Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Cell Rep ; 10(11): 1793-802, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25801020

ABSTRACT

Embryonic stem cells (ESCs) hold promise for the treatment of many medical conditions; however, their utility is limited by immune rejection. The objective of our study is to establish tolerance or promote engraftment of transplanted ESCs as well as mature cell populations derived from ESCs. Luciferase (luc(+))-expressing ESCs were utilized to monitor the survival of the ESCs and differentiated progeny in living recipients. Allogeneic recipients conditioned with fractioned total lymphoid irradiation (TLI) and anti-thymocyte serum (ATS) or TLI plus regulatory T cells (T(reg)) promoted engraftment of ESC allografts after transplantation. Following these treatments, the engraftment of transplanted terminally differentiated endothelial cells derived from ESCs was also significantly enhanced. Our findings provide clinically translatable strategies of inducing tolerance to adoptively transferred ESCs for cell replacement therapy of medical disorders.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/transplantation , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/immunology , Endothelial Cells/cytology , Lymphatic Irradiation/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Stem Cell Transplantation/methods
2.
Chem Biol ; 21(10): 1278-1292, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25308275

ABSTRACT

Store-operated calcium (SOC) channels are vital for activation of the immune cells, and mutations in the channel result in severe combined immunodeficiency in human patients. In lymphocytes, SOC entry is mediated by the Orai1 channel, which is activated by direct binding of STIM1. Here we describe an alternative approach for identifying inhibitors of SOC entry using minimal functional domains of STIM1 and Orai1 to screen a small-molecule microarray. This screen identified AnCoA4, which inhibits SOC entry at submicromolar concentrations and blocks T cell activation in vitro and in vivo. Biophysical studies revealed that AnCoA4 binds to the C terminus of Orai1, directly inhibiting calcium influx through the channel and also reducing binding of STIM1. AnCoA4, unlike other reported SOC inhibitors, is a molecule with a known binding site and mechanism of action. These studies also provide proof of principle for an approach to ion channel drug discovery.


Subject(s)
Benzodioxoles/therapeutic use , Chromones/therapeutic use , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Small Molecule Libraries/chemistry , Animals , Benzodioxoles/chemistry , Benzodioxoles/pharmacology , Chromones/chemistry , Chromones/pharmacology , Disease Models, Animal , Drosophila , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Fura-2/chemistry , Gene Expression/drug effects , HEK293 Cells , Humans , Hypersensitivity, Delayed/drug therapy , Hypersensitivity, Delayed/metabolism , Hypersensitivity, Delayed/pathology , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Lymphocyte Activation/drug effects , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Molecular Docking Simulation , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Stromal Interaction Molecule 1 , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
3.
Blood ; 124(11): 1832-42, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25030062

ABSTRACT

Acute graft-versus-host disease (GVHD) is induced by alloreactivity of donor T cells toward host antigens presented on antigen-presenting cells (APCs). Apoptotic cells are capable of inducing tolerance by altering APC maturation. Apoptosis can be induced by extracorporeal photopheresis (ECP). We demonstrate that the use of ECP as a prophylaxis prior to conditioning significantly improves survival (P < .0001) after bone marrow transplantation (BMT) by inhibiting the initiation phase of acute GVHD in a murine BMT model. ECP-treated autologous splenocytes resulted in immune tolerance in the host, including reduced dendritic cell activation with decreased nuclear factor-κB engagement, increased regulatory T-cell (Treg) numbers with enhanced expression of cytolytic T lymphocyte-associated antigen 4, potentiating their suppressive function. The protective effect required host production of interleukin-10 and host Tregs. Conventional T cells that entered this tolerant environment experienced reduced proliferation, as well as a reduction of tissue homing and expression of activation markers. The induction of this tolerant state by ECP was obviated by cotreatment with lipopolysaccharide, suggesting that the inflammatory state of the recipient prior to treatment would play a role in potential clinical translation. The use of prophylactic ECP may provide an alternative and safe method for immunosuppression in the bone marrow transplant setting.


Subject(s)
Antigen-Presenting Cells/transplantation , Apoptosis , Graft vs Host Disease/prevention & control , Immune Tolerance , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/pathology , Autografts , Bone Marrow Transplantation , Disease Models, Animal , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Interleukin-10/genetics , Interleukin-10/immunology , Mice , Mice, Inbred AKR , Mice, Inbred BALB C , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
4.
PLoS One ; 9(1): e86551, 2014.
Article in English | MEDLINE | ID: mdl-24475140

ABSTRACT

Lag-3 has emerged as an important molecule in T cell biology. We investigated the role of Lag-3 in conventional T cell (Tcon) and regulatory T cell (Treg) function in murine GVHD with the hypothesis that Lag-3 engagement diminishes alloreactive T cell responses after bone marrow transplantation. We demonstrate that Lag-3 deficient Tcon (Lag-3(-/-) Tcon) induce significantly more severe GVHD than wild type (WT) Tcon and that the absence of Lag-3 on CD4 but not CD8 T cells is responsible for exacerbating GVHD. Lag-3(-/-) Tcon exhibited increased activation and proliferation as indicated by CFSE and bioluminescence imaging analyses and higher levels of activation markers such as CD69, CD107a, granzyme B, and Ki-67 as well as production of IL-10 and IFN-g early after transplantation. Lag-3(-/-) Tcon were less responsive to suppression by WT Treg as compared to WT Tcon. The absence of Lag-3, however, did not impair Treg function as both Lag-3(-/-) and WT Treg equally suppress the proliferation of Tcon in vitro and in vivo and protect against GVHD. Further, we demonstrate that allogeneic Treg acquire recipient MHC class II molecules through a process termed trogocytosis. As MHC class II is a ligand for Lag-3, we propose a novel suppression mechanism employed by Treg involving the acquisition of host MHC-II followed by the engagement of Lag-3 on T cells. These studies demonstrate for the first time the biologic function of Lag-3 expression on conventional and regulatory T cells in GVHD and identify Lag-3 as an important regulatory molecule involved in alloreactive T cell proliferation and activation after bone marrow transplantation.


Subject(s)
Antigens, CD/immunology , Bone Marrow Transplantation/adverse effects , Graft vs Host Disease/immunology , T-Lymphocytes/immunology , Transplantation, Homologous/adverse effects , Analysis of Variance , Animals , Antigens, CD/genetics , Cell Proliferation , Flow Cytometry , Fluoresceins , Luminescent Measurements , Mice , Mice, Knockout , Succinimides , Lymphocyte Activation Gene 3 Protein
5.
Blood ; 122(22): 3659-65, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24030387

ABSTRACT

To investigate the role of mast cells in hematopoietic cell transplantation, we assessed graft-versus-host disease (GVHD) in C57BL/6-Kit(W-sh/W-sh) recipients, which virtually lack mast cells, compared with C57BL/6 WT recipients. GVHD was severely exacerbated in C57BL/6-Kit(W-sh/W-sh) mice (median survival time = 13 vs 60 days in wild-type [WT] mice; P < .0001). The increased mortality risk in C57BL/6-Kit(W-sh/W-sh) hosts correlated with increased T-cell numbers in lymph nodes, liver, and gastrointestinal tract sites, as indicated by bioluminescence imaging (P < .001). We did not detect any deficit in the number or function of CD4(+)CD25(+) regulatory T cells (Tregs) in C57BL/6-Kit(W-sh/W-sh) mice. Furthermore, Tregs were equally effective at reducing GVHD in C57BL/6-Kit(W-sh/W-sh) recipients compared with WT recipients containing mast cells. Furthermore, we found that survival of C57BL/6-Kit(W-sh/W-sh) mice during GVHD was significantly improved if the mice were engrafted with bone marrow-derived cultured mast cells from WT C57BL/6 mice but not from interleukin (IL)-10-deficient C57BL/6 mice. These data indicate that the presence of mast cells can significantly reduce GVHD independently of Tregs, by decreasing conventional T-cell proliferation in a mechanism involving IL-10. These experiments support the conclusion that mast cells can mediate a novel immunoregulatory role during hematopoietic cell transplantation.


Subject(s)
Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Mast Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Proliferation , Cell Survival , Female , Graft vs Host Disease/pathology , Hematopoietic Stem Cell Transplantation/adverse effects , Immune Tolerance , Interleukin-10/biosynthesis , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Male , Mast Cells/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/immunology , T-Lymphocytes, Regulatory/classification
6.
Biol Blood Marrow Transplant ; 19(11): 1557-65, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23921175

ABSTRACT

Regulatory T cell (Treg) immunotherapy is a promising strategy for the treatment of graft rejection responses and autoimmune disorders. Our and other laboratories have shown that the transfer of highly purified CD4(+)CD25(+)Foxp3(+) natural Treg can prevent lethal graft-versus-host disease (GVHD) after allogeneic hematopoietic cell transplantation across both major and minor histocompatibility barriers. However, recent evidence suggests that the Treg suppressive phenotype can become unstable, a phenomenon that can culminate in Treg conversion into IL-17-producing cells. We hypothesized that the intense proinflammatory signals released during an ongoing alloreaction might redirect a fraction of the transferred Treg to the Th17 cell fate, thereby losing immunosuppressive potential. We therefore sought to evaluate the impact of Il17 gene ablation on Treg stability and immunosuppressive capacity in a major MHC mismatch model. We show that although Il17 gene ablation results in a mildly enhanced Treg immunosuppressive ability in vitro, such improvement is not observed when IL-17-deficient Treg are used for GVHD suppression in vivo. Similarly, when we selectively blocked IL-1 signaling in Treg, that was shown to be necessary for Th17 conversion, we did not detect any improvement on Treg-mediated GVHD suppressive ability in vivo. Furthermore, upon ex vivo reisolation of transferred wild-type Treg, we detected little or no Treg-mediated IL-17 production upon GVHD induction. Our results indicate that blocking Th17 conversion does not affect the GVHD suppressive ability of highly purified natural Treg in vivo, suggesting that IL-17 targeting is not a valuable strategy to improve Treg immunotherapy after hematopoietic cell transplantation.


Subject(s)
Bone Marrow Transplantation/methods , Graft vs Host Disease/immunology , Immunotherapy, Adoptive/methods , Interleukin-17/genetics , T-Lymphocytes, Regulatory/transplantation , Animals , Graft vs Host Disease/genetics , Humans , Interleukin-17/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
7.
Blood ; 118(8): 2342-50, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21734238

ABSTRACT

Previous work has demonstrated that both rapamycin (RAPA) and IL-2 enhance CD4⁺CD25⁺Foxp3⁺ regulatory T-cell (Treg) proliferation and function in vitro. We investigated whether the combination of RAPA plus IL-2 could impact acute GVHD induction after bone marrow transplantation (BMT). RAPA plus IL-2 resulted in improved survival and a reduction in acute GVHD lethality associated with an increased expansion of donor type CD4⁺Foxp3⁺ Tregs and reduced CD4⁺CD25⁻ conventional T cells (Tcons). RAPA plus IL-2, but not either drug alone, increased both expansion of donor natural Tregs and conversion of induced Tregs from donor CD25⁻ Tcons while IL-2 alone increased conversion of Tregs from CD25⁻ Tcon. RAPA plus IL-2 treatment resulted in less production of IFN-γ and TNF, cytokines known to be important in the initiation of acute GVHD. These studies indicate that the pharmacologic stimulation of T cells with IL-2 and the suppression of Tcon proliferation with RAPA result in a selective expansion of functional Tregs and suppression of acute GVHD.


Subject(s)
Graft vs Host Disease/prevention & control , Interleukin-2/pharmacology , Sirolimus/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Acute Disease , Animals , Bone Marrow Transplantation/adverse effects , Bone Marrow Transplantation/immunology , Female , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/immunology , Interferon-gamma/biosynthesis , Interleukin-2/administration & dosage , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Sirolimus/administration & dosage , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/classification , Transplantation, Homologous , Tumor Necrosis Factor-alpha/biosynthesis
8.
Cell Stem Cell ; 8(3): 309-17, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21362570

ABSTRACT

Embryonic stem cells (ESCs) are an attractive source for tissue regeneration and repair therapies because they can be differentiated into virtually any cell type in the adult body. However, for this approach to succeed, the transplanted ESCs must survive long enough to generate a therapeutic benefit. A major obstacle facing the engraftment of ESCs is transplant rejection by the immune system. Here we show that blocking leukocyte costimulatory molecules permits ESC engraftment. We demonstrate the success of this immunosuppressive therapy for mouse ESCs, human ESCs, mouse induced pluripotent stem cells (iPSCs), human induced pluripotent stem cells, and more differentiated ESC/(iPSCs) derivatives. Additionally, we provide evidence describing the mechanism by which inhibition of costimulatory molecules suppresses T cell activation. This report describes a short-term immunosuppressive approach capable of inducing engraftment of transplanted ESCs and iPSCs, providing a significant improvement in our mechanistic understanding of the critical role costimulatory molecules play in leukocyte activation.


Subject(s)
Embryonic Stem Cells/transplantation , Immunosuppression Therapy , Induced Pluripotent Stem Cells/transplantation , Animals , Antigens, CD/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/immunology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Graft Rejection/immunology , Humans , Immunosuppressive Agents/pharmacology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/immunology , Leukocytes/cytology , Leukocytes/drug effects , Leukocytes/metabolism , Mice , Time Factors , Transplantation, Heterologous , Transplantation, Homologous
9.
Blood ; 117(11): 3220-9, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21258007

ABSTRACT

CD4(+) natural killer T (NKT) cells, along with CD4(+)CD25(+) regulatory T cells (Tregs), are capable of controlling aberrant immune reactions. We explored the adoptive transfer of highly purified (> 95%) CD4(+)NKT cells in a murine model of allogeneic hematopoietic cell transplantation (HCT). NKT cells follow a migration and proliferation pattern similar to that of conventional T cells (Tcons), migrating initially to secondary lymphoid organs followed by infiltration of graft-versus-host disease (GVHD) target tissues. NKT cells persist for more than 100 days and do not cause significant morbidity or mortality. Doses of NKT cells as low as 1.0 × 10(4) cells suppress GVHD caused by 5.0 × 10(5) Tcons in an interleukin-4 (IL-4)-dependent mechanism. Protective doses of NKT cells minimally affect Tcon proliferation, but cause significant reductions in interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α) production by donor Tcons and in skin, spleen, and gastrointestinal pathology. In addition, NKT cells do not impact the graft-versus-tumor (GVT) effect of Tcons against B-cell lymphoma-1 (BCL-1) tumors. These studies elucidate the biologic function of donor-type CD4(+)NKT cells in suppressing GVHD in an allogeneic transplantation setting, demonstrating clinical potential in reducing GVHD in HCT.


Subject(s)
Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Interleukin-4/immunology , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Acute Disease , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Cell Proliferation , Inflammation Mediators/metabolism , Interferon-gamma/biosynthesis , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-4/biosynthesis , Mice , Organ Specificity , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/biosynthesis
10.
Blood ; 115(21): 4293-301, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20233969

ABSTRACT

Natural killer (NK) cells suppress graft-versus-host disease (GVHD) without causing GVHD themselves. Our previous studies demonstrated that allogeneic T cells and NK cells traffic similarly after allogeneic bone marrow transplantation (BMT). We therefore investigated the impact of donor NK cells on donor alloreactive T cells in GVHD induction. Animals receiving donor NK and T cells showed improved survival and decreased GVHD score compared with controls receiving donor T cells alone. Donor T cells exhibited less proliferation, lower CD25 expression, and decreased interferon-gamma (IFN-gamma) production in the presence of NK cells. In vivo, we observed perforin- and Fas ligand (FasL)-mediated reduction of donor T cell proliferation and increased T cell apoptosis in the presence of NK cells. Further, activated NK cells mediated direct lysis of reisolated GVHD-inducing T cells in vitro. The graft-versus-tumor (GVT) effect was retained in the presence of donor NK cells. We demonstrate a novel mechanism of NK cell-mediated GVHD reduction whereby donor NK cells inhibit and lyse autologous donor T cells activated during the initiation of GVHD.


Subject(s)
Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Graft vs Tumor Effect/immunology , Killer Cells, Natural/immunology , T-Lymphocytes/immunology , Animals , Apoptosis , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Fas Ligand Protein/deficiency , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Female , Inflammation Mediators/metabolism , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Killer Cells, Natural/transplantation , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pore Forming Cytotoxic Proteins/deficiency , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/immunology , T-Lymphocytes/cytology , T-Lymphocytes/transplantation , T-Lymphocytes, Regulatory/immunology , Transplantation, Homologous
11.
Blood ; 111(1): 453-62, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17967941

ABSTRACT

Based on their ability to control T-cell homeostasis, Foxp3(+)CD4(+)CD25(+) regulatory T cells (Tregs) are being considered for treatment of autoimmune disorders and acute graft-versus-host disease (aGVHD). When combining Tregs with the immunosuppressant rapamycin (RAPA), we observed reduced alloreactive conventional T-cell (Tconv) expansion and aGVHD lethality compared with each treatment alone. This synergistic in vivo protection was paralleled by intact expansion of polyclonal Tregs with conserved high FoxP3 expression. In contrast to Tconv, activation of Tregs with alloantigen and interleukin-2 preferentially led to signal transducer and activator of transcription 5 (STAT5) phosphorylation and not phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway activity. Expression of phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a negative regulator of the PI3K/Akt/mTOR pathway, remained high in Tregs but not Tconv during stimulation. Conversely, targeted deletion of PTEN increased susceptibility of Tregs to mTOR inhibition by RAPA. Differential impact of RAPA as a result of reduced usage of the mTOR pathway in Tregs compared with conventional T cells explains the synergistic effect of RAPA and Tregs in aGVHD protection, which has important implications for clinical trials using Tregs.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/drug therapy , Immunosuppressive Agents/pharmacology , Protein Kinases/metabolism , Sirolimus/pharmacology , T-Lymphocytes, Regulatory/drug effects , Animals , CD4-Positive T-Lymphocytes/cytology , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Hematopoietic Stem Cell Transplantation/adverse effects , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT5 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/cytology , TOR Serine-Threonine Kinases
12.
Biochemistry ; 43(44): 14257-64, 2004 Nov 09.
Article in English | MEDLINE | ID: mdl-15518576

ABSTRACT

The transporter associated with antigen presentation (TAP) is an ATP-binding cassette (ABC) protein which transports peptides for presentation to the immune system. TAP is composed of two half transporters, TAP1 (ABCB2) and TAP2 (ABCB3), which heterodimerize to function. In humans, the TAP family consists of TAP1, TAP2, and TAPL (ABCB9). While the TAP1-TAP2 complex is well characterized, TAPL's dimerization state and function are unknown. To identify interactions within the human TAP family, we adapted the dihydrofolate reductase protein-fragment complementation assay (DHFR PCA) to half ABC transporters. This assay has been shown to be suitable for the study of membrane-bound proteins in vivo [Remy, I., Wilson, I. A., and Michnick, S. W. (1999) Science 283, 990-993]. With this method, in vivo TAP1-TAP2 heterodimerization was confirmed, no homodimerizations were detected with TAP1 or TAP2, and TAPL did not show any interaction with TAP1 or TAP2. However, we found strong evidence that TAPL forms homodimers. These results provide evidence of a novel homomeric TAPL interaction and demonstrate that the DHFR PCA will be of general utility in studies of half ABC transporter interactions in vivo.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Multigene Family , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP Binding Cassette Transporter, Subfamily B, Member 3 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/physiology , Animals , Antigen Presentation/genetics , CHO Cells , Cell Survival/genetics , Clone Cells , Cricetinae , Dimerization , False Positive Reactions , Flow Cytometry , Genetic Complementation Test , Humans , Mice , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding/genetics , Protein Interaction Mapping , RNA, Messenger/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL