Network


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

Hotspot


Dive into the research topics where Elizabeth Mayhew is active.

Publication


Featured researches published by Elizabeth Mayhew.


Journal of Immunology | 2000

Human Uveal Melanoma Cells Produce Macrophage Migration-Inhibitory Factor to Prevent Lysis by NK Cells

Amanda C. Repp; Elizabeth Mayhew; Jerry Y. Niederkorn

Human uveal melanoma arises in an immune privileged ocular environment in which both adaptive and innate immune effector mechanisms are suppressed. Uveal melanoma is the most common intraocular tumor in adults and is derived from tissues in the eye that produce macrophage migration-inhibitory factor (MIF), a cytokine that has recently been demonstrated to produce immediate inhibition of NK cell-mediated lytic activity. Although NK cell-mediated lysis of uveal melanomas is inhibited in the eye, melanoma cells that disseminate from the eye are at risk for surveillance by NK cells. Moreover, uveal melanoma cells demonstrate a propensity to metastasize to the liver, an organ with one of the highest levels of NK activity in the body. Therefore, we speculated that uveal melanomas produced MIF as a means of escaping NK cell-mediated lysis. Accordingly, seven primary uveal melanoma cell lines and two cell lines derived from uveal melanoma metastases were examined for their production of MIF. MIF was detected in melanoma culture supernatants by both ELISA and the classical bioassay of macrophage migration inhibition. Melanoma-derived MIF inhibited NK cell-mediated lysis of YAC-1 and uveal melanoma cells. Cell lines derived from uveal melanoma metastases produced approximately twice as much biologically active MIF as cultures from primary uveal melanomas. Inhibition of NK cell-mediated killing by uveal melanoma-derived MIF was specifically inhibited in a dose-dependent manner by anti-MIF Ab. The results suggest that human uveal melanoma cells maintain a microenvironment of immune privilege by secreting active MIF that protects against NK cell-mediated killing.


Transplantation | 2005

Cd4+ T-cell–mediated Mechanisms of Corneal Allograft Rejection: Role of Fas-induced Apoptosis

Sushma Hegde; Clay Beauregard; Elizabeth Mayhew; Jerry Y. Niederkorn

Background. The role of CD4+ T cells as effector cells in corneal allograft rejection is poorly understood. We investigated the role of CD4+ T cells as helper cells in the generation of allospecific effector macrophages in corneal graft rejection and the role of CD4+ T cells as apoptosis-inducing effector cells. Methods. Corneal allografts were transplanted to CD4 knockout, FasL-deficient, and macrophage-depleted hosts. An Annexin-V binding assay was used to evaluate the susceptibility of corneal cells to both Fas-dependent and CD4+ T-cell–mediated apoptosis in vitro. Results. Macrophages were essential for graft rejection, but not as effector cells. Anti-BALB/c CD4+ T cells from immunized C57BL/6 mice induced apoptosis of BALB/c corneal epithelial and endothelial cells. However, anti-BALB/c CD4+ T cells from FasL-deficient gld/gld mice did not induce apoptosis of BALB/c corneal endothelial cells. Moreover, gld/gld mice had a reduced capacity to reject BALB/c corneal allografts. Although the initial results suggested a role for Fas-induced apoptosis in corneal graft rejection, additional experiments indicated otherwise. The incidence and tempo of immune rejection of Fas-deficient lpr/lpr corneal allografts were no different than those for corneal grafts from Fas-bearing C57BL/6 donors. Moreover, CD4+ T-cell–mediated apoptosis of corneal cells could not be blocked with either Fas-Fc fusion protein or anti-FasL blocking antibody. Conclusions. The results suggest that CD4+ T cells function directly as effector cells and not as helper cells in the rejection of corneal allografts. Although the corneal endothelium is highly susceptible to Fas-induced apoptosis, this is apparently not the primary mechanism of CD4+ T-cell–dependent rejection.


Journal of Immunology | 2001

Ocular Immune Privilege Promoted by the Presentation of Peptide on Tolerogenic B Cells in the Spleen. II. Evidence for Presentation by Qa-1

Thomas J. D'Orazio; Elizabeth Mayhew; Jerry Y. Niederkorn

Ocular immune privilege is the result of several unique features of the eye, including the systemic down-regulation of Th1 immune responses to Ags encountered in the anterior chamber of the eye—a phenomenon termed anterior chamber-associated immune deviation (ACAID). The induction of ACAID requires the participation of three cell populations: the ocular ACAID APC, the splenic B cell, and the splenic T cell. Because B cells have been implicated in tolerogenic Ag presentation in other systems, we hypothesized that B cells were responsible for the induction of regulatory T cells in ACAID. The central hypothesis for this study is that APC from the eye migrate to the spleen where they release antigenic peptides (OVA) that are captured and presented to T cells by splenic B cells. A combination of in vitro and in vivo studies demonstrated that splenic B cells, incubated with ACAID APC in vitro, were capable of inducing ACAID when transferred to naive mice. The induction of ACAID required the normal expression of β2-microglobulin on both the B cell and ACAID APC, but not on the T suppressor cells. Moreover, the induction of ACAID regulatory cells required histocompatibility between the B cells and regulatory T cells at the TL/Qa region. The results indicate that: 1) B cells are necessary for the induction of ACAID; 2) ACAID B cells do not directly suppress the expression of delayed-type hypersensitivity; and 3) the induction of Ag-specific regulatory T cells by ACAID B cells requires histocompatibility at the TL/Qa region.


Immunology | 2003

CD25+, interleukin‐10‐producing CD4+ T cells are required for suppressor cell production and immune privilege in the anterior chamber of the eye

Molly E. Skelsey; Elizabeth Mayhew; Jerry Y. Niederkorn

An important factor in the establishment of ocular immune privilege is the dynamic down regulation of T helper 1 (Th1) immune responses that occurs in response to antigens delivered intraocularly; a phenomenon that has been termed anterior chamber‐associated immune deviation (ACAID). ACAID is characterized by the generation of splenic regulatory cells that inhibit the expression of delayed‐type hypersensitivity. Previous studies have shown that antigens introduced into the anterior chamber of the eye induce the generation of a CD4+ T‐cell population that suppress the induction of Th1 immune responses and the appearance of a second population of CD8+ T regulatory cells that suppresses the expression of Th1 inflammatory responses (= efferent suppressor cells). Experiments described here characterized the function of the CD4+ ACAID suppressor cell population and its effect on the generation of CD8+ efferent suppressor cells that inhibit the expression of DTH in situ. Both in vivo and in vitro experiments demonstrated that CD4+ T cells are required for the generation of CD8+ efferent suppressor cells. CD4+ T cells do not require cell contact with CD8+ T cells; instead they produce soluble IL‐10 that is sufficient for the generation of ACAID suppressor cells. Finally, the CD4+ afferent T suppressor cells are not natural killer T cells, but do express the CD25 cell surface marker.


Journal of Immunology | 2005

Cutting Edge: Atopy Promotes Th2 Responses to Alloantigens and Increases the Incidence and Tempo of Corneal Allograft Rejection

Clay Beauregard; Christina Stevens; Elizabeth Mayhew; Jerry Y. Niederkorn

A large body of evidence suggests that corneal allograft rejection is mediated by a type 1 Th cell response and that deviation toward type 2 immunity favors graft survival. However, clinical observations indicate that patients with severe ocular allergies have increased risk of corneal allograft rejection. We used a mouse model of atopic conjunctivitis to evaluate the effects of Th2 immune deviation on corneal allograft survival and possible mechanisms of graft rejection. Our results reveal the following novel findings: 1) atopic conjunctivitis promotes systemic Th2 immune responses to corneal graft donor alloantigens; 2) corneal allografts in atopic host eyes have an increased incidence and swifter tempo of rejection; 3) increased rejection is associated with alterations in systemic T cell-mediated responses to donor alloantigens; and 4) corneal allograft rejection in atopic hosts does not require the direct involvement of infiltrating eosinophils.


Transplantation | 2006

CD4+ T-cell-independent rejection of corneal allografts.

Jerry Y. Niederkorn; Christina Stevens; Jessamee Mellon; Elizabeth Mayhew

Background. Several studies suggest that a significant number of corneal allografts undergo rejection in the absence of CD4+ T cells. This study examined the role of CD4+ T cell-independent mechanisms of corneal allograft rejection. Methods. BALB/c corneal allografts were transplanted to C57BL/6 beige nude mice that received either CD8− or CD8+ T cells from C57BL/6 CD4 knockout (KO) mice that had rejected BALB/c corneal allografts. Immune effector functions of CD8− or CD8+ T cells from C57BL/6 CD4 KO mice were assessed using delayed-type hypersensitivity assays and Annexin V apoptosis assays respectively. Results. Both CD8− and CD8+ T cells from CD4 KO corneal allograft rejector mice mediated corneal allograft rejection following adoptive transfer to nude mice. CD8− T cells, but not CD8+ T cells, from CD4 KO mice adoptively transferred donor-specific DTH and induced apoptosis of BALB/c corneal endothelial cells in vitro. Apoptosis of BALB/c corneal endothelial cells was mediated by double negative (DN) T cells, as treatment of CD8− cells from CD4 KO mice with anti-Thy 1.2 plus complement abolished their effector function. Conclusion. The results support the proposition that CD4+ T cell-independent rejection of corneal allografts can be mediated by either CD8+ or CD8− T cells. The CD8− T cells represent a unique DN T cell population that might mediate rejection by either direct cytolysis or by inducing apoptosis of the donor corneal endothelium.


Transplant Immunology | 2003

Are corneal cells susceptible to antibody-mediated killing in corneal allograft rejection?

Sylvia L. Hargrave; Elizabeth Mayhew; Sushma Hegde; Jerry Y. Niederkorn

PURPOSE The precise role of antibodies in corneal transplantation is controversial. Clinical and experimental evidence both supports and refutes the contribution of donor-derived alloantibody in corneal allograft rejection. Accordingly, we prospectively evaluated the presence of donor-derived alloantibody in two high-risk donor-host combinations. We also evaluated the ability of this alloantibody to kill corneal epithelial, keratocytes, and endothelial cells in complement-dependent and complement-independent fashions. METHODS C3H/Hej (H-2(k)) and Balb/c (H-2(d)) corneal grafts were transplanted orthotopically to CB6F1 (H-2(b/d)) and C57BL/6 (H-2(b)) recipients, respectively. These two donor-host combinations represent disparity at the entire MHC and multiple minor histocompatibility loci. Four objectives were addressed. First, we wished to determine if there was a correlation between the appearance of donor-specific serum IgG antibody and corneal graft rejection. Second, we evaluated the effect of passive transfer of hyperimmune donor-specific antibody on corneal allograft rejection. Third, we examined the capacity of donor-specific alloantibody to mediate complement-dependent cytolysis of corneal cells. Finally, we determined the capability of donor-specific alloantibody to mediate apoptosis of corneal cells. RESULTS The presence of donor-specific serum IgG alloantibodies did not correlate with corneal graft rejection. One hundred percent of CB6F1 and C57BL/6 hosts rejected their C3H and Balb/c orthotopic corneal allografts, respectively. However, two of these seven CB6F1 hosts and one C57BL/6 host did not produce donor-specific IgG alloantibody that was significantly different from naive donors. Passive transfer of hyperimmune allo-antiserum prior to corneal transplantation did not increase the incidence, severity, or tempo of corneal allograft rejection in either donor-host combination. Hyperimmune allo-antiserum produced complement-mediated lysis of C3H corneal endothelial but not C3H corneal epithelial cells in the C3H-CB6F1 donor-host combination. Interestingly, all three corneal cell layers were vulnerable to complement-mediated cytolysis in the Balb/c-C57BL/6 donor-host combination. Additionally, Balb/c corneal epithelial, keratocytes, and endothelial cells were vulnerable to complement-independent, antibody induced apoptosis. CONCLUSIONS Corneal graft rejection does not appear to correlate with the production of IgG alloantibody and can occur in the absence of donor-specific IgG alloantibody. Antibody-mediated killing of the corneal endothelium can occur in a complement-dependent or complement-independent fashion.


Transplantation | 2002

Phenotypic analysis of oral tolerance to alloantigens: evidence that the indirect pathway of antigen presentation is involved.

Jerry Y. Niederkorn; Elizabeth Mayhew

Background. Oral administration of alloantigens induces down-regulation of Th1 immune responses and reduces the incidence of corneal graft rejection. This study examined the role of Th1 and Th2 cytokines, accessory cells, and lymphoid organs that are known to be instrumental in other forms of antigen-specific tolerance. Methods. Allogeneic dendritic cells (DC) were administered orally using a protocol that is known to reduce the incidence of corneal allograft rejection and prevent the generation of allospecific delayed-type hypersensitivity (DTH). Hosts included normal mice and gene knockout (KO) mice, including B cell-deficient &mgr;MT, interleukin (IL)-4 KO, IL-10 KO, and interferon (IFN)-&ggr; KO mice. The requirement for either an intact spleen or thymus was also examined. Orally administered paraformaldehyde-fixed, UVB-treated, or sonicated allogeneic cells were tested to determine if dead cells were capable of inducing tolerance. Results. Studies on gene KO mice indicated that a Th1 cytokine (IFN-&ggr;) and a Th2 cytokine (IL-4) were needed for the development of oral tolerance to alloantigens. By contrast, IL-10 was not required. Although an intact spleen was necessary for the development of tolerance, removal of the thymus did not affect down-regulation of DTH. Conclusions. Oral tolerance induced with allogeneic cells shares characteristics with antigen-specific unresponsiveness induced by other routes, yet there are some noteworthy differences. The capacity of killed or sonicated allogeneic cells to induce oral tolerance and enhance corneal graft survival indicates that oral tolerance to alloantigens can occur via the indirect pathway of alloantigen presentation. These results also emphasize the remarkable redundancy in the mechanisms that the immune system employs to produce antigen-specific unresponsiveness.


Experimental Eye Research | 2002

Adaptive immune responses to Acanthamoeba cysts

Kathy McClellan; Kevin Howard; Elizabeth Mayhew; Jerry Y. Niederkorn; Hassan Alizadeh

Acanthamoeba cysts are not eliminated from the corneas of human subjects or experimentally infected animals. The persistence of Acanthamoeba cysts in the cornea indicates that either the cysts escape immunological elimination or are not recognized by the hosts immunological elements. The aim of this study was to determine the immunogenicity and antigenicity of the Acanthamoeba cyst. Mice were immunized intraperitoneally and serum anti-Acanthamoeba IgG was measured by ELISA. Lymphoproliferative assay and delayed type hypersensitivity (DTH) responses to Acanthamoeba castellanii cyst and trophozoite antigens were used to determine the cell mediated immune responses against Acanthamoeba cysts. A. castellanii cysts were both immunogenic and antigenic, producing anti-Acanthamoeba serum IgG, T lymphocyte proliferation, and delayed type hypersensitivity responses. These results indicate that Acanthamoeba cysts are recognized by the immune system. The persistence of the organism in the human cornea means that these adaptive immune responses fail to kill Acanthamoeba cysts.


Current Eye Research | 1993

Effect of anti-ganglioside antibodies on the metastatic spread of intraocular melanomas in a nude mouse model of human uveal melanoma

Jerry Y. Niederkorn; Jessamee Mellon; M. Pidherney; Elizabeth Mayhew; Rajiv Anand

In vivo and in vitro studies were performed to determine: (a) if human uveal melanoma cells expressed GD2 and GD3 gangliosides; (b) if anti-GD2 monoclonal antibodies would inhibit the propensity of human uveal melanoma cells to localize in the liver following intravenous injection; and (c) if anti-GD2 monoclonal antibody would reduce the spontaneous metastasis of primary intraocular melanomas in nude mice. The results showed that all three of the human uveal melanoma cell lines tested expressed GD2 and GD3 gangliosides in vitro and in vivo. The human uveal melanoma cell lines preferentially localized in the liver and entered the hepatic parenchyma following spontaneous metastasis from the eyes of nude mice. In vivo administration of anti-GD2 monoclonal antibody produced a sharp reduction in the number of uveal melanoma cells that disseminated to the liver following either intravenous injection or by spontaneous metastasis from primary intraocular melanomas. Collectively, the results demonstrate that uveal melanoma cells display a propensity to localize in the liver after entering the bloodstream; however, this localization can be significantly inhibited by in vivo administration of anti-ganglioside antibodies. The expression of GD2 and GD3 surface gangliosides on uveal melanomas and the capacity of anti-ganglioside antibodies to inhibit metastasis formation in mouse models of ocular and cutaneous melanomas raise the possibility of implementing anti-ganglioside antibodies as potential therapeutic agents for the management of uveal melanoma.

Collaboration


Dive into the Elizabeth Mayhew's collaboration.

Top Co-Authors

Avatar

Jerry Y. Niederkorn

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jessamee Mellon

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peter W. Chen

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Christina Stevens

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hassan Alizadeh

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sushma Hegde

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Clay Beauregard

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kevin Howard

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rajendra S. Apte

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yu Guang He

University of Texas Southwestern Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge