Network


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

Hotspot


Dive into the research topics where Amar Nijagal is active.

Publication


Featured researches published by Amar Nijagal.


Journal of Clinical Investigation | 2011

Maternal T cells limit engraftment after in utero hematopoietic cell transplantation in mice

Amar Nijagal; Marta Wegorzewska; Erin Jarvis; Tom Le; Qizhi Tang; Tippi C. MacKenzie

Transplantation of allogeneic stem cells into the early gestational fetus, a treatment termed in utero hematopoietic cell transplantation (IUHCTx), could potentially overcome the limitations of bone marrow transplants, including graft rejection and the chronic immunosuppression required to prevent rejection. However, clinical use of IUHCTx has been hampered by poor engraftment, possibly due to a host immune response against the graft. Since the fetal immune system is relatively immature, we hypothesized that maternal cells trafficking into the fetus may pose the true barrier to effective IUHCTx. Here, we have demonstrated that there is macrochimerism of maternal leukocytes in the blood of unmanipulated mouse fetuses, with substantial increases in T cell trafficking after IUHCTx. To determine the contribution of these maternal lymphocytes to rejection after IUHCTx, we bred T and/or B cell-deficient mothers to wild-type fathers and performed allogeneic IUHCTx into the immunocompetent fetuses. There was a marked improvement in engraftment if the mother lacked T cells but not B cells, indicating that maternal T cells are the main barrier to engraftment. Furthermore, when the graft was matched to the mother, there was no difference in engraftment between syngeneic and allogeneic fetal recipients. Our study suggests that the clinical success of IUHCTx may be improved by transplanting cells matched to the mother.


PLOS Biology | 2011

Pancreatic mesenchyme regulates epithelial organogenesis throughout development.

Limor Landsman; Amar Nijagal; Theresa J. Whitchurch; Renee L. VanderLaan; Warren E. Zimmer; Tippi C. MacKenzie; Matthias Hebrok

Genetic disruption of the pancreatic mesenchyme reveals that it is critical for the expansion of epithelial progenitors and for the proliferation of insulin-producing beta cells.


Molecular and Cellular Biology | 2006

Normal immune development and glucocorticoid-induced thymocyte apoptosis in mice deficient for the T-cell death-associated gene 8 receptor.

Caius G. Radu; Donghui Cheng; Amar Nijagal; Mireille Riedinger; Jami McLaughlin; Li V. Yang; James Johnson; Owen N. Witte

ABSTRACT T-cell death-associated gene 8 (TDAG8) is a G-protein-coupled receptor transcriptionally upregulated by glucocorticoids (GCs) and implicated by overexpression studies in psychosine-mediated inhibition of cytokinesis and in GC-induced apoptosis. To examine the physiological function of TDAG8, we generated knockout (KO) mice by homologous recombination. An enhanced green fluorescent protein reporter was knocked into the disrupted tdag8 locus to allow the analysis of TDAG8 expression in living cells. Interestingly, we found that during thymocyte development, TDAG8 expression resembled the dynamic regulation described for known modulators of GC-induced apoptosis, including Bcl-2, Notch1, and GC receptor. TDAG8 was expressed in double-negative cells, was downregulated at the double-positive transition, and was upregulated in single-positive thymocytes. However, despite this striking expression pattern, maturation and selection of thymocytes, as well as major immune functions, were not affected in TDAG8 KO mice. In contrast to previous overexpression results, TDAG8 was dispensable for psychosine-induced formation of multinucleated cells. Furthermore, TDAG8 KO thymocytes showed normal apoptosis following in vivo and in vitro GC treatment. These results, while establishing a useful reporter strain to study T-lymphocyte maturation, argue against a critical role for TDAG8 in immune development, psychosine-mediated inhibition of cytokinesis, and GC-induced cell death.


American Journal of Transplantation | 2012

Decreased risk of graft failure with maternal liver transplantation in patients with biliary atresia.

Amar Nijagal; Shannon Fleck; Nancy K. Hills; S. Feng; Qizhi Tang; Sang-Mo Kang; P. Rosenthal; Tippi C. MacKenzie

The presence of maternal cells in offspring may promote tolerance to noninherited maternal antigens (NIMAs). Children with biliary atresia (BA) have increased maternal cells in their livers, which may impact tolerance. We hypothesized that patients with BA would have improved outcomes when receiving a maternal liver. We reviewed all pediatric liver transplants recorded in the SRTR database from 1996 to 2010 and compared BA and non‐BA recipients of maternal livers with recipients of paternal livers for the incidences of graft failure and retransplantation. Rejection episodes after parental liver transplantation were examined for patients transplanted at our institution. BA patients receiving a maternal graft had lower rates of graft failure compared to those receiving a paternal graft (3.7% vs. 10.5%, p = 0.02) and, consequently, fewer episodes of retransplantation (2.7% vs. 7.5%, p = 0.04). These differences were not seen among non‐BA patients or among BA patients who received female deceased donor grafts. In patients transplanted at our institution, paternal liver transplantation was associated with an increased incidence of refractory rejection compared to maternal liver transplantation only in BA. Our data support the concept that maternal cells in BA recipients promote tolerance to NIMAs and may be important in counseling BA patients who require liver transplantation.


Molecular Diagnosis & Therapy | 2006

Molecular markers for gastric adenocarcinoma: an update.

Casandra Anderson; Amar Nijagal; Joseph Kim

Gastric cancer is the second most common cancer worldwide. Treatment of localized gastric cancer relies primarily on surgical intervention, although growing evidence suggests that the addition of chemoradiation may improve disease-free intervals and overall survival. In this regard, the current high rates of recurrence and subsequent poor survival have prompted an ever-increasing use of multimodal strategies, even for early-stage disease. However, these therapies are often limited by debilitating toxicities and varying degrees of response efficacy. As a result, pharmacogenomics, the study of specific genetic and molecular signatures that may be predictive of treatment outcomes, has gained considerable interest. For example, studies have demonstrated that the expression of enzymes involved in the metabolism or conjugation of commonly used chemotherapy agents, such as fluoropyrimidines and cisplatin, can serve as surrogate markers predictive of chemotherapy response. Polymorphisms in the genes encoding these enzymes have also been identified and may further account for altered expression patterns, resulting in varied clinical responses. Future work is necessary to further refine the list of molecular genetic markers and to identify novel markers for prognostic and predictive purposes. Technologies such as microarray analysis may be useful in identifying new molecular genetic markers, and further work may determine whether these markers can be employed to help stratify patients into different multimodal treatment regimens.


Blood | 2013

Direct and indirect antigen presentation lead to deletion of donor-specific T cells after in utero hematopoietic cell transplantation in mice

Amar Nijagal; Chris Derderian; Tom Le; Erin Jarvis; Linda Nguyen; Qizhi Tang; Tippi C. MacKenzie

In utero hematopoietic cell transplantation (IUHCTx) is a promising method to induce donor-specific tolerance but the mechanisms of antigen presentation that educate host T cells and the relative importance of deletion vs regulation in this setting are unknown. We studied the roles of direct and indirect antigen presentation (mediated by donor- and host-derived antigen-presenting cells [APCs], respectively) in a mouse model of IUHCTx. We found that IUHCTx leads to precocious maturation of neonatal host dendritic cells (DCs) and that there is early differentiation of donor-derived DCs, even after transplantation of a stem cell source without mature APCs. We next performed allogeneic IUHCTx into donor-specific T-cell receptor transgenic mice and confirmed that both direct and indirect antigen presentation lead to clonal deletion of effector T cells in chimeras. Deletion did not persist when chimerism was lost. Importantly, although the percentage of regulatory T cells (Tregs) after IUHCTx increased, there was no expansion in Treg numbers. In wild-type mice, there was a similar deletion of effector cells without expansion of donor-specific Tregs. Thus, tolerance induction after IUHCTx depends on both direct and indirect antigen presentation and is secondary to thymic deletion, without de novo Treg induction.


Journal of Pediatric Surgery | 2012

Long-term outcomes after fetal therapy for congenital high airway obstructive syndrome

Payam Saadai; Eric B. Jelin; Amar Nijagal; Samuel C. Schecter; Shinjiro Hirose; Tippi C. MacKenzie; Larry Rand; Ruth B. Goldstein; Jody A. Farrell; Michael R. Harrison; Hanmin Lee

BACKGROUND/PURPOSE Congenital high airway obstructive syndrome (CHAOS) is a rare and devastating condition that is uniformly fatal without fetal intervention. We sought to describe fetal treatment and long-term outcomes of CHAOS at a single referral center. METHODS The medical records of patients with fetal CHAOS evaluated at our center between 1993 and 2011 were reviewed. Maternal history, radiographic findings, antenatal management, and postnatal outcomes were compared. RESULTS Twelve fetuses with CHAOS were identified. Eleven had concomitant hydrops at diagnosis. Six were electively terminated, and 2 had intra- or peripartum demise. Four patients underwent fetal intervention. Two underwent delivery via ex utero intrapartum treatment (EXIT) procedure with tracheostomy placement only, and 2 underwent fetal bronchoscopy with attempted wire tracheoplasty followed by EXIT with tracheostomy at delivery. All 4 patients who underwent EXIT were alive at last follow-up. One patient was ventilator and tracheostomy free and feeding by mouth. CONCLUSION Long-term and tracheostomy-free survival is possible with appropriate fetal intervention even in the presence of hydrops. Fetal intervention earlier in pregnancy may improve long-term outcomes, but patient selection for intervention remains challenging. Magnetic resonance imaging may help select those patients for whom fetal intervention before EXIT delivery may be beneficial.


Journal of Immunology | 2014

Fetal Intervention Increases Maternal T Cell Awareness of the Foreign Conceptus and Can Lead to Immune-Mediated Fetal Demise

Marta Wegorzewska; Amar Nijagal; Charissa M. Wong; Tom Le; Ninnia Lescano; Qizhi Tang; Tippi C. MacKenzie

Fetal interventions to diagnose and treat congenital anomalies are growing in popularity but often lead to preterm labor. The possible contribution of the maternal adaptive immune system to postsurgical pregnancy complications has not been explored. We recently showed that fetal intervention in mice increases maternal T cell trafficking into the fetus and hypothesized that this process also may lead to increased maternal T cell recognition of the foreign conceptus and subsequent breakdown in maternal–fetal tolerance. In this study, we show that fetal intervention in mice results in accumulation of maternal T cells in the uterus and that these activated cells can produce effector cytokines. In adoptive transfer experiments, maternal T cells specific for a fetal alloantigen proliferate after fetal intervention, escape apoptosis, and become enriched compared with endogenous T cells in the uterus and uterine-draining lymph nodes. Finally, we demonstrate that such activation and accumulation can have a functional consequence: in utero transplantation of hematopoietic cells carrying the fetal alloantigen leads to enhanced demise of semiallogeneic fetuses within a litter. We further show that maternal T cells are necessary for this phenomenon. These results suggest that fetal intervention enhances maternal T cell recognition of the fetus and that T cell activation may be a culprit in postsurgical pregnancy complications. Our results have clinical implications for understanding and preventing complications associated with fetal surgery such as preterm labor.


Clinics in Perinatology | 2012

In Utero Hematopoietic Cell Transplantation for the Treatment of Congenital Anomalies

Amar Nijagal; Alan W. Flake; Tippi C. MacKenzie

In utero hematopoietic cell transplantation (IUHCTx) is a promising strategy for the treatment of common hematopoietic disorders and for inducing immune tolerance in the fetus. Although the efficacy of IUHCTx has been demonstrated in multiple small and large animal models, the clinical application of this technique in humans has had limited success. Recent studies in mice have demonstrated that the maternal immune system plays a critical role in limiting engraftment in the fetus. This article reviews the therapeutic rationale of IUHCTx, potential barriers to its applications, and recent experimental strategies to improve its clinical success.


Journal of Pediatric Surgery | 2012

Alterations in maternal-fetal cellular trafficking after fetal surgery

Payam Saadai; Tzong Hae Lee; Geoanna Bautista; Kelly D. Gonzales; Amar Nijagal; Michael P. Busch; Chong Jai Kim; Roberto Romero; Hanmin Lee; Shinjiro Hirose; Larry Rand; Douglas N. Miniati; Diana L. Farmer; Tippi C. MacKenzie

BACKGROUND/PURPOSE Bidirectional trafficking of cells between the mother and the fetus is routine in pregnancy and a component of maternal-fetal tolerance. Changes in fetal-to-maternal cellular trafficking have been reported in prenatal complications, but maternal-to-fetal trafficking has never been studied in the context of fetal intervention. We hypothesized that patients undergoing open fetal surgery would have altered maternal-fetal cellular trafficking. METHODS Cellular trafficking was analyzed in patients with myelomeningocele (MMC) who underwent open fetal surgical repair (n = 5), patients with MMC who had routine postnatal repair (n = 6), and healthy control healthy patients (n = 9). As an additional control for the fetal operation, trafficking was also analyzed in patients who were delivered by an ex utero intrapartum treatment procedure (n = 6). Microchimerism in maternal and cord blood was determined using quantitative real-time polymerase chain reaction for nonshared alleles. RESULTS Maternal-to-fetal trafficking was significantly increased in patients who underwent open fetal surgery for MMC compared with healthy controls, patients who underwent postnatal MMC repair, and patients who underwent ex utero intrapartum treatment. There were no differences in fetal-to-maternal cell trafficking among groups. CONCLUSION Patients undergoing open fetal surgery for MMC have elevated levels of maternal microchimerism. These results suggest altered trafficking and/or increased proliferation of maternal cells in fetal blood and may have important implications for preterm labor.

Collaboration


Dive into the Amar Nijagal's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tom Le

University of California

View shared research outputs
Top Co-Authors

Avatar

Qizhi Tang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hanmin Lee

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew Tucker

University of California

View shared research outputs
Top Co-Authors

Avatar

Caius G. Radu

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric B. Jelin

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge