Autoantibodies/blood , Chickenpox/complications , Chickenpox/immunology , Complement Factor H/antagonists & inhibitors , Hemolytic-Uremic Syndrome/etiology , Hemolytic-Uremic Syndrome/immunology , Membrane Cofactor Protein/deficiency , Chickenpox/genetics , Child, Preschool , Complement Factor H/immunology , Female , Hemolytic-Uremic Syndrome/genetics , Heterozygote , Humans , Male , Membrane Cofactor Protein/genetics , Point Mutation , Time Factors
12.
Pediatr Nephrol ; 24(9): 1757-60, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19373492

ABSTRACT

Patients with atypical haemolytic uremic syndrome (aHUS) with a mutation in the gene encoding membrane cofactor protein (CD46) are known to have a better prognosis than those with mutations in factor H (CFH) or factor I (CFI), but a small number of the former still proceed to end-stage renal failure. Plasma therapy (PE) is the recommended approach to treat both acute episodes and prevent recurrences in aHUS, but studies have yet to show PE efficacy in aHUS associated with a CD46 mutation. The factors determining failure to treatment are not clear and may be related to the mutation involved or to insufficient treatment. Our experience of PE in a family of three sisters with CFH-associated aHUS suggests that intensive and prophylactic PE allows renal function to be maintained in both native kidneys and allografts. The success of this strategy has led us to use it in all cases of aHUS. Here, we describe the effect of this strategy in a child with aHUS and a CD46 mutation. The initial episode was treated with daily PE, resulting in the recovery of renal function. However, over the next 4 years, there was a progressive decline in renal function to end-stage renal failure, with evidence of an on-going thrombotic microangiopathy despite continuous prophylactic PE. Prophylactic PE does not influence the natural course of aHUS and CD46 mutation.


Subject(s)
Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/prevention & control , Membrane Cofactor Protein/genetics , Mutation , Plasma Exchange , Child, Preschool , Hemolytic-Uremic Syndrome/surgery , Humans , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/physiopathology , Kidney Failure, Chronic/surgery , Kidney Transplantation , Male , Membrane Cofactor Protein/deficiency , Membrane Cofactor Protein/metabolism , Purpura, Thrombotic Thrombocytopenic/genetics , Purpura, Thrombotic Thrombocytopenic/physiopathology , Treatment Outcome
13.
Am J Kidney Dis ; 52(2): e5-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18514989

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is a rare thrombotic microangiopathic disorder that may be familial or sporadic. Complement factor H (CFH), factor I, and membrane cofactor protein (MCP; CD46), 3 regulators of the alternative pathway of the complement system activation, have been implicated in this pathological state. To date, 29 different mutations of CD46 have been reported, with incomplete penetrance and better clinical outcome compared with CFH mutations. Of those mutations, only 6 were found to be homozygous (accounting for 8 patients), and 5 resulted in a lack of or dramatically decreased cell-surface CD46 expression. We report here the seventh patient with a null mutation associated with recurrent aHUS. This mutation, a guanine to cytosine substitution in the first nucleotide of intron 2, disrupts a splice donor site. Interestingly, the patient's disease-free sister showed the same homozygous mutation. Extensive analysis of other complement regulatory protein- and polymorphism-associated risk factors did not uncover a difference between the patient and his sister. In conclusion, we describe for the first time a disease-free individual with complete CD46 deficiency, confirming the extremely variable penetrance and genetic complexity of aHUS.


Subject(s)
Gene Expression , Genetic Predisposition to Disease , Hemolytic-Uremic Syndrome/genetics , Membrane Cofactor Protein/deficiency , Membrane Cofactor Protein/genetics , RNA, Messenger/genetics , Siblings , Adolescent , Adult , Female , Hemolytic-Uremic Syndrome/blood , Homozygote , Humans , Male , Mutation , Nephelometry and Turbidimetry , Pedigree , Recurrence , Reverse Transcriptase Polymerase Chain Reaction
14.
Mol Immunol ; 44(7): 1559-68, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17027083

ABSTRACT

Mutations in complement regulatory proteins predispose to the development of aHUS. Approximately 50% of patients bear a mutation in one of three complement control proteins, factor H, factor I, or membrane cofactor protein (MCP; CD46). Another membrane regulator that is closely related to MCP, decay accelerating factor (DAF; CD55) thus far has shown no association with aHUS and continues to be investigated. The goal of this study was to compare the regulatory profile of MCP and DAF and to assess how alterations in MCP predispose to complement dysregulation. We employed a model system of complement activation on Chinese hamster ovary (CHO) cell transfectants. The four regularly expressed isoforms of MCP and DAF inhibited C3b deposition by the alternative pathway. DAF, but not MCP, inhibited the classical pathway. Most patients with MCP-aHUS are heterozygous and express only 25-50% of the wild-type protein. We, therefore, analyzed the effect of reduced levels of wild-type MCP and found that cells with lowered expression levels were less efficient in inhibiting alternative pathway activation. Further, a dysfunctional MCP mutant, expressed at normal levels and identified in five patients with aHUS (S206P), failed to protect against C3b amplification on CHO cells, even if expression levels were increased 10-fold. Our results add new information relative to the necessity for appropriate expression levels of MCP and further implicate the alternative pathway in disease processes such as aHUS.


Subject(s)
CD55 Antigens/metabolism , Complement C3b/metabolism , Hemolytic-Uremic Syndrome/immunology , Membrane Cofactor Protein/deficiency , Models, Biological , Animals , CD55 Antigens/chemistry , CD55 Antigens/genetics , CHO Cells , Cells, Cultured , Complement Factor H/metabolism , Complement Pathway, Classical , Cricetinae , Cricetulus , Genetic Predisposition to Disease , Hemolytic-Uremic Syndrome/genetics , Humans , Membrane Cofactor Protein/chemistry , Membrane Cofactor Protein/genetics , Mutation , Protein Conformation , Protein Isoforms/metabolism
15.
J Am Soc Nephrol ; 17(7): 2017-25, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16762990

ABSTRACT

Hemolytic uremic syndrome (HUS) is characterized by the triad of thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure. The non-Shiga toxin-associated HUS (atypical HUS [aHUS]) has been shown to be a disease of complement dysregulation. Mutations in the plasma complement regulators factor H and factor I and the widely expressed membrane cofactor protein (MCP; CD46) have been described recently. This study looked for MCP mutations in a panel of 120 patients with aHUS. In this cohort, approximately 10% of patients with aHUS (11 patients; nine pedigrees) have mutations in MCP. The onset typically was in early childhood. Unlike patients with factor I or factor H mutations, most of the patients do not develop end-stage renal failure after aHUS. The majority of patients have a mutation that causes reduced MCP surface expression. A small proportion expressed normal levels of a dysfunctional protein. As in other studies, incomplete penetrance is shown, suggesting that MCP is a predisposing factor rather than a direct causal factor. The low level of recurrence of aHUS in transplantation in patients with MCP mutations is confirmed, and the first MCP null individuals are described. This study confirms the association between MCP deficiency and aHUS and further establishes that a deficiency in complement regulation, specifically cofactor activity, predisposes to severe thrombotic microangiopathy in the renal vasculature.


Subject(s)
Hemolytic-Uremic Syndrome/genetics , Membrane Cofactor Protein/genetics , Mutation , Adolescent , Adult , Cell Line , Child , Child, Preschool , Complement Activation , Complement System Proteins/analysis , Exons , Female , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/metabolism , Humans , Infant , Male , Membrane Cofactor Protein/deficiency , Membrane Cofactor Protein/metabolism , Prognosis , Reverse Transcriptase Polymerase Chain Reaction
16.
Kidney Int ; 70(1): 16-23, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16760911

ABSTRACT

Thrombotic microangiopathy, which includes thrombotic thrombocytopenic purpura (TTP), shiga-toxin-associated hemolytic uremic syndrome (Stx-HUS) and atypical HUS, is characterized by the development of hyaline thrombi in the microvasculature resulting in thrombocytopenia, microangiopathic hemolysis, and organ dysfunction. Renal failure is a predominant complication of both Stx-HUS and atypical HUS, whereas neurological complications are more prominent in TTP. Other disorders such as lupus or bone marrow transplantations may occasionally present with features of thrombotic microangiopathy. Recent studies have found autoimmune inhibitors or genetic mutations of a von Willebrand factor (VWF) cleaving metalloprotease ADAMTS13 in patients with TTP. In approximately 30-50% of patients with atypical HUS, mutations have been detected in complement factor H, membrane cofactor protein (CD46), or factor I. All three proteins are involved in the regulation of complement activation. Additionally, autoantibodies of factor H have been described in patients without genetic mutations. These advances illustrate that dysregulation of VWF homeostasis or complement activation owing to genetic or autoimmune mechanisms may lead to the syndrome of thrombotic microangiopathy.


Subject(s)
ADAM Proteins/deficiency , Complement Factor H/deficiency , Hemolytic-Uremic Syndrome/genetics , Membrane Cofactor Protein/deficiency , Purpura, Thrombotic Thrombocytopenic/genetics , ADAM Proteins/genetics , ADAMTS13 Protein , Animals , Complement Factor H/genetics , Hemolytic-Uremic Syndrome/metabolism , Humans , Membrane Cofactor Protein/genetics , Mice , Microcirculation/physiopathology , Purpura, Thrombotic Thrombocytopenic/metabolism
17.
Medicina (B Aires) ; 66 Suppl 3: 2-5, 2006.
Article in Spanish | MEDLINE | ID: mdl-17354469

ABSTRACT

An up-date of the causes and pathogenesis of the HUS is reported. After more than 40 years of research we are able to define the infectious agents and the toxin involved. The mechanisms and the molecules involved in the non-diarrheal (atypical) entities producing HUS have also been characterized. This new situation allows us to develop a diagnostic algorithm that enables us to better define preventive and therapeutic measures, based on more rational evidence.


Subject(s)
Hemolytic-Uremic Syndrome/etiology , ADAM Proteins/deficiency , ADAMTS13 Protein , Algorithms , Complement Activation/physiology , Complement Factor H/deficiency , Glomerulonephritis/complications , Graft Rejection/complications , Hemolytic Agents/adverse effects , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/metabolism , Humans , Membrane Cofactor Protein/deficiency , Purpura, Thrombotic Thrombocytopenic/complications , Shiga Toxin/metabolism , von Willebrand Factor/metabolism
18.
Medicina (B.Aires) ; 66(supl.2): 2-5, 2006. tab
Article in Spanish | BINACIS | ID: bin-123544

ABSTRACT

An up-date of the causes and pathogenesis of the HUS is reported. After more than 40 years of research we are able to define the infectious agents and the toxin involved. The mechanisms and the molecules involved in the non-diarrheal (atypical) entities producing HUS have also been characterized. This new situation allows us to develop a diagnostic algorithm that enables us to better define preventive and therapeutic measures, based on more rational evidence.(AU)


Subject(s)
Humans , Hemolytic-Uremic Syndrome/etiology , ADAM Proteins/deficiency , Algorithms , Membrane Cofactor Protein/deficiency , Complement Activation/physiology , Complement Factor H/deficiency , Glomerulonephritis/complications , Graft Rejection/complications , Hemolytic Agents/adverse effects , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/metabolism , Purpura, Thrombotic Thrombocytopenic/complications , Shiga Toxin/metabolism , von Willebrand Factor/metabolism
19.
Springer Semin Immunopathol ; 27(3): 345-58, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16200405

ABSTRACT

In the past 20 years, our understanding of the workings of complement regulatory protein, CD46 (membrane cofactor protein), has grown as has the impressive list of pathogens interacting with this membrane-bound complement inhibitor. Referred to as a "pathogen magnet," CD46 serves as a receptor for seven human pathogens. Initially discovered as a widely expressed C3b- and C4b-binding protein, it was subsequently shown to be a cofactor for the serine protease factor I to inactivate by limited proteolysis these two opsonins and components of the convertases. The involvement of CD46 in reproductive processes continues to be an emerging story. It is a protector of placental tissue, but it may also play a more direct role in reproduction through its expression on the inner acrosomal membrane of spermatozoa. Cross-linking CD46 with antibodies or natural or pathogenic ligands induces rapid turnover and signaling events. In this regard, much attention is currently focused on generating human T lymphocyte regulatory cells by cross-linking CD46. Finally, highlighting its importance in protecting cells against excessive complement activation is the discovery that even a heterozygous deficiency of CD46 predisposes to hemolytic uremic syndrome.


Subject(s)
Membrane Cofactor Protein/immunology , Amino Acid Sequence , Animals , Complement Activation , Female , Gene Expression , Humans , Male , Membrane Cofactor Protein/chemistry , Membrane Cofactor Protein/deficiency , Membrane Cofactor Protein/genetics , Models, Molecular , Molecular Sequence Data , Pregnancy , Receptors, Virus/immunology , Reproduction/immunology , T-Lymphocytes, Regulatory/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...