Try a new search

Format these results:

Searched for:

person:nardim01

Total Results:

112


A new α1-globin mutation, Hb Brugg [α20(B1)His→Gln] [Case Report]

Rizzi, Mattia; Zurbriggen, Karin; Schmid, Marlis; Goede, Jeroen S; Nardi, Michael A; Schmugge, Markus; Speer, Oliver
A 2½-year-old male child and a 23-year-old woman with no clinical symptoms were investigated during routine consultations. Cation exchange high performance liquid chromatography (HPLC) revealed an additional peak eluting before Hb A. DNA sequencing showed a novel heterozygous mutation at codon 20 of the α1-globin gene. The hemoglobin (Hb) variant was named Hb Brugg. Analysis of oxygen affinity Hb and Hb stability did not show any changes compared to normal Hb constellation.
PMID: 21797708
ISSN: 1532-432x
CID: 4709552

Specific cross-reaction of anti-dsDNA antibody with platelet integrin GPIIIa49-66

Zhang, Wei; Dang, Suying; Wang, Jianhui; Nardi, Michael A; Zan, Hong; Casali, Paolo; Li, Zongdong
Anti-platelet autoantibodies are frequently found in systemic lupus erythematosus (SLE) patients and contribute to the development of SLE-associated immunologic thrombocytopenia (SLE-ITP). Although the correlation of anti-dsDNA autoantibody with platelet-associated antibody has been reported, the potential mechanism underlying such a correlation is incompletely understood. We have reported that anti-platelet integrin GPIIIa49-66 (CAPESIEFPVSEARVLED) autoantibodies play a major role in the development of HIV-1-related thrombocytopenia (HIV-1-ITP). The strong negative charge of GPIIIa49-66 prompts us to investigate whether GPIIIa49-66 can be an epitope mimicking dsDNA. We report here that anti-GPIIIa49-66 antibodies are found in three out of nine SLE-ITP patients. Double-stranded (ds) DNA competitively inhibited the binding of purified patient anti-dsDNA antibodies to GPIIIa49-66 peptide. Both polyclonal and monoclonal anti-GPIIIa49-66 antibodies are able to cross-react with dsDNA. Consistent with previous reports, the DNA binding activities of anti-GPIIIa49-66 antibodies are mainly dependent on the positively charged amino acid in the heavy-chain complementarity-determining region 3 (HCDR3). The HCDR3 of human SLE anti-dsDNA monoclonal antibody (mAb) 412.67 demonstrates a similar positively charged amino acid chain orientation compared with that of anti-GPIIIa49-66 mAb A11, and it cross-reacts with GPIIIa49-66 peptide. Purified anti-GPIIIa49-66 antibodies from SLE-ITP patients are able to induce platelet fragmentation in vitro and to induce thrombocytopenia in vivo. Thus, our data suggest that specific epitope cross-reaction between GPIIIa49-66 and dsDNA could be a mechanism involved in the development of SLE-associated thrombocytopenia
PMCID:4702250
PMID: 20828249
ISSN: 1607-842x
CID: 114820

The Effect of Anti GPIIIa49 66 Antibody on Megakaryocyte Differentiation [Meeting Abstract]

Wang, Jianhui; Pan, Ruimin; Nardi, Michael A.; Li, Zongdong
ISI:000285025201534
ISSN: 0006-4971
CID: 130853

Dissolution of arterial platelet thrombi in vivo with a bifunctional platelet GPIIIa49-66 ligand which specifically targets the platelet thrombus

Zhang, Wei; Li, Yong-Sheng; Nardi, Michael A; Dang, Suying; Yang, Jing; Ji, Yong; Li, Zongdong; Karpatkin, Simon; Wisniewski, Thomas
Patients with HIV-1 immune-related thrombocytopenia have a unique antibody (Ab) against integrin GPIIIa49-66 capable of inducing oxidative platelet fragmentation via Ab activation of platelet nicotinamide adenine dinucleotide phosphate oxidase and 12-lipoxygenase releasing reactive oxygen species. Using a phage display single-chain antibody (scFv) library, we developed a novel human monoclonal scFv Ab against GPIIIa49-66 (named A11) capable of inducing fragmentation of activated platelets. In this study, we investigated the in vivo use of A11. We show that A11 does not induce significant thrombocytopenia or inhibit platelet function. A11 can prevent the cessation of carotid artery flow produced by induced artery injury and dissolve the induced thrombus 2 hours after cessation of blood flow. In addition, A11 can prevent, as well as ameliorate, murine middle cerebral artery stroke, without thrombocytopenia or brain hemorrhage. To further optimize the antithrombotic activity of A11, we produced a bifunctional A11-plasminogen first kringle agent (SLK), which homes to newly deposited fibrin strands within and surrounding the platelet thrombus, reducing effects on nonactivated circulating platelets. Indeed, SLK is able to completely reopen occluded carotid vessels 4 hours after cessation of blood flow, whereas A11 had no effect at 4 hours. Thus, a new antithrombotic agent was developed for platelet thrombus clearance
PMCID:2953838
PMID: 20525921
ISSN: 1528-0020
CID: 114164

C-terminal ADAMTS-18 fragment induces oxidative platelet fragmentation, dissolves platelet aggregates and protects against carotid artery occlusion and cerebral stroke

Li, Zongdong; Nardi, Michael A; Li, Yong-Sheng; Zhang, Wei; Pan, Ruimin; Dang, Suying; Yee, Herman; Quartermain, David; Jonas, Saran; Karpatkin, Simon
Anti-platelet integrin GPIIIa49-66 Ab induces complement-independent platelet oxidative fragmentation and death by generation of platelet peroxide following NADPH oxidase activation. A C-terminal 385 amino acid fragment of ADAMTS-18 (A Disintegrin Metalloproteinase with Thrombospondin motifs produced in endothelial cells) induces oxidative platelet fragmentation in an identical kinetic fashion as anti-GPIIIa49-66 Ab. Endothelial cell ADAMTS-18 secretion is enhanced by thrombin and activated by thrombin cleavage to fragment platelets. Platelet aggregates produced ex vivo with ADP and fibrinogen are destroyed by the C-terminal ADAMTS-18 fragment. Anti-ADAMTS-18 Ab shortens the tail vein bleeding time. The C-terminal fragment protects against FeCl3 induced carotid artery thrombosis as well as cerebral infarction in a post-ischemic stroke model. Thus, a new mechanism is proposed for platelet thrombus clearance, via platelet oxidative fragmentation induced by thrombin cleavage of ADAMTS-18
PMCID:2699219
PMID: 19218546
ISSN: 1528-0020
CID: 93983

Clinical and molecular genetic analysis of a family with macrothrombocytopenia and early onset sensorineural hearing loss

Mhatre, Anand N; Janssens, Sandra; Nardi, Michael A; Li, Yan; Lalwani, Anil K
A kindred with inherited macrothrombocytopenia (MTCP) and sensorineural hearing loss (SNHL) from Ghent, Belgium was identified. Currently, joint expression of MTCP and hearing loss are linked to mutations within MYH9 only. Thus, we tested the hypothesis that a mutation within MYH9 is responsible for the autosomal dominant inheritance of MTCP and hearing loss in the Ghent family. A mutation screen of MYH9 coding region including its intron-exon junctions, as well as common hearing loss genes GJB2, GJB3, and GJB6, was performed. However, no pathogenic sequence alteration was identified. Patients' leukocytes were determined to be normal for NMMHC-A distribution via immunofluorescence analysis and free of Dohle body-like inclusions, identified as aggregates of mutant NMHC-IIA in MYH9 disorders. Also, western blot analysis with anti-NMHC-IIA antibody identified a single 220 kDa immunoreactive band with normal expression level of NMHC-IIA within the platelets and leukocytes of the affected family members. The immunoblot analysis eliminates the possibility of a large deletion within MYH9 that can escape detection by direct sequencing. Collectively, these results suggest that molecular genetic etiology of the Ghent family disorder may be due to as yet unidentified gene whose mutation(s) yields a phenocopy of the MYH9-related disease
PMID: 19285578
ISSN: 1878-0849
CID: 101277

Role of molecular mimickry of hepatitis C-virus (HCV) protein with platelet GPIIIa in hepatitis C-related immunologic thrombocytopenia

Zhang, Wei; Nardi, Michael A; Borkowsky, William; Li, Zongdong; Karpatkin, Simon
HIV-1-ITP patients have a unique Ab against platelet GPIIIa49-66 capable of inducing oxidative platelet fragmentation in the absence of complement. HIV-1-seropositive drug abusers are more prone to develop immune thrombocytopenia (HIV-1-ITP) than non-drug abusers and have a higher coinfection with Hepatitis C virus than non-drug abusers (90% vs 30%). Molecular mimickry with a Hepatitis C protein was sought by screening a phage peptide library with anti-GPIIIa49-66 antibody as bait for peptides sharing homology sequences with HCV protein. Several phage peptide clones had 70% homology with HCV protein. Sera from dually infected thrombocytopenic patients with HCV and HIV-ITP reacted strongly with 4 non-conserved peptides from HCV core-envelope 1. Reactivity correlated inversely with platelet count, r(2)=0.7, p<0.01. Ab raised against peptide PHC09 in GPIIIa(-/-) mice induced severe thrombocytopenia in wild-type mice. Affinity-purified IgG against PHC09 induced oxidative platelet fragmentation in vitro. Drug abusers dually infected with HCV and HIV-1 had a greater incidence and severity of thrombocytopenia as well as greater incidence and titer of anti-GPIIIa49-66/PHC09 Ab. NZB/W F1 mice injected with recombinant core envelope 1 developed Ab vs PHC09 and significantly decreased their platelet count, p<0.001. Thus, HCV core envelope 1 can induce thrombocytopenia by molecular mimicry with GPIIIa49-66
PMCID:2673130
PMID: 19023115
ISSN: 1528-0020
CID: 95182

Role of Molecular Mimickry of Hepatitis C Virus (HCV) Protein with Platelet GPIIIa in Hepatitis C-Related Immunologic Thrombocytopenia [Meeting Abstract]

Zhang, W; Nardi, MA; Borkowsky, W; Karpatkin, S
ISI:000262104700399
ISSN: 0006-4971
CID: 93283

The Orphan Receptor GPR31 Is the Platelet and HUVEC Receptor for 12(S)-HETE [Meeting Abstract]

Feinmark, SJ; Nardi, MA; Harleton, E; Hu, L; Pan, R; Karpatkin, S
ISI:000262104700414
ISSN: 0006-4971
CID: 93284

Platelet fragmentation requires a specific structural conformation of human monoclonal antibody against beta3 integrin

Li, Zongdong; Nardi, Michael A; Wu, Jing; Pan, Ruimin; Zhang, Wei; Karpatkin, Simon
We have described an autoantibody against beta3 (GPIIIa49-66), a region of platelet integrin alphaIIbbeta3 that is unique. It induces platelet fragmentation in the absence of complement via antibody activation of platelet NADPH oxidase and 12-lipoxygenase to release reactive oxygen species, which destroy platelets. To study the mechanism of anti-GPIIIa antibody-induced platelet fragmentation, we screened a human single chain Fv antibody library with the GPIIIa49-66 peptide. Nine monoclonal antibodies were identified that were capable of binding to GPIIIa49-66. Surprisingly, binding avidity for GPIIIa49-66 did not correlate with activity of induction of platelet fragmentation. We therefore investigated the requirements for platelet fragmentation. Mutations were introduced into the heavy chain complementary-determining region-3 of clones 11, 43, and 54 by site-directed mutagenesis. The capability of these clones to induce platelet fragmentation or bind to GPIIIa49-66 subsequently changed. Molecular modeling of these clones with their mutants revealed that the ability to induce platelet fragmentation is affected by the side chain orientation of positively charged amino acids in the heavy chain of residues 99-102. Thus, a structural change in the conformation of anti-GPIIIa49-66 antibody contributes to its binding to the beta3 integrin and subsequent antibody-induced platelet fragmentation and aggregate dissolution
PMID: 18056258
ISSN: 0021-9258
CID: 76762