scholarly journals Current Anti-HPA-1a Standard Antibodies React with the β3 Integrin Subunit but not with αIIbβ3 and αvβ3 Complexes

2019 ◽  
Vol 119 (11) ◽  
pp. 1807-1815 ◽  
Author(s):  
Behnaz Bayat ◽  
Annalena Traum ◽  
Heike Berghöfer ◽  
Silke Werth ◽  
Jieging Zhu ◽  
...  

Abstract Background Fetal/neonatal alloimmune thrombocytopenia (FNAIT) results from maternal alloantibodies (abs) reacting with fetal platelets expressing paternal human platelet antigens (HPAs), mostly HPA-1a. Anti-HPA-1a abs, are the most frequent cause of severe thrombocytopenia and intracranial hemorrhage (ICH). Objectives Titration of anti-HPA-1a in maternal serum using standard National Institute for Biological Standards and Control (NIBSC) 03/152 is one diagnostic approach to predict the severity of FNAIT. Recently, we found three anti-HPA-1a subtypes reacting with the β3 subunit independently or dependently from complexes with αIIb and αv. Endothelial cell-reactive anti-αvβ3 abs were found predominantly in cases with ICH. Our aim was to assess whether available standard material represents all anti-HPA-1a subtypes. Materials and Methods In this study, anti-HPA-1a sera (NIBSC 03/152) and human monoclonal antibodies (moabs) against HPA-1a (moabs 26.4 and 813) were evaluated using transfected cell lines expressing αIIbβ3, αvβ3 or monomeric cβ3. Results Flow cytometry analyses with well-characterized murine moabs recognizing αIIbβ3, αvβ3, or β3 alone demonstrated that AP3 reacts compound-independently, whereas compound-dependent moabs Gi5 and 23C6 reacted only with complexes. NIBSC 03/152, moabs 26.4, and 813 against HPA-1a reacted like AP3, same results were obtained with monomeric cβ3 in immunoblotting. Antigen capture assay targeting endothelial cells showed anti-HPA-1a reactivity disappearance after cβ3 beads adsorption. Furthermore, in contrast to anti-HPA-1a abs from ICH cases, none of NIBSC 03/152, 26.4, and 813 inhibited tube formation. Conclusion These results suggest that current anti-HPA-1a standard material contains only the anti-β3 subtype. The absence of anti-αvβ3 makes NIBSC 03/152 less suitable as standard to predict the severity of FNAIT.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. SCI-50-SCI-50
Author(s):  
Heyu Ni

Abstract Fetal and neonatal alloimmune thrombocytopenia (FNAIT) is a severe alloimmune disorder that results from fetal/neonatal platelet opsonization by maternal antibodies, which cross the placenta and result in fetal/neonatal platelet destruction. The frequency of FNAIT has been estimated at 0.5-1.5/1,000 liveborn neonates. However, this number does not include fetuses that die from this disease, since the incidence of FNAIT miscarriage has not been adequately studied. Analogous to autoimmune thrombocytopenia (ITP), the major target antigens in FNAIT are the platelet GPIIIa (β3 integrin) and GPIbα. However, severe bleeding is much more frequent in FNAIT, particularly the occurrence of intracranial hemorrhage (ICH). It is not known why the reported incidence of FNAIT mediated by anti-GPIbα antibodies is at a much lower frequency when compared to ITP, and whether the severe bleeding tendency in FNAIT is due to β3 integrin expression on angiogenic vessels in the developing fetus, which are targeted by cross-reacting maternal anti-β3 integrin antibodies. To study the pathogenesis and to develop new strategies for prevention and treatment, we have established murine models of FNAIT using β3 integrin and GPIbα deficient (-/-) mice. We first transfused these deficient mice with wild-type (WT) platelets to induce anti-β3 or anti-GPIbα antibody responses; we then bred these immunized female mice with WT males, causing FNAIT in the offspring. We found that maternal antiplatelet antibody titer correlated with the severity of FNAIT. These two murine models have revealed fundamental differences between the pathogenesis of anti-β3 and anti-GPIbα-mediated FNAIT. In anti-β3-mediated FNAIT, we found severe thrombocytopenia, ICH, and miscarriage. We also found the impairment of angiogenesis, which may contribute to ICH and intrauterine growth retardation. In contrast, the anti-GPIbα-mediated model revealed a nonclassical form of FNAIT (e.g., miscarriage but not bleeding disorders in neonates). We found that anti-GPIbα antibodies caused thrombosis in the placentas and miscarriage in most pregnant mice, which may partially explain the rarity of anti-GPIbα-mediated FNAIT reported in humans. Despite these substantial differences, there are also similarities between anti-β3 and anti-GPIbα-mediated FNAIT, such as the role of the neonatal Fc receptor (FcRn). FcRn is important for serum IgG homeostasis and for IgG transportation across the placenta. Using FcRn-/- mice, we demonstrated that fetal (but not maternal) FcRn is required to transport maternal antibodies to the fetal circulation and is indispensable for FNAIT. Blocking FcRn with an anti-FcRn antibody markedly reduced the severity of both anti-β3 and anti-GPIbα-mediated FNAIT. This important finding may lead to the development of new therapies (e.g., anti-FcRn antibody) against FNAIT and other maternal pathogenic antibody-mediated fetal/neonatal diseases. These models of FNAIT have allowed us to investigate the efficacy and mechanism of action of several therapies, including the aforementioned anti-FcRn antibody as well as antibody-mediated immune suppression (AMIS) and intravenous IgG (IVIG). In β3-/- mice, prophylactic administration of anti-HPA-1a antibody or murine β3 antisera induced AMIS against human HPA-1a-positive or murine WT platelets, respectively. Importantly, AMIS induced by β3 antisera suppressed the antibody response, thrombocytopenia, and miscarriage in FNAIT mice. These findings support the hypothesis that anti-HPA-1a antibody administration to HPA-1a-negative women after delivery of an HPA-1a-positive child may prevent FNAIT in subsequent pregnancies. The efficacy of IVIG is inconsistent, sources are limited, and the mechanism of action is not fully understood. We demonstrated that IVIG ameliorated both anti-β3 and anti-GPIbα-mediated FNAIT. IVIG decreased antiplatelet antibodies in both maternal and neonatal circulations, fetal platelet clearance, bleeding, and fetal mortality. In summary, we have uncovered fundamental differences in the pathogenesis of anti-β3 and anti-GPIbα-mediated FNAIT and have greatly enhanced our understanding of emerging and existing therapies. We will continue to investigate the pathogenesis of FNAIT so that we may develop more tailored and accessible therapeutic strategies for patients. Disclosures: No relevant conflicts of interest to declare.


Author(s):  
Noor Aqilah Binti Ashamuddin ◽  
Sabariah Binti Mohd Noor ◽  
Irni Binti Mohd Yasin

Neonatal alloimmune thrombocytopenia (NAIT) is the leading cause of thrombocytopenia in otherwise healthy new-born. (1,2) Maternal antibodies raised against paternally inherited alloantigen carried on fetal platelet causing NAIT. Maternal IgG antibodies passed through to the fetal via the placenta, attack and cause the destruction of the fetal platelet. (3) We present a case of NAIT without any complications in a premature baby (35 weeks) with VACREL association, G6PD deficiency, left calcified cephalohaematoma, cardiomegaly and hypospadias with severe thrombocytopenia (platelet counts is 23 109/L) at day two of life and received twice platelet transfusion. Platelet count initially 123 109/L at birth but significantly drop and persistently less than 50 109/L until day 10 of life before it normalized. Maternal serum antibody screening was negative, but platelet immunology test detected maternal platelet-reactive antibody Anti-HLA Class I and correlates with incompatible parental crossmatch indicating that parent had “platelet-antigen incompatibility”. The goal of obstetric management is to identify pregnancies at risk and prevent intracranial haemorrhage. (4) There is no evidence to support routine screening for pregnancies as per current practice. (2, 5) The latest treatments include maternal administration of intravenous immunoglobulin to suppress maternal antibody production and or to reduce placental transfer of antibodies; with or without steroids during antepartum period besides planning of mode, timing and method of delivery. (2, 5, 6, 7) This is a rare and unique case of NAIT secondary to Anti-HLA Class I antibody and hence clinician should be au fait with the diagnosis and management as it is infrequent among Malaysian.International Journal of Human and Health Sciences Supplementary Issue-2: 2021 Page: S21


Blood ◽  
2006 ◽  
Vol 107 (7) ◽  
pp. 2976-2983 ◽  
Author(s):  
Heyu Ni ◽  
Pingguo Chen ◽  
Christopher M. Spring ◽  
Ebrahim Sayeh ◽  
John W. Semple ◽  
...  

AbstractFetal and neonatal alloimmune thrombo cytopenia (FNAITP) is a life-threatening bleeding disorder caused by maternal antibodies directed against fetal platelet antigens. The immunoreactive epitopes in FNAITP are primarily located in the extracellular regions of the platelet glycoprotein IIIa (β3 integrin). Here we have established a novel animal model of FNAITP using β3 integrin–deficient (β3-/-) mice. We demonstrated first that these mice are immunoresponsive to β3 integrin; β3-/- mice transfused with wild-type platelets generated specific anti–β3 antibodies which were able to induce thrombocytopenia in wild-type mice. Subsequently, β3-/- female mice (both naive and immunized) were bred with wild-type male mice to recapitulate the features of FNAITP. The titer of generated maternal antibodies correlated with the severity of FNAITP. High titer maternal anti–β3 anti-bodies caused severe fetal thrombocytopenia, intracranial hemorrhage, and even miscarriage. Furthermore, maternal administration of intravenous immunoglobulin G (IgG) ameliorated FNAITP and down-regulated pathogenic antibodies in both the maternal and fetal circulations.


2014 ◽  
Vol 12 (1) ◽  
pp. 96-99 ◽  
Author(s):  
Fabiana Mendes Conti ◽  
Sergio Hibner ◽  
Thiago Henrique Costa ◽  
Marcia Regina Dezan ◽  
Maria Giselda Aravechia ◽  
...  

Neonatal alloimmune thrombocytopenia is a serious disease, in which the mother produces antibodies against fetal platelet antigens inherited from the father; it is still an underdiagnosed disease. This disease is considered the platelet counterpart of the RhD hemolytic disease of the fetus and newborn, yet in neonatal alloimmune thrombocytopenia the first child is affected with fetal and/or neonatal thrombocytopenia. There is a significant risk of intracranial hemorrhage and severe neurological impairment, with a tendency for earlier and more severe thrombocytopenia in subsequent pregnancies. This article reports a case of neonatal alloimmune thrombocytopenia in the second pregnancy affected and discusses diagnosis, management and the clinical importance of this disease.


2020 ◽  
Author(s):  
Xiaolin Wang ◽  
Yongqian Bian ◽  
Yuejun Li ◽  
Jing Li ◽  
Congying Zhao ◽  
...  

Abstract Background: DARC (The Duffy antigen receptor for chemokines) is a kind of glycosylated membrane protein that binds to members of the CXC chemokine family associated with angiogenesis and has recently been reported to be implicated in diverse normal physiologic processes. This study aimed to investigate the involvement of DARC in angiogenesis, which is known to generate new capillary blood vessels from preexisting ones. Methods: HDMECs (Human dermal microvascular endothelial cells) were divided into two groups (DARC overexpression group, and control group). We used Brdu staining to detect cell proliferation, and wound healing assay to detect cell migration. Then tube formation assay were observed. Also, western blot and immunofluorescent staining were used to estimate the relationship between DARC and RhoA (Ras homolog gene family, member A). Results: HDMECs proliferation, migration, and tube formation were inhibited significantly when DARC was overexpressed intracellular. DARC impaired microfilament dynamics and intercellular connection in migrating cells, and RhoA activation underlay the effect of DARC on endothelial cell. Furthermore, DARC inhibited the formation of new capillaries in vitro. Conclusion: Our findings revealed the role of DARC in the angiogenic process and provided a novel mechanism for RhoA activation during endothelial cell migration and angiogenesis.


Blood ◽  
2003 ◽  
Vol 101 (3) ◽  
pp. 937-942 ◽  
Author(s):  
Julie A. Peterson ◽  
Contessa E. Nyree ◽  
Peter J. Newman ◽  
Richard H. Aster

Abstract Drug-dependent antibodies (DDAbs) can cause the precipitous destruction of platelets if a patient is exposed to the drug for which the antibodies are specific. The molecular character of the epitopes recognized is poorly understood, and the mechanism by which drugs promote tight binding of these antibodies to platelet glycoproteins without linking covalently to protein or antibody is not yet known. We studied a group of quinine-dependent antibodies that react with human glycoprotein IIIa (GPIIIa; β3-integrin subunit) but fail to recognize rat GPIIIa, despite close homology between the 2 proteins. By characterizing reactions of these antibodies with human/rat GPIIIa chimeras and selected GPIIIa mutants, we found that each of 3 quinine-dependent antibodies requires a 17-amino acid sequence in the newly recognized “hybrid” and PSI homology domains of GPIIIa for drug-dependent binding. Disulfide bonds are required to stabilize the target epitope. Monoclonal antibody AP3, which blocks the binding of these DDAbs to GPIIIa, was found to require a more limited stretch of the same peptide for its reaction with the glycoprotein. The findings suggest this region of GPIIIa may be a favored target for quinine-dependent antibodies and may provide a basis for further studies to elucidate the molecular basis of glycoprotein–drug–antibody interaction.


2012 ◽  
Vol 2012 ◽  
pp. 1-5 ◽  
Author(s):  
Maria Therese Ahlen ◽  
Anne Husebekk ◽  
Mette Kjær Killie ◽  
Jens Kjeldsen-Kragh ◽  
Martin L. Olsson ◽  
...  

Background. Maternal alloantibodies against HPA-1a can cross placenta, opsonize foetal platelets, and induce neonatal alloimmune thrombocytopenia (NAIT). In a study of 100, 448 pregnant women in Norway during 1995–2004, 10.6% of HPA-1a negative women had detectable anti-HPA-1a antibodies.Design and Methods. A possible correlation between the maternal ABO blood group phenotype, or underlying genotype, and severe thrombocytopenia in the newborn was investigated.Results. We observed that immunized women with blood group O had a lower risk of having a child with severe NAIT than women with group A; 20% with blood group O gave birth to children with severe NAIT, compared to 47% among the blood group A mothers (relative risk 0.43; 95% CI 0.25–0.75).Conclusion. The risk of severe neonatal alloimmune thrombocytopenia due to anti-HPA-1a antibodies is correlated to maternalABOtypes, and this study indicates that the observation is due to genetic properties on the maternal side.


Gene ◽  
2008 ◽  
Vol 408 (1-2) ◽  
pp. 9-17 ◽  
Author(s):  
Angeles Jiménez-Marín ◽  
Noemí Yubero ◽  
Gloria Esteso ◽  
Angela Moreno ◽  
Juana Martín de las Mulas ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1232-1232
Author(s):  
Jens Kjeldsen-Kragh ◽  
Mette K. Killie ◽  
Geir Tomter ◽  
Elzbieta Golebiowska ◽  
Helene Pedersen ◽  
...  

Abstract Background: Neonatal alloimmune thrombocytopenia (NAIT) is most frequently caused by antibodies against the human platelet antigen (HPA) 1a. The objective of the present study was to identify HPA 1a negative women, and to offer them an intervention program aimed to reduce morbidity and mortality of NAIT. Methods: A total of 100,448 pregnant women were HPA 1 typed. The HPA 1a negative women were screened for anti-HPA 1a, which was quantified when present. Immunized women were referred to a university hospital for clinical follow-up, including ultrasonographic examination of the fetal brain. Caesarean section was performed 2–4 weeks prior to term with platelets from HPA 1bb donors reserved for immediate transfusion if petechiae were present and/or if platelet count was < 35 × 109/L. Results: Of all women typed 2.1% were HPA 1a negative. Anti-HPA 1a was detected in 210 of 1,990 HPA 1a negative women. A total of 170 pregnancies in 154 HPA 1a negative women were managed according to the intervention program. These women gave birth to 161 HPA 1a positive children of whom 55 had severe thrombocytopenia (<50 × 109/L) including two with ICH. There were no intrauterine deaths. In 13 previously published prospective studies comprising 131,465 women of whom 2,290 were HPA 1a negative, there were 10 cases with severe NAIT-related complications (3 intrauterine deaths and 7 neonates with ICH), which are significantly higher than in our study (p < 0.05). Conclusions: The screening and intervention program seems to reduce mortality and serious morbidity associated with NAIT.


Sign in / Sign up

Export Citation Format

Share Document