Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Roberta Attanasio is active.

Publication


Featured researches published by Roberta Attanasio.


Journal of Immunology | 2006

IgG Fc Receptor III Homologues in Nonhuman Primate Species: Genetic Characterization and Ligand Interactions

Kenneth A. Rogers; Franco Scinicariello; Roberta Attanasio

Ig Fc receptors bind to immune complexes through interactions with the Fc regions of specific Ab subclasses to initiate or inhibit the defense mechanisms of the leukocytes on which they are expressed. The mechanism of action of IgG-based therapeutic molecules, which are routinely evaluated in nonhuman primate models, involves binding to the low-affinity FcRIII (CD16). The premise that IgG/CD16 interactions in nonhuman primates mimic those present in humans has not been evaluated. Therefore, we have identified and characterized CD16 and associated TCR ζ-chain homologues in rhesus macaques, cynomolgus macaques, baboons, and sooty mangabeys. Similar to humans, CD16 expression was detected on a lymphocyte subpopulation, on monocytes, and on neutrophils of sooty mangabeys. However, CD16 was detected only on a lymphocyte subpopulation and on monocytes in macaques and baboons. A nonhuman primate rCD16 generated in HeLa cells interacted with human IgG1 and IgG2. By contrast, human CD16 binds to IgG1 and IgG3. As shown for humans, the mAb 3G8 was able to block IgG binding to nonhuman primate CD16 and inhibition of nonhuman primate CD16 N-glycosylation enhanced IgG binding. Clearly, differences in interaction with IgG subclasses and in cell-type expression should be considered when using these models for in vivo evaluation of therapeutic Abs.


Immunology | 2004

Rhesus macaque antibody molecules: sequences and heterogeneity of alpha and gamma constant regions

Franco Scinicariello; Carrie N. Engleman; Lakshmi Jayashankar; Harold M. McClure; Roberta Attanasio

Rhesus macaques (Macaca mulatta) are extensively used in vaccine development. Macaques infected with simian immunodeficiency viruses (SIV) or simian‐human immunodeficiency viruses (SHIV) are the best animal model currently available for acquired‐immune‐deficiency‐syndrome‐related studies. Recent results emphasize the importance of antibody responses in controlling HIV and SIV infection. Despite the increasing attention placed on humoral immunity in these models, very limited information is available on rhesus macaque antibody molecules. Therefore, we sequenced, cloned and characterized immunoglobulin gamma (IGHG) and alpha (IGHA) chain constant region genes from rhesus macaques of Indian and Chinese origin. Although it is currently thought that rhesus macaques express three IgG subclasses, we identified four IGHG genes, which were designated IGHG1, IGHG2, IGHG3 and IGHG4 on the basis of sequence similarities with the four human genes encoding the IgG1, IgG2, IgG3 and IgG4 subclasses. The four genes were expressed at least at the messenger RNA level, as demonstrated by real‐time reverse transcription polymerase chain reaction (RT‐PCR). The level of intraspecies heterogeneity was very high for IGHA genes, whereas IGHG genes were remarkably similar in all animals examined. However, single amino acid substitutions were present in IGHG2 and IGHG4 genes, indicating the presence of IgG polymorphism possibly resulting in the expression of different allotypes. Two IgA alleles were identified in several animals and RT‐PCR showed that both alleles may be expressed. Presence of immunoglobulin gene polymorphism appears to reflect the unusually high levels of intraspecies heterogeneity already demonstrated for major histocompatibility complex genes in this non‐human primate species.


Immunology | 2001

Intraspecies heterogeneity of immunoglobulin α-chain constant region genes in rhesus macaques

Franco Scinicariello; Roberta Attanasio

Immunoglobulin A (IgA) is the major antibody class present in external secretions and is also an important component of serum immunoglobulins. On mucosal surfaces, IgA represents a first line of defence by neutralizing invading pathogens. The number of IgA constant‐region genes (Cα) present in different mammalian species is variable. Immunoglobulin Cα genes differ mainly in the sequences located in the hinge region. IgA molecules, whose hinge regions are remarkably similar to those of the respective human molecules, are present in hominoid primates. In this report, we show that two alleles of a single immunoglobulin Cα are present in rhesus macaques (Macaca mulatta). In addition, we show that intraspecies immunoglobulin Cα allelic polymorphism is very high in this non‐human primate species. Specifically, five different hinge regions, some of which are proline‐rich, were identified from a total of eight rhesus macaque immunoglobulin Cα‐chains. The five hinge regions were different from those present in hominoid primates, both in length and in sequence. These results represent the first example of high levels of intraspecies immunoglobulin constant‐region variability and suggest that IgAs of variable structure and function may be present in rhesus macaques. As rhesus macaques are widely used as animal models for the development of vaccines for acquired immune deficiency syndrome (AIDS), the possible presence of structurally and functionally variable IgA molecules in different animals should be taken into account when designing experimental strategies to induce mucosal antibody responses to human immunodeficiency virus (HIV).


Clinical and Experimental Immunology | 2001

Age-related autoantibody production in a nonhuman primate model

Roberta Attanasio; Kathleen M. Brasky; S. H. Robbins; L. Jayashankar; R. J. Nash; T. M. Butler

Autoantibody production increases with ageing. However, the pathological significance of this increase as well as the corresponding underlying mechanisms are poorly understood. To further our understanding of the role that ageing plays in the development of autoantibody responses, we used a novel nonhuman primate model consisting of healthy baboons of ages representing the entire lifespan of this animal species. Results from this study indicate that production of antinuclear antibodies, anticell extract antibodies and natural autoantibodies gradually and significantly increases from young age to old age without a corresponding increase in neither serum immunoglobulin concentration nor in levels of selected markers of immune dysregulation (sTNF‐RI, sTNF‐RII, IL‐2 sRα and IFN‐γ). Therefore, in the baboon model, autoantibodies may be produced in absence of recognizable pathological conditions of the ageing immune system.


Immunogenetics | 2011

Characterization and allelic polymorphisms of rhesus macaque (Macaca mulatta) IgG Fc receptor genes

Doan C. Nguyen; Franco Scinicariello; Roberta Attanasio

Macaque models are invaluable for AIDS research. Indeed, initial development of HIV-1 vaccines relies heavily on simian immunodeficiency virus-infected rhesus macaques. Neutralizing antibodies, a major component of anti-HIV protective responses, ultimately interact with Fc receptors on phagocytic and natural killer cells to eliminate the pathogen. Despite the major role that Fc receptors play in protective responses, there is very limited information available on these molecules in rhesus macaques. Therefore, in this study, rhesus macaque CD32 (FcγRII) and CD64 (FcγRI) homologues were genetically characterized. In addition, presence of CD16 (FcγRIII), CD32, and CD64 allelic polymorphisms were determined in a group of nine animals. Results from this study show that the predicted structures of macaque CD32 and CD64 are highly similar to their human counterparts. Macaque and human CD32 and CD64 extracellular domains are 88–90% and 94–95% homologous, respectively. Although all cysteines are conserved between the two species, macaque CD32 exhibits two additional N-linked glycosylation sites, whereas CD64 lacks three of them when compared to humans. Five CD32, three CD64, and three CD16 distinct allelic sequences were indentified in the nine animals examined, indicating a relatively high level of polymorphism in macaque Fcγ receptors. Together, these results validate rhesus macaques as models for vaccine development and antibody responses, while at the same time, underscoring the need to take into account the high degree of genetic heterogeneity present in this species when designing experimental protocols.


Immunology | 2004

Identification and characterization of macaque CD89 (immunoglobulin A Fc receptor)

Kenneth A. Rogers; Franco Scinicariello; Roberta Attanasio

The interaction of the immunoglobulin A (IgA) molecule with its specific cellular receptor is necessary to trigger a variety of effector functions able to clear IgA‐opsonized antigens. The human IgA‐specific Fc receptor, FcαRI or CD89, is expressed on cells of the myeloid lineage. Recently, CD89 homologues have been identified in rats and cattle. Because non‐human primates represent well established models for a variety of human diseases and for the testing of immunotherapeutic strategies, we cloned and sequenced cDNAs corresponding to the CD89 gene from rhesus (Macaca mulatta) and cynomolgus (Macaca fascicularis) macaques. Macaque sequences of full‐length CD89 consist of five exons of length identical to the corresponding human CD89 exons. The rhesus and cynomolgus macaque derived amino acid sequences are highly homologous to each other (99·3% identity) and exhibit 86·5% and 86·1% identity to the human counterpart, respectively. Transfection of HeLa cells with plasmids containing the cloned macaque cDNAs resulted in the expression of surface molecules recognized by an anti‐human CD89 antibody. Five splice variants were identified in rhesus macaques. Three of the five variants are similar to described human CD89 splice variants, whereas two variants have not been described in humans. Three splice variants were identified in cynomolgus macaques. Of the three variants, one is present also in humans and rhesus macaques, whereas the other two are shared with rhesus macaques but not humans. Similarly to the human CD89, macaque CD89 is expressed on myeloid cells from peripheral blood. The characterization of macaque CD89 represents an essential step in establishing a non‐human primate model for the testing of immunotherapeutic approaches based on the manipulation of the IgA/CD89 interaction.


Vaccine | 2011

17β-Estradiol restores antibody responses to an influenza vaccine in a postmenopausal mouse model.

Doan C. Nguyen; Feda Masseoud; Xiuhua Lu; Franco Scinicariello; Suryaprakash Sambhara; Roberta Attanasio

Post-menopausal women belong to an age group that is highly susceptible to influenza infection and its most serious complications. However, data on the immunogenicity of influenza vaccines in these women is limited. Therefore, the antibody response to influenza vaccination was assessed in a postmenopausal mouse model. An inactivated-detergent-split vaccine from the A/New Caledonia/20/99 (H1N1) influenza virus strain was given to three groups of mice: ovariectomized (OVEX), OVEX with 17β-estradiol replacement (OVEX+E2), and sham-OVEX. The OVEX+E2 group produced influenza virus-specific serum antibodies, including neutralizing antibodies, at significantly higher levels (p<0.001) than did OVEX mice. These levels matched those observed in the sham-OVEX group, indicating that ovariectomy negatively modulates the antibody response to the influenza vaccine, whereas 17β-estradiol replacement restores this response to levels observed in intact animals. Our findings suggest that immunogenicity and efficacy of influenza vaccines need to be evaluated in postmenopausal women, including women receiving hormone replacement therapy.


Immunology | 2006

Molecular characterization of immunoglobulin D in mammals: immunoglobulin heavy constant delta genes in dogs, chimpanzees and four old world monkey species

Kenneth A. Rogers; Jennifer P. Richardson; Franco Scinicariello; Roberta Attanasio

Antibodies are adaptor molecules that neutralize pathogens and link humoral and cellular defence mechanisms. Immunoglobulin D (IgD), one of the five antibody classes present in mammals, is expressed as an antigen receptor on naïve B cells. The functional role that IgD plays in the immune response is still poorly understood, but the recent characterization of immunoglobulin heavy constant delta genes (IGHD) in a variety of species challenges the view that IgD is of minor importance and is not present in many animals. On the basis of serological studies, IgD appears to be expressed in the majority of mammalian species examined. To confirm, at the molecular level, that IgD is present in different species, we cloned and sequenced IGHD cDNA from dogs and five non‐human primate species (chimpanzee, rhesus macaque, cynomolgus macaque, baboon and sooty mangabey). Our results show that in all six species, IgD heavy chains possess three immunoglobulin domains and a long hinge region encoded by two exons. Only the hinge region of non‐human primates is similar to the human hinge region, with conservation of O‐glycosylation sites and multiple charged residues at opposing ends. The preservation of IgD in primates, dogs and previously characterized species suggests an important functional role for IgD, possibly involving binding to a receptor. The high degree of similarity existing between the structural features of human and non‐human primate IgD suggests that non‐human primates are suitable for in vivo studies designed to define the role that IgD plays in the immune response.


Journal of Clinical Immunology | 2002

Immunomodulatory Effects of Estrogen and Progesterone Replacement in a Nonhuman Primate Model

Roberta Attanasio; Deborah A. Gust; Mark E. Wilson; Tracy Meeker; Thomas P. Gordon

A novel postmenopausal nonhuman primate model consisting of healthy young and old ovariectomized rhesus macaques was used to assess the short-term immunomodulatory effects of transdermally administered estrogen and progesterone. Specifically, we determined estrogen- and progesterone-induced changes in absolute numbers of circulating lymphocytes (B lymphocytes, CD4+ lymphocytes, and CD8+ lymphocytes) as well as lymphocytes expressing the activation markers CD25 and CD69. In addition, we assessed B and T lymphocyte activity, i.e, immunoglobulin (Ig) and interferon-γ (IFN-γ) production by peripheral blood mononuclear cells (PBMCs). In general, treatment with estrogen or progesterone resulted in decreased lymphocyte numbers and in down-modulation of activation markers. In addition, hormone replacement resulted in a decreasing trend for PBMC IFN-γ production, whereas PBMC Ig production was minimally affected. Hormone treatment seemed to influence young and old animals differently, with the young animals appearing more susceptible to its immune system-related effects. These results indicate that, in our animal model exogenously administered hormones may dynamically interact with the immune system, resulting in in vivo modulation of lymphocyte numbers and activity.


Immunogenetics | 2002

Baboon immunoglobulin constant region heavy chains: identification of four IGHG genes

Roberta Attanasio; Lakshmi Jayashankar; Carrie N. Engleman; Franco Scinicariello

Abstract. The increasing use of nonhuman primate models in biomedical research and especially in vaccine development requires the characterization of their immunoglobulin genes and corresponding products. Therefore, we sequenced, cloned and characterized the four immunoglobulin gamma chain constant region genes (IGHG) present in baboons. These four genes were designated IGHG1, IGHG2, IGHG3 and IGHG4 on the basis of sequence similarities with the four human genes encoding the IgG1, IgG2, IgG3 and IgG4 subclasses and the three known rhesus macaque IGHG genes. Specifically, the baboon IgG1, IgG2, IgG3 and IgG4 sequences exhibit 90.3%, 88.3%, 86.7% and 89.6% amino acid identity to their human counterpart. The percent of amino acid identity of baboon IgG1, IgG2 and IgG3 to the corresponding rhesus macaque sequences is 98.5, 93.1 and 94.4, respectively. Therefore, baboon and rhesus macaque IGHG genes are highly homologous to each other. The majority of differences existing between baboon and human sequences are clustered in the hinge region, with the upper hinge being the most diverse and containing several proline residues. Similar to rhesus macaques, the hinge regions of all baboon IGHG genes consist of a single exon, whereas in humans the IgG3 molecule is encoded by multiple exons. These results confirm the evolutionary instability of the hinge region and indicate that functional properties associated with the hinge regions of baboon and human IgG molecules might differ between the two species.

Collaboration


Dive into the Roberta Attanasio's collaboration.

Top Co-Authors

Avatar

Franco Scinicariello

U.S. Agency for Toxic Substances and Disease Registry

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kathleen M. Brasky

Texas Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Feda Masseoud

Georgia State University

View shared research outputs
Top Co-Authors

Avatar

Benjamin C. Blount

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge