@prefix vivo: . @prefix edm: . @prefix ns0: . @prefix dcterms: . @prefix skos: . vivo:departmentOrSchool "Science, Faculty of"@en, "Zoology, Department of"@en ; edm:dataProvider "DSpace"@en ; ns0:degreeCampus "UBCV"@en ; dcterms:creator "Johnson, Laura Alexandra"@en ; dcterms:issued "2009-12-23T22:52:22Z"@en, "2004"@en ; vivo:relatedDegree "Doctor of Philosophy - PhD"@en ; ns0:degreeGrantor "University of British Columbia"@en ; dcterms:description "The ATP-binding cassette (ABC) transporter TAP (ABCB2/3) is an integral component of the endogenous antigen processing pathway. Without TAP, antigenic peptides cannot enter the ER to bind to class I MHC molecules en route to the cell surface, and as a result, no cellmediated immune response can occur. All normal cells can present endogenous antigens, but exogenous antigen presentation is conducted solely by professional antigen presenting cells (APCs). The precise mechanism of intracellular transport of these exogenous antigens remains unknown. As TAP plays an integral role in endogenous peptide transport, it is conceivable that other ABC transporters play a similar role in exogenous antigen processing. Using RT-PCR, gene expression of all 46 known murine ABC transporters was assayed in APCs and non-APCs in response to various stimulatory cytokines and exogenous antigens. A pattern emerged, whereby APCs induced many specific ABC genes compared to non-APCs, and activated dendritic cells (DCs) expressed the highest number. Incorporating information based on the evolutionary phylogeny of sequence similarity to known antigen transporters, 18 candidate antigen presenting genes were identified. The ABCB9 transporter gene shared the highest sequence homology to the TAP genes, was differentially expressed in activated DCs, and localized to endosome and lysosomal compartments. An ABCB9[sup -/-] mouse was made and characterized for an immunological phenotype. While no defect in live viral immune response was observed, a defective T-cell response to cross-presented exogenous antigens such as ovalbumin and heat-inactivated Sendai virus was discovered, but not to MHC class-II restricted antigens. This defect was traced back to an inability of ABCB9[sup -/-] DCs to present these antigens on the cell surface, and was consistent with a lack of antigen transport between the endo/lysosome and the cytoplasm, as introduction of endogenous antigen corrected the defect. The contribution of the ABCB9 gene to cancer immunology was then evaluated, and it was found that upon challenge with live tumour cells, pre-immunized ABCB9[sup -/-] mice were no more susceptible than wild-type mice. Thus, exogenous antigen presentation is not the primary method by which a cellular anti-tumour response is primed in-vivo."@en ; edm:aggregatedCHO "https://circle.library.ubc.ca/rest/handle/2429/17320?expand=metadata"@en ; skos:note "T H E A B C s OF A N T I G E N PRESENTAT ION: DEF IN ING A N T I G E N PROCESS ING P A T H W A Y S by L A U R A A L E X A N D R A JOHNSON B.Sc, Simon Fraser University, 1998 M.Sc, The University of British Columbia, 2000 A THESIS SUBM ITTED IN PART IAL F U L F I L M E N T OF T H E REQU IREMENTS FOR THE D E G R E E OF D O C T O R OF PH ILOSOPHY in THE F A C U L T Y OF G R A D U A T E STUDIES D E P A R T M E N T OF Z O O L O G Y A N D B I O T E C H N O L O G Y L A B O R A T O R Y We accept this thesis as conforming , to the required standard THE UN IVERS ITY OF BRITISH C O L U M B I A O C T O B E R 2004 © Laura Alexandra Johnson, 2004 A B S T R A C T The ATP-binding cassette (ABC) transporter TAP (ABCB2/3) is an integral component of the endogenous antigen processing pathway. Without TAP, antigenic peptides cannot enter the ER to bind to class I M H C molecules en route to the cell surface, and as a result, no cell-mediated immune response can occur. A l l normal cells can present endogenous antigens, but exogenous antigen presentation is conducted solely by professional antigen presenting cells (APCs). The precise mechanism of intracellular transport of these exogenous antigens remains unknown. As TAP plays an integral role in endogenous peptide transport, it is conceivable that other A B C transporters play a similar role in exogenous antigen processing. Using RT-PCR, gene expression of all 46 known murine A B C transporters was assayed in APCs and non-APCs in response to various stimulatory cytokines and exogenous antigens. A pattern emerged, whereby APCs induced many specific A B C genes compared to non-APCs, and activated dendritic cells (DCs) expressed the highest number. Incorporating information based on the evolutionary phylogeny of sequence similarity to known antigen transporters, 18 candidate antigen presenting genes were identified. The A B C B 9 transporter gene shared the highest sequence homology to the T A P genes, was differentially expressed in activated DCs, and localized to endosome and lysosomal compartments. An ABCB9\" 7 \" mouse was made and characterized for an immunological phenotype. While no defect in live viral immune response was observed, a defective T-cell response to cross-presented exogenous antigens such as ovalbumin and heat-inactivated Sendai virus was discovered, but not to M H C class-II restricted antigens. This defect was traced back to an inability of ABCB9\"~ DCs to present these antigens on the cell surface, and was consistent i i with a lack of antigen transport between the endo/lysosome and the cytoplasm, as introduction of endogenous antigen corrected the defect. The contribution of the A B C B 9 gene to cancer immunology was then evaluated, and it was found that upon challenge with live tumour cells, pre-immunized A B C B 9 / _ mice were no more susceptible than wild-type mice. Thus, exogenous antigen presentation is not the primary method by which a cellular anti-tumour response is primed in-vivo. i i i T A B L E OF CONTENTS Abstract ii Table of Contents iv List of Tables vi List of Figures vi List of Abbreviations viii Acknowledgements xii CHAPTER I WHAT DO A B C TRANSPORTERS H A V E TO DO WITH ANTIGEN PRESENTATION? 1 1.1 Introduction 2 1.2 Antigen presentation 3 1.3 ATP-binding cassette transporters 14 1.4 Rationale and hypotheses 15 1.5 Goals 18 1.6 Bibliography 20 CHAPTER II EXPRESSION PROFILING OF ABC TRANSPORTERS IN APCs 24 2.1 Overview 25 2.2 Introduction 26 2.3 Materials and methods 27 2.4 Results 30 2.5 Discussion 41 2.6 Conclusions 50 2.7 Future directions 51 2.8 Bibliography 53 CHAPTER III IMMUNOLOGICAL CHARACTERIZATION OF THE ABCB9 KNOCKOUT MOUSE 58 3.1 Overview 59 iv 3.2 Introduction 60 3.3 Materials and methods 61 3.4 Results 65 3.5 Discussion 81 3.6 Bibliography 85 C H A P T E R IV E X O G E N O U S CROSS-PR IMING A N D T U M O U R I M M U N O L O G Y 87 4.1 Overview 88 4.2 Introduction 89 4.3 Materials and methods 92 4.4 Results 94 4.5 Discussion 105 4.6 Conclusions 107 4.7 Future directions 107 4.8 Bibliography 109 C H A P T E R V A V I E W TO THE F U T U R E I l l 5.1 In review 112 5.2 A B C B 9 113 5.3 Cross-presentation and cancer immunology 114 5.4 New avenues for A B C B 9 115 5.5 Multiple RT-PCR screen for A B C transporter gene expression 115 5.6 Conclusion 117 5.7 Bibliography 119 A P P E N D I X I M U L T I P L E A B C T R A N S P O R T E R RT-PCR D A T A 121 v LIST OF T A B L E S C H A P T E R II Table 2.1 RT-PCR primer sequences for mouse A B C transporter genes 33 Table 2.2 Differential A B C expression in APCs under various stimuli 37 LIST OF F IGURES C H A P T E R I Figure 1.1 The M H C class I peptide-loading complex 4 Figure 1.2 ER-Phagosome cross-presentation 8 Figure 1.3 Antigen processing and presentation pathways 11 Figure 1.4 Dendritic cell maturation markers 13 C H A P T E R II Figure 2.1 Cytokine treatment induces antigen presenting A B C transporter expression 31 Figure 2.2. RT-PCR of untreated and TNFa treated DCs 32 Figure 2.3 Splenic D C activation markers 35 Figure 2.4 Phylogenetic analysis of A B C transporter protein superfamily 40 C H A P T E R III Figure 3.1 A B C B 9 gene knockout construct and tissue expression 66 Figure 3.2 Cellular profiles of ABCB9\" 7 \" mice 68 Figure 3.3 ABCB9~'~ mice generate normal C T L response to live virus 70 Figure 3.4 A B C B 9 m i c e generate a deficient C T L response to killed virus 72 Figure 3.5 HI Sendai antigen processing is proteasome-dependent 73 Figure 3.6 A B C B 9 m i c e have defective D C cross-presentation 75 Figure 3.7 ABCB9\" / \"DCs are defective in ovalbumin cross-presentation 77 Figure 3.8 Endogenously supplied O V A rescues surface ICVOVA expression 78 Figure 3.9 ABCB9 \" \" mice have normal MHC-II exogenous antigen presentation 80 Figure 3.10 Proposed model of A B C B 9 as an endo/lysosomal antigen transporter 83 C H A P T E R IV Figure 4.1 B16 tumour progression 96 Figure 4.2 Survivorship of GV1T.64 injected mice 98 Figure 4.3 Survivorship of CMT.1-4 injected mice 100 Figure 4.4 Survivorship of pre-immunized CMT.64 injected mice 102 Figure 4.5 Survivorship of pre-immunized CMT. 1 -4 injected mice 104 vi i LIST OF ABBREV IAT IONS (RT)-PCR (Reverse transcriptase)-polymerase chain reaction Ab Antibody A B C ATP-binding cassette A P C Antigen presenting cell A T P Adenosine triphosphate B04 Helper T-cell clone recognizing H E L peptide on I-Ab B16 Murine melanoma cell line (B6 background) B220 B-cell specific marker B3Z C T L clone recognizing O V A peptide on H2-K b B-cell Bone-marrow derived lymphocyte C57BL/6 B6 background inbred mouse C D Cellular differentiation marker C D l l b M 0 marker (also found on some DCs) C D l l c D C specific marker CD19 B-cell specific marker CD3 T-cell specific marker CD4 Helper T-cell marker CD40 D C maturation marker CD8 Cytotoxic T-cell marker CD80 B-7.1 'danger' signal, D C maturation marker CD86 B-7.2 'danger' signal, D C maturation marker c D N A Complementary D N A CIIV Class II vesicle CMT. 1 -4 CMT.64 transfected with TAP 1 CMT.64 Murine SCLC cell line (B6 background) CNS Central nervous system C T L Cytotoxic T-lymphocytes D C Dendritic cell DEC-205 D C surface receptor and maturation marker D N A Deoxyribonucleic acid D T H Delayed type hypersensitivity ER Endoplasmic reticulum E R A A P ER-associated amino peptidase ES cell Embryonic stem cell FITC Fluorescein isothiocyanate fluorescent marker H-2K b (K b) Class I M H C molecule in C57BI76 mice H E L Hen egg lysozyme Hl-Sendai Heat inactivated Sendai (parainfluenza) virus H L A Human leukocyte antigen i.p. Intraperitoneal I-Ab Class II M H C molecule in C57BL/6 mice IB3B Anti-human A B C B 9 specific polyclonal Ab IFNy Interferon gamma IL-2 Interleukin 2 ix IR Immune response IRE Interferon response element L M P Low molecular-weight protein (part of the proteasome) LPS Lipopolysaccharide Ltk Murine fibroblast cell line mAb Monoclonal antibody M D R Multiple drug resistance M H C Major histocompatibility complex MIIC M H C class II compartment M 0 Macrophage NIC Natural killer cell N O D Non-obese diabetic mouse model O V A Chicken egg ovalbumin PE Phycoerythrin fluorescent marker PGP P-glycoprotein R N A Ribonucleic acid s.c. Subcutaneous SCLC Small-cell lung carcinoma Sv/Ev B6 background inbred mouse T A A Tumour associated antigen T A P Transporter associated with antigen presentation T-cell Thymically educated lymphocyte TCR T-cell receptor x TD 5 o Timepoint at which 5 0 % mortality is reached TIL Tumour infiltrating lymphocytes TNFa Tumour necrosis factor alpha V S V Vesicular stomatitus virus V V Vaccinia virus P2-m Beta-2-microglobulin xi A C K N O W L E D G E M E N T S The process of completing this dissertation has been a long and not always easy one. Many challenges had to be overcome, which has resulted in much growth, both personal and professional. Having completed this thesis feels as thought I've walked through a doorway emerging as a different person. There are a number of people who have been very important to me during this time, and I would like to acknowledge their support. For the generation and sharing of the ABCB9 _ / \" mouse, reagents and discussion, I would like to thank following individuals: V. Ling, F. Zhang, R. Wang, A. Chan, and D. Leveson-Gower. Thanks to T. Mak for his generous gift of ES cells, R. Grewal and C.D.Helgason for blastocyst injection in the generation of chimeric mice, and L. Munroe for embryo re-derivation of the A B C B 9 7 \" mouse. 1 thank A. Jeffries and R. Gopaul for mouse breeding and technical assistance. I would like to acknowledge J. Yew dell for the kind gift of ovalbumin vaccinia-virus constructs, J. Blum for protocols, N. Shastri for making the B3Z and B04 T-cell clones available, and R. Germain for providing the 25-D1.16 antibody. Thanks to Q-J. Zhang, G. Basha and G. Lizee for discussions and procedural advice. For endless repetitions of cell purification and RT-PCR for the A B C transporter screen, I thank my project student N. Tracey. Thanks also to technicians L. L i and S. Lok, and to R. Seipp for proofreading and editing my thesis without complaint. 1 also want to thank my supervisor W i l f Jefferies, who has been pivotal in my growth and maturation as a scientist, and has helped guide my decisions with his advice. On a personal level, I want to thank my husband, Ken Johnson, who has always supported my decisions, and encouraged my career without concern for himself, all the while providing a xii warm and welcoming home environment. Last but not least, I would like to thank my parents, Eric and Sandra Merz who got me interested in science at an early age when they gave me my first microscope and chemistry set. Since my childhood, they have always created an environment where my goals were encouraged, but I never felt pressured. To this day, my father remains the one of the most knowledgeable people I have ever met. xi i i C H A P T E R I W H A T DO A B C T R A N S P O R T E R S H A V E TO D O WITH A N T I G E N PRESENTAT ION? C H A P T E R I. W H A T DO A B C T R A N S P O R T E R S H A V E TO D O WITH A N T I G E N PRESENTAT ION? 1.1 Introduction The immune system normally destroys diseased cells in the body by sampling antigens presented in the context of major histocompatibility molecules (MHC) on cell surfaces and generating a T-cell response against those antigens determined to be dangerous (i.e. of viral, bacterial or cancerous origin). These primed T-cells then circulate throughout the body, conducting immuno-surveillance by sampling cell surface antigens to identify and destroy diseased cells. Problems arise when components of this immuno-surveillance mechanism break down. If for some reason diseased cells are unable to present antigens, no antigen-specific immune response wi l l take place, and the disease can progress. Indeed, many cancers and pathogenic infections occur in which known components of the antigen presentation pathway are defective (Ehrlich 1997; Gabathuler, Alimonti et al. 1998; Alimonti, Zhang et al. 2000; Tortorella, Gewurz et al. 2000). Although there are many different pathogens and genetic defects that can cause disease, there appears to be a uniform mechanism of endogenous antigen processing and presentation (Heath and Carbone 2001). Recent work in a tumour model has established that in-vivo correction of a known antigen presenting defect was sufficient to resurrect antigen presentation and result in elimination of the tumour (Alimonti, Zhang et al. 2000). Characterization of all the genes involved in the antigen presentation process would present an enormous potential to cure a broad spectrum of diseases through gene therapy. 2 1.2 Antigen presentation Most cells throughout the body present endogenous antigens for immuno-surveillance, while specialized antigen presenting cells (APC) (macrophages (M0) , B lymphocytes (B-cells) and dendritic cells (DC)) also process and present exogeous antigens. This exogenous antigen presentation utilizes a unique mechanism, and it results in the antigen-specific priming of naive T-cells (Heath and Carbone 2001). Currently, the full process involved in A P C antigen processing and T-cell priming remains unknown. Endogenous antigen presentation Endogenous antigen presentation involves the processing of cytosolic antigens, such as viral antigens in infected cells or self antigens, into peptides to mount on class I M H C molecules. Cytosolic antigen is degraded by the proteasome (Hirsch and Ploegh 2000; Thery and Amigorena 2001; Yewdell and Bennink 2001) and is transported into the endoplasmic reticulum (ER) by the transporter associated with antigen presentation (TAP), formed by the TAP1 (ABCB2) and TAP2 (ABCB3) subunits (Kelly, Powis et al. 1992), there to be further processed by the ER-associated aminopeptidase (ERAP) (Serwold, Gonzalez et al. 2002). The resulting peptides can then be mounted on nascent class I M H C molecules with the help of accessory molecules tapasin (Ortmann, Copeman et al. 1997), calnexin (Saito, lhara et al. 1999), calreticulin (Sadasivan, Lehner et al. 1996) and ERp57 (Hughes and Cresswell 1998) as shown in Figure 1.1. The complete peptide-MHC complex is then moved to the cell surface to be presented to CD8+ cytolytic T-cells (CTL) for immuno-surveillance. This endogenous pathway occurs in 3 essentially all nucleated cells in the body, and its primary function is to present cellular antigens for immuno-surveillance by T-cells. ERp57 Nucleotide- Peptide- Pore domain Pore domain Peptide- Nucleotide-bmding binding hmdmg binding dorr.ain domain domain domain Figure 1.1. The M H C class I peptide-loading complex. ERp57, calreticulin and tapasin are all involved in facilitating loading of TAP-transported, class I restricted, E R A P -trimmed peptides onto the M H C heavy chain / p2-m complex. Possible modes of interaction are indicated (Ackerman and Cresswell 2004). Reproduced from Nature Immunology. 4 Exogenous antigen presentation In contrast, exogenous antigens are endocytosed by APCs, where they are processed in one of several ways (Moore, Carbone et al. 1988; Trowsdale, Hanson et al. 1990; Kovacsovics-Bankowski and Rock 1995). Exogenous antigen presentation requires extracellular antigens and peptides to pass from phagosomes or endosomes into endolysosomes or M H C class II compartments (MHC) (Peters, Neefjes et al. 1991; Amigorena, Drake et al. 1994) for exogenous presentation on class II M H C or cross-presentation on class I M H C (Bevan 1987; Bachmann, Lutz et al. 1996; den Haan, Lehar et al. 2000). In the MIIC, class II M H C restricted antigens undergo cathepsin cleavage to form peptides (Kleijmeer, Ossevoort et al. 1995; Pierre and Mellman 1998; Trombetta, Ebersold et al. 2003; Watts 2004). These peptides can then be loaded onto post-golgi class II M H C molecules for transport to the surface for presentation to CD4+ helper T-cells. Cross-presentation Cross-presentation is the process of presenting exogenous antigens on class I M H C molecules, previously thought to bind only endogenous antigens. Antigen presentation by dendritic cells (DCs) in the form of cross-presentation is pivotal to the initiation of T-cell mediated immunity and in particular, the priming of naive T-cells (Banchereau and Steinman 1998; Mellman and Steinman 2001). Uptake and presentation of exogenous antigens by DCs is integral to the production of an effective adaptive immune response against viruses, bacteria, tumour antigens, as well as for the generation of tolerance to self proteins (von Boehmer and 5 Hafen 1986; Matzinger and Guerder 1989; Germain 1994; Kurts, Heath et al. 1996; Kurts, Carbone et al. 1997; Steinman, Turley et al. 2000; Steinman and Nussenzweig 2002). Evidence has supported three principal, non-mutually exclusive models explaining how DCs cross-present exogenous antigens on class I M H C molecules (Heath and Carbone 2001)a,b: (1) TAP-dependent ER-loaded peptides (conventional), (2) phagosome-ER fusion loading (involving cathepsins, TAP, Sec61, and proteosomes), and (3) TAP-independent endolysosome or MIIC loaded peptides (via cathepsin degradation). Specific antigens are known to undergo processing via each of these pathways as shown in Figure 1.2a: 1. The TAP-dependent pathway (Moore, Carbone et al. 1988; Kovacsovics-Bankowski and Rock 1995) processes endocytosed antigens and exogenous proteins in the cytosol (Brossart and Bevan 1997). TAP-dependent cross-presentation involves exogenous antigen uptake whereby fluid antigens or antigens taken up by receptor-mediated endocytosis are endocytosed. These antigens are then transferred into the cytosol to be degraded into peptides by the proteasome. The peptides are then transported by TAP into the ER, similarly to the well-defined endogenous antigen presentation pathway. Alternatively, at this point the peptides may enter specialized antigen presenting compartments, such as the endolysosome or MIIC, where they can access post-golgi class 1 M H C molecules and then proceed to the cell surface. 6 2. Cross-presentation via the ER-phagolysosome has recently been demonstrated in DCs (Ackerman, Kyritsis et al. 2003; Guermonprez, Saveanu et al. 2003; Houde, Bertholet et al. 2003). The phagolysosome is a compartment capable of phagocytosing particulate antigens or dying cells, translocating those to the cytoplasm via Sec61 for degradation by the proteasome, and passing the generated peptides back into the phagosome through localized T A P transporters for access to class 1 M H C (Figure 1.2b). 3. A third pathway has been described as the TAP-independent pathway (Bachmann, Oxenius et al. 1995; L iu, Zhou et al. 1995; Gromme and Neefjes 2002). In this pathway, endocytosed exogenous proteins move into MI IC vesicles or endolysosomal compartments, where antigens are degraded by proteolytic cathepsins, similarly to the class II M H C restricted antigen presentation pathway (Amigorena, Drake et al. 1994; Mellman, Pierre et al. 1995). Peptides generated in this way are then able to access class I M H C molecules in these secretory vesicles, which have presumably been recycled from the cell surface or post-golgi membrane. Additional support for this pathway comes from recent work, showing that M H C I molecules lacking a sorting signal within their cytoplasmic domain renders them unable to access this endolysosomal compartment and are thus incapable of cross-presenting antigens (Lizee, Basha et al. 2003). 7 Antigen presentation to CTLs f MHC class I CO TAP J ~ ) Proteasome 00 MHC class II Ervdogertous pathway Viral proteins Exogenous pathway • Virus — Extracellular pathogen ( C P ) Phagosome x-.-'W m Antigen presentation j to helper T cells Endoplasmic reticulum Figure 1.2. ER-phagosome cross-presentation. Professional antigen presenting cells process intracellular and extracellular pathogens differently, a) In the endogenous pathway, proteins from intracellular pathogens, such as viruses, are degraded by the proteasome and the resulting peptides are shuttled into the endoplasmic reticulum by T A P proteins. These peptides are loaded onto M H C class I molecules and the complex is delivered to the cell surface, where it stimulates cytotoxic T-lymphocytes (CTLs) that k i l l the infected cells. In contrast, extracellular pathogens are engulfed by phagosomes (exogenous pathway) (Roy 2003). Reproduced from Nature, b) Inside the phagosome, the pathogen-derived peptides are either loaded directly onto M H C class II molecules, which activate helper T-cells that stimulate the production of antibodies, or degraded via the proteasome and cross-presented on M H C class I molecules (Desjardins 2003). Reproduced from Nature Immunology Reviews. Antigen presentation pathways for common antigens Different pathogens or antigens are processed and presented in different ways. Viral infections, such as vesicular stomatitis virus (VSV), are injected directly into the cell cytoplasm and are processed through the endogenous pathway (endogenous Class I). D C cross-presentation of these endogenous viral antigens from phagocytosed infected cells is required to prime the anti-viral immune response (Sigal, Crotty et al. 1999; Sigal and Rock 2000). Chicken egg ovalbumin (OVA) is taken up exogenously by endosomes and cross-presented by DCs in a 9 TAP-dependent manner. The immunodominant H-2K b restricted peptide, OVA(257-264) is generated in the cytoplasm via proteasome-dependent processing (exogenous class I) (Moore, Carbone et al. 1988; Kovacsovics-Bankowski and Rock 1995). Heat-inactivated Sendai virus (Hl-Sendai) is an endocytosed antigen cross-presented in a TAP-independent manner. This antigen produces an H-2K b restricted immunodominant peptide, Sendai(324-332), which is processed through the endosome to endolysosome pathway (exogenous class 1) (Bachmann, Oxenius et al. 1995; Liu, Zhou et al. 1995; Gromme and Neefjes 2002). Hen egg lysozyme (HEL) is another exogenous protein, endocytosed and processed in the endosome to lysosome or MI IC pathway, producing an immunodominant class II M H C restricted peptide, H E L ( 7 4 _ 8 8 ) on the I-Ab class II molecule (exogenous class II). These pathways are represented graphically in Figure 1.3 below, reproduced from Nature Immunology Reviews. 10 MHC I MHC l-pefctKte MHO *4 Figure 1.3. Antigen processing and presentation pathways, (a) Endogenous antigens, such as viral antigens in infected cells, are presented on ER-localized class I M H C via the T A P transporter, (b) Exogenous 'TAP-independent' antigens are presented on class II or cross-presented on class I M H C molecules, (c) Exogenous 'TAP-dependent' antigens are cross-presented on class I M H C (Heath and Carbone 200l)a. Reproduced from Nature Immunology Reviews. 11 Cytokine production and activation of APC The cellular immune system, and in particular the APCs, act in response to stimulatory cytokines. Infection by different pathogens generates an immune response, which results in production of different cytokines within the body. Interferon gamma (IFNy) and tumour necrosis factor alpha (TNFa) are generally produced in response to various D N A and RNA-based viral infections (Wong and Goeddel 1986). Both of these cytokines have been shown capable of activating antigen presenting cells, and DCs in particular (Brossart and Bevan 1997). Upon activation, the B7 family (CD80, CD86) of co-stimulatory 'danger' signals are upregulated, along with CD40 and DEC-205 in DCs, and antigen presentation is maximized through a marked increase in both class I and class II M H C expression at the cell surface, as shown in Figure 1.4. It is in this state, through the combined expression of MHC-bound antigen and co-stimulatory signals, that APCs are able to prime an antigen-specific T-cell immune response. 12 Immature DC Intermediate DC L$* J3S0 S i. ft 1/ 1 \" I ' CD11c Mature DC i l •V 1 , l-Ad (MHCII) CD40 C D 8 0 CD86 i -(N/A) Figure 1.4. Dendritic cell activation markers. Fluorescent activated cell sorting (FACS) analysis of D C (CD1 lc) cells, demonstrating the upregulation of D C cell surface markers as the cells progress from immature DCs (CD40-, CD80-, CD86-, MHC - I I lo), through intermediate DCs (CD40 lo, CD80 lo, CD86 lo, MHC - I I mid), and into mature 'activated' DCs (CD40 hi, CD80 hi, CD86 hi, MHC - II hi). Shaded histograms show negative staining controls, thin black outlines show stained cells (Calder, Liversidge et al. 2004). Reproduced from Journal of Immunological Methods. 13 1.3. ATP-binding cassette transporters The ATP-binding cassette (ABC) transporter family of proteins is the largest known group of transmembrane (TM) proteins. Wel l represented in organisms ranging from bacteria to humans, the predicted numbers of A B C transporters range from 29 in yeast (Michaelis and Berkower 1995; Decottignies and Goffeau 1997) to as many as 114 in the plant Arabidopsis (Dean, 2002), with humans and mice numbering in the middle, with an estimated 48-51 each (Dean, Rzhetsky et al. 2001; Dean 2002). Structurally, functional A B C proteins are comprised of 2 T M regions, each several (6-7) membrane-spanning alpha-helices long, which determine the substrate specificity; T M domains are paired together with 2 cytosolic nucleotide binding domains (NBD), which bind A T P and power the transport (Higgins 1992; Childs and Ling 1994; Dean and Allikmets 1995; Dean 2002). A B C transporters generally take 1 of 2 forms: a ' f u l l ' transporter, in which 1 gene encodes both T M and N B D paired regions, or ' ha l f transporters, coding for 1 N B D and 1 TM . Half-transporters must pair up to form homo- or hetero-dimers to function. In prokaryotes, A B C s are generally embedded in the cellular membrane and function to transport metabolic substrates into the cell from the surrounding environment. Most of the known functions of A B C transporters in eukaryotes are to shuttle compounds throughout the cell or from inside the cell out to the extracelluar environment for metabolic, protective or immunological purposes (Dean 2002). A B C transporter proteins have been localized to many different subcellular compartments within cells from phylogenetically diverse groups. The physiological functions for most of the 14 48-51 transporters found in mammals remain unknown, with the exception of a few, including the peptide transporters TAP1 and TAP2. Recent research has shown a peptide-transport role for an A B C transporter in an organelle other than the ER. In yeast, the A B C transporter M d l l is located in mitochondria and has been shown to transport self peptides from the mitochondria into the cytosol (Young, Leonhard et al. 2001). A B C transporters are involved in processes as diverse as lipid and sterol transport, drug efflux, eye colour pigment movement, and peptide or chemical transport (Dean 2002). The downstream results of these processes become very apparent once A B C function becomes disrupted. Such human diseases as cystic fibrosis, cholesterol defects, anemia, and multidrug resistance in cancerous tumours are all attributable to abnormal A B C transporter expression. 1.4 Rationale and hypotheses Rationale The goals of this dissertation were to further clarify the cell biological mechanisms by which exogenous antigen presentation occurs, by identifying candidate antigen presenting genes and characterizing those for their role in this process. I have selected one particular family of genes to evaluate, the A B C transporters. There is precedence for the involvement of A B C transporters in antigen processing, in the well-characterized movement of cytosolic peptides through the TAP transporters. TAP has been shown to be crucial for endogenous antigen 15 movement from the cytosol into the ER, and its presence is required for peptides to access M H C class I molecules prior to surface expression (Moore, Carbone et al. 1988; Trowsdale, Hanson et al. 1990; Kovacsovics-Bankowski and Rock 1995). The existence of the M d l l mitochondrial peptide transporter in eukaryotic cells (Young, Leonhard et al. 2001) provides precedent for the existence of additional peptide transporters in organelles other than the ER. The different pathways of exogenous antigen presentation require antigenic movement through subcellular compartments (Heath and Carbone 2001). This suggests the need for additional transporter involvement. Since antigens and peptides must be transported between intracellular vesicles, and T A P and M d l l are known to transport endogenous peptides, the family of A B C transporters may contain other such peptide-transport proteins. O f the 48 putative murine A B C transporter genes, on a phylogenetic basis, ABCB9/TAPL (TAP-like) shows exceptional similarity to the two known T A P antigen-presenting transporters (Zhang, Zhang et al. 2000). Since conserved structure is often indicative of conserved function, this gene holds the most interest for a potential role in antigen presentation. There are however several notable differences between A B C B 9 and the T A P genes. A B C B 9 is known to be expressed in only a few tissues, and it localizes to the endosome and lysosome membranes (Zhang, Zhang et al. 2000), while T A P expression is ubiquitous. TAP1 and 2 are located genomically within the class 11 M H C region on chromosome 6p21 (human) and 17 (mouse), while A B C B 9 is located on 12q24, and 5F, respectively. While many immune response genes involved in antigen presentation are located within the M H C locus, there are exceptions. For example, the P2 -microglobul in ( P 2 - m ) subunit of the class I M H C complex is located on 15q21 (human) and 2 (mouse), which is outside of the M H C region. 16 Endogenous antigen presentation for immuno-surveillance does not utilize endosomal or lysosomal compartments, but exogenous antigen presentation for T-cell priming does. Combined with its similarity to TAP, the endosomal and lysosomal localization of A B C B 9 suggests a possible role in exogenous antigen presentation. The restriction of exogenous antigen presentation to APCs may also account for the reduced array of tissue expression of A B C B 9 compared to the known T A P genes. A knock-out mouse model of the A B C B 9 gene was made available by the laboratory of Dr. Victor Ling at the B C Cancer Research Centre (Canada). Hypotheses Hypotheses and predictions were as follows: 1. Since APCs process exogenous antigens through several intracellular compartments, and there is precedent for peptide transport by A B C transporters (TAP), this intracellular antigen movement wi l l be facilitated via localized A B C transporters. 2. Since resting APCs take up antigen from their surroundings, but do not process nor present it on the cell surface, and activated APCs primarily process and present antigens, activated APCs should express higher numbers of A B C transporters than non-activated APCs, and these should be involved specifically in antigen transport. 3. Since A B C B 9 bears significant similarity to the T A P transporter, and is located in those compartments in which exogenous antigens are processed (i.e. endosomes and 17 lysosomes) of activated dendritic cells, it may function in the transport and processing of exogenous antigens. 4. It is expected that DCs from an animal lacking A B C B 9 may be unable to process and present exogenous antigens, and therefore these animals may be unable to prime a cell-mediated immune response upon challenge with either M H C class I or class II restricted exogenous antigens. 1.5 Goals Expression of all mouse A B C transporters wi l l be evaluated in primary APCs derived from C57BL/6 wild-type mice. This expression profile wi l l be repeated following exposure to the A P C activating cytokines T N F a and IFNy, and to the exogenous antigens ovalbumin and HI Sendai virus. The profile of transporter expression in untreated APCs wi l l be compared with that of treated APCs, as well as to a non-antigen presenting cell fibroblast control. We expect the results to reveal a series of A B C transporters that are inducible in activated APCs. These genes w i l l be subjected to phylogenetic analysis, and the results w i l l be used to predict A B C transporters as potential candidates for a role in antigen presentation. The A B C B 9 m o u s e representing the knockout of the TAP- l ike A B C transporter gene wi l l be evaluated for an immunological phenotype. Lymphoid organs wi l l be analyzed for a differential cellular distribution and compared to wild-type mice. Immune function w i l l be evaluated in-vivo by challenge with both live and heat-killed virus, as well as for the ability to respond to exogenous antigens on class I and class II M H C . Finally, the ability to generate an 18 anti-tumour response wi l l be evaluated by challenge with M H C expressing and non-expressing tumour cells. 19 1.6 Bibliography Ackerman, A. L. and P. Cresswell (2004). \"Cellular mechanisms governing cross-presentation of exogenous antigens.\" Nat Immunol 5(7): 678-84. Ackerman, A. L., C. Kyritsis, et al. (2003). \"Early phagosomes in dendritic cells form a cellular compartment sufficient for cross presentation of exogenous antigens.\" Proc Natl Acad Sci U S A 100(22): 12889-94. Alimonti, J . , Q. J . Zhang, et al. (2000). \" TAP expression provides a general method for improving the recognition of malignant cells in vivo.\" Nat Biotechnol 18(5): 515-20. Amigorena, S., J. R. Drake, et al. (1994). \"Transient accumulation of new class II M H C molecules in a novel endocytic compartment in B lymphocytes.\" Nature 369(6476): 113-20. Bachmann, M. F., M. B. Lutz, et al. (1996). \"Dendritic cells process exogenous viral proteins and virus-like particles for class I presentation to CD8+ cytotoxic T lymphocytes.\" Eur J Immunol 26(11): 2595-600. Bachmann, M. F., A. Oxenius, et al. (1995). \"TAP 1-independent loading of class I molecules by exogenous viral proteins.\" Eur J Immunol 25(6): 1739-43. Banchereau, J . and R. M . Steinman (1998). \"Dendritic cells and the control of immunity.\" Nature 392(6673): 245-52. Bevan, M . J. (1987). \"Antigen recognition. Class discrimination in the world of immunology [news].\" Nature 325(6101): 192-4. Brossart, P. and M . J. Bevan (1997). \"Presentation of exogenous protein antigens on major histocompatibility complex class I molecules by dendritic cells: pathway of presentation and regulation by cytokines.\" Blood 90(4): 1594-9. Calder, C. J . , J . Liversidge, et al. (2004). \"Murine respiratory tract dendritic cells: isolation, phenotyping and functional studies.\" J Immunol Methods 287(1-2): 67-77. Childs, S. and V. Ling (1994). \"The M D R superfamily of genes and its biological implications.\" Important Adv Oncol: 21-36. Dean, M . (2002). The Human ATP-Binding Cassette (ABC) Transporter Superfamily. Bethesda, MD, National Library of Medicine. Dean, M. and R. Allikmets (1995). \"Evolution of ATP-binding cassette transporter genes.\" Curr Opin Genet Dev 5(6): 779-85. Dean, M., A. Rzhetsky, et al. (2001). \"The human ATP-binding cassette (ABC) transporter superfamily.\" Genome Res 11.(7): 1156-66. Decottignies, A. and A. Goffeau (1997). \"Complete inventory of the yeast A B C proteins.\" Nat Genet 15(2): 137-45. den Haan, J . M., S. M. Lehar, et al. (2000). \"CD8(+) but not CD8(-) dendritic cells cross-prime cytotoxic T cells in vivo.\" J Exp Med 192(12): 1685-96. Desjardins, M. (2003). \"ER-mediated phagocytosis: a new membrane for new functions.\" Nat Rev Immunol 3(4): 280-91. Ehrlich, R. (1997). \"Modulation of antigen processing and presentation by persistent virus infections and in tumors.\" Hum Immunol 54(2): 104-16. Gabathuler, R., J. Alimonti, et al. (1998). \"Surrogate antigen processing mediated by TAP -dependent antigenic peptide secretion.\" j Cel l B io l 140(1): 17-27. 20 Germain, R. N. (1994). \"MHC-dependent antigen processing and peptide presentation: providing ligands for T lymphocyte activation.\" Cell 76(2): 287-99. Gromme, M. and J. Neefjes (2002). \"Antigen degradation or presentation by M H C class I molecules via classical and non-classical pathways.\" Mol Immunol 39(3-4): 181-202. Guermonprez, P., L. Saveanu, et al. (2003). \"ER-phagosome fusion defines an MHC class I cross-presentation compartment in dendritic cells.\" Nature 425(6956): 397-402. Heath, W. R. and F. R. Carbone (2001). \"Cross-presentation in viral immunity and self-tolerance.\" I^RexImmunoi 1(2): 126-34. Heath, W. R. and F. R. Carbone (2001). \"Cross-presentation, dendritic cells, tolerance and immunity.\" Annu Rev Immunol 19: 47-64. Higgins, C. F. (1992). \"ABC transporters: from microorganisms to man.\" Annu Rev Cell Biol 8: 67-113. Hirsch, C. and H. L. Ploegh (2000). \"Intracellular targeting of the proteasome.\" Trends Cell Biol 10(7): 268-72. Houde, M. , S. Bertholet, et al. (2003). \"Phagosomes are competent organelles for antigen cross-presentation.\" Nature 425(6956): 402-6. Hughes, E. A. and P. Cresswell (1998). \"The thiol oxidoreductase ERp57 is a component of the MHC class I peptide-loading complex.\" Curr Biol 8(12): 709-12. Kelly, A., S. H. Powis, et al. (1992). \"Assembly and function of the two A B C transporter proteins encoded in the human major histocompatibility complex.\" Nature 355(6361): 641-4. Kleijmeer, M. J., M. A. Ossevoort, et al. (1995). \"MHC class II compartments and the kinetics of antigen presentation in activated mouse spleen dendritic cells.\" J Immunol 154(11): 5715-24. Kovacsovics-Bankowski, M . and K. L. Rock (1995). \"A phagosome-to-cytosol pathway for exogenous antigens presented on MHC class I molecules.\" Science 267(5195): 243-6. Kruh, G. D. and M. G. Belinsky (2003). \"The MRP family of drug efflux pumps.\" Oncogene 22(47): 7537-52. Kurts, C , F. R. Carbone, et al. (1997). \"CD4+ T cell help impairs CD8+ T cell deletion induced by cross-presentation of self-antigens and favors autoimmunity.\" J Exp Med 186(12): 2057-62. Kurts, C , W. R. Heath, et al. (1996). \"Constitutive class I-restricted exogenous presentation of self antigens in vivo.\" J Exp Med 184(3): 923-30. Liu, T., X. Zhou, etal. (1995). \"Heat-inactivated Sendai virus can enter multiple M H C class I processing pathways and generate cytotoxic T lymphocyte responses in vivo.\" J Immunol 154(7): 3147-55. Lizee, G., G. Basha, et al. (2003). \"Control of dendritic cell cross-presentation by the major histocompatibility complex class I cytoplasmic domain.\" Nat Immunol 4(11): 1065-73. Matzinger, P. and S. Guerder (1989). \"Does T-cell tolerance require a dedicated antigen-presenting cell?\" Nature 338(6210): 74-6. Mellman, I., P. Pierre, et al. (1995). \"Lonely MHC molecules seeking immunogenic peptides for meaningful relationships.\" Curr Opin Cell Biol 7(4): 564-72. Mellman, I. and R. M. Steinman (2001). \"Dendritic cells: specialized and regulated antigen processing machines.\" Cell 106(3): 255-8. Michaelis, S. and C. Berkower (1995). \"Sequence comparison of yeast ATP-binding cassette proteins.\" Cold Spring Harb Svmp Quant Biol 60: 291-307. 21 Moore, M. W., F. R. Carbone, et al. (1988). \"Introduction of soluble protein into the class I pathway of antigen processing and presentation.\" Cel l 54(6): 777-85. Ortmann, B., J. Copeman, et al. (1997). \" A critical role for tapasin in the assembly and function of multimeric M H C class I-TAP complexes.\" Science 277(5330): 1306-9. Peters, P. J., J. J. Neefjes, et al. (1991). \"Segregation of M H C class II molecules from M H C class I molecules in the Golgi complex for transport to lysosomal compartments.\" Nature 349(6311): 669-76. Pierre, P. and 1. Mellman (1998). \"Developmental regulation of invariant chain proteolysis controls M H C class 11 trafficking in mouse dendritic cells.\" Cel l 93(7): 1135-45. Roy, C. R. (2003). \"Immunology: professional secrets.\" Nature 425(6956): 351-2. Sadasivan, B., P. J. Lehner, et al. (1996). \"Roles for calreticulin and a novel glycoprotein, tapasin, in the interaction of M H C class I molecules with TAP.\" Immunity 5(2): 103-14. Saito, Y., Y. Ihara, et al. (1999). \"Calreticulin functions in vitro as a molecular chaperone for both glycosylated and non-glycosylated proteins.\" Embo J 18(23): 6718-29. Serwold, T., F. Gonzalez, et al. (2002). \" E R A A P customizes peptides for M H C class 1 molecules in the endoplasmic reticulum.\" Nature 419(6906): 480-3. Sigal, L., S. Crotty, et al. (1999). \"Cytotoxic T-cell immunity to virus-infected non-haematopoietic cells requires presentation of exogenous antigen.\" Nature 398(6722): 77-80. Sigal, L. J. and K. L. Rock (2000). \"Bone marrow-derived antigen-presenting cells are required for the generation of cytotoxic T lymphocyte responses to viruses and use transporter associated with antigen presentation (TAP)-dependent and -independent pathways of antigen presentation.\" J Exp Med 192(8): 1143-50. Steinman, R. M. and M. C. Nussenzweig (2002). \"Avoiding horror autotoxicus: the importance of dendritic cells in peripheral T cell tolerance.\" Proc Natl Acad Sci U S A 99(1): 351-8. Steinman, R. M., S. Turley, et al. (2000). \"The induction of tolerance by dendritic cells that have captured apoptotic cells.\" J Exp Med 191(3): 411-6. Thery, C. and S. Amigorena (2001). \"The cell biology of antigen presentation in dendritic cells.\" Curr Opin Immunol 13(1): 45-51. Tortorella, D., B. E. Gewurz, et al. (2000). \"Viral subversion of the immune system.\" Annu Rev Immunol 18: 861-926. Trombetta, E. S., M . Ebersold, et al. (2003). \"Activation of lysosomal function during dendritic cell maturation.\" Science 299(5611): 1400-3. Trowsdale, J., 1. Hanson, et al. (1990). \"Sequences encoded in the class II region of the M H C related to the ' A B C superfamily of transporters [see comments].\" Nature 348(6303): 741-4. von Boehmer, H. and K. Hafen (1986). \"Minor but not major histocompatibility antigens of thymus epithelium tolerize precursors of cytolytic T cells.\" Nature 320(6063): 626-8. Watts, C. (2004). \"The exogenous pathway for antigen presentation on major histocompatibility complex class II and CD1 molecules.\" Nat Immunol 5(7): 685-92. Wong, G. H. and D. V. Goeddel (1986). \"Tumour necrosis factors alpha and beta inhibit virus replication and synergize with interferons.\" Nature 323(6091): 819-22. Yewdell, J. W. and J. R. Bennink (2001). \"Cut and trim: generating M H C class I peptide ligands.\" Curr Opin Immunol 13(1): 13-8. Young, L., K. Leonhard, et al. (2001). \"Role of the A B C transporter M d l l in peptide export from mitochondria.\" Science 291(5511): 2135-8. 22 Zhang, F., W. Zhang, et al. (2000). \"Characterization of A B C B 9 , an A T P binding cassette protein associated with lysosomes.\" J B io l Chem 275(30): 23287-94. 23 C H A P T E R II E XPRESS ION PROF IL ING OF A B C T R A N S P O R T E R S IN APCs C H A P T E R II. EXPRESS ION PROF IL ING OF A B C TRANSPORTERS IN APCs 2.1 Overview Ubiquitious endogenous antigen presentation on class I major histocompatibility (MHC) molecules drives immuno-surveillance by CD8+ T-cells. The transporter associated with antigen presentation (TAP), comprised of 2 half A B C transporters, A B C B 2 (TAP1) and A B C B 3 (TAP2), transports peptides for endogenous antigen presentation. Specialized antigen presenting cells (APCs) conduct both endogenous and exogenous antigen presentation, which is crucial for priming the immune response. Exogenous antigen presentation requires uptake and processing of extracellular antigens through subcellular compartments for presentation on M H C molecules. In this study, we examined gene expression of 46 murine A B C s in resting and stimulated APCs, compared with non-APC controls. Genes that were inducible in APCs by immune cytokines were also found to be induced by exposure to exogenous antigen and were identified as potential antigen transporters. These results were combined with a phylogenetic analysis of these 46 ABCs , and they revealed clustering of many of our candidate antigen transporters including TAP1 and TAP2. In this manner, 18 A B C genes were identified for their potential involvement in antigen presentation. 25 2.2 Introduction The ability of the adaptive immune system to mount a response to antigens requires peptide processing and presentation by antigen presenting cells (APCs) (Unanue 2002). Dendritic cells (DC), macrophages (M0) , and B-cells are all known to act as specialized APCs. APCs are known for their ability to process and present exogenous antigens, both on class I and on class II M H C , as required for priming a cellular immune response to viral, bacterial, or tumour antigens, as well as for generation of tolerance to self proteins. Dendritic cells in particular have been shown to function exceptionally in this role (Mellman and Steinman 2001). Endogenous antigen presentation is constitutive in most cells, and the mechanisms of this pathway are well known (Monaco, Cho et al. 1990; Trowsdale, Hanson et al. 1990; Powis, Townsend et al. 1991; Sadasivan, Cariappa et al. 1995; Hughes and Cresswell 1998). Endogenous antigens are cleaved into peptides by the proteasome before being transported through the TAP transporter into the E R to bind to nascent class 1 M H C en route to the cell surface. In less well-defined pathways, exogenous antigens are taken up by APCs and processed through various intracellular compartments to be presented on MHC-II or cross-presented on MHC- I (Moore, Carbone et al. 1988; Trowsdale, Hanson et al. 1990; Kovacsovics-Bankowski and Rock 1995). In the cellular immune system, APCs are activated by exposure to cytokines, or specific receptor-binding by pathogens. IFNy is produced in response to a variety of viral infections, while tumour necrosis factor alpha (TNFa) is commonly released in response to viral or bacterial infection and/or inflammation (Holler 2002). Both of these cytokines have been shown to be 26 capable of activating APCs, and dendritic cells in particular (Brossart and Bevan 1997; Gallucci and Matzinger 2001). The purpose of this investigation was to determine whether additional A B C transporters other than TAPs might be candidates for involvement in antigen processing and presentation. To address this question, differential gene expression of 46 identified A B C transporters was evaluated in DCs, M 0 s and B-cells, and compared to non-APC controls. Each subset of APC s was exposed to stimulatory immune cytokines, or to various antigens, and the differential A B C gene expression was evaluated. The results were used to determine that specialized APCs express differential A B C transporters compared to non-APCs, that these transporters are cytokine-inducible, and that specific antigens induce these same A B C transporters, suggesting that exposure to antigen alone is capable of inducing A B C transporter activity. Several of those transporters induced in APCs share sequence similarity, and are phylogenetically clustered into groups with the known T A P antigen presenting genes. It was also revealed that DCs express specific A B C transporters that differ from those expressed in other APCs, suggesting genes that may be specific to cross-presentation. 2.3 Materials and Methods Media 27 A l l assays were conducted in C T L media: RPM I 1640 + 1% antibiotics (penicillin/streptomycin) + 1% N E A A + 1% L-glutamine + 1% HEPES + 0.1% 2-Mercaptoethanol + 10% Hyclone (UT) fetal bovine serum (FBS). Cell purification A l l cells were obtained from C57BL/6 mice. Dendritic cells, macrophages and B-cells were purified from mouse spleen using positive selection for C D l l c + , C D l l b + (following C D l l c + depletion), and CD19+ antibody-bound microbeads (Miltenyi Biotec, Canada) respectively, as per manufacturer's directions on an Au toMACS (Miltenyi Biotec, Canada) cell separator. Briefly, spleens were injected with lmg/mL Collagenase D (Boehringer-Mannheim, NJ) in PBS, disaggregated, and incubated for 1 hr at 37 °C. Splenocytes were then passed through 70 pm nylon mesh and washed with PBS supplemented with 0.5% FBS (buffer). The cells were resuspended in 400 pL buffer per two spleens, and 100 pL microbeads was added. After 15 minutes incubation at 4 °C, cells were washed, resuspended in 500uL buffer and separated on Au toMACS by double positive selection (POSSELD). Antigenic induction Cells were cultured overnight in 6-well plates in 5 m L of C T L media. Cells were stimulated with 10 ng/mL TNFa ( R & D Systems, MN ) to induce general inflammatory immune response genes. To activate the endosome to lysosome exogenous M H C Class 1 pathway of antigen presentation, heat-inactivated Sendai (Hl-Sendai) virus (65 degrees C , 2 hours) was added to the media prior to overnight incubation at 37 °C. Chicken egg ovalbumin (Worthington, NJ) was added (2 mg/mL) to the media to stimulate the endosome to cytosol 28 exogenous class 1 pathway. Hen egg lysosome (HEL) (Sigma-aldrich, Canada) was added (2 mg/mL) to stimulate the exogenous class II antigen presentation pathway. To simulate viral infection initiation of endogenous antigen presentation, 50 ng/mL IFNy (R&D Systems, M N ) was added to the cells prior to incubation. An untreated control was included for all cell types. Bio informatics Phylogenetic analysis was conducted using ClustalX software, version 1.81 (National Center for Biotechnology Information). Sequences for alignment were obtained based upon the GenBank accession numbers published on http://www.humanabc.org, hosted by the Nutrition, Metabolism and Genomics Group, Wageningen University, the Netherlands. Phylograms were obtained by ClustalX multiple amino acid sequence alignment of all available sequences for human and mouse, followed by creation of a tree using neighbour-joining (N-J) and bootstrap algorithms. Sequence specific RT -PCR primers (shown in Table 2.1) were selected corresponding to each of the murine A B C transporter expressed sequences recorded in N C B I GenBank database (accession numbers published at http://www.humanabc.org). Primer selection was conducted using the web-based Primer3 software and checked against a rodent mispriming library (Rozen 1996,1997). RT-PCR R N A was extracted using the Qiagen (Canada) RNEasy kit. Concentration of R N A was determined by spectrophotometric quantification, and equal amounts (1.0 ug) of R N A from each cell type treatment were used for each reverse-transcription (RT) reaction. Complementary D N A 29 (cDNA) was produced from the R N A using random hexanucleotide primers and Superscript!! RT enzyme (Gibco-BRL, Canada) as per manufacturers' directions. Equal amounts of c D N A were then used as template for each PCR reaction. Expression of A B C genes was evaluated by RT-PCR with Taq polymerase (Gibco-BRL, Canada) using the primers indicated in Table 2.1 for 35 PCR cycles, annealing temperature of 60° C with 1 minute each denaturing, annealing and elongation. Equal volumes of the PCR product were run on a 1% agarose gel alongside a 100 bp D N A ladder (Gibco-BRL, Canada). 2.4. Results Interferon-y (IFNy) is produced in response to immune stimulation by several stimuli, including viral infections and results in a primarily Th-1 type T-cell response. Many genes involved in the immune response are regulated by this cytokine, and are considered to be interferon responsive (IRE) (York, Goldberg et al. 1999). TAP1 and TAP2 have been shown to be upregulated in response to interferon, and other immune stimulatory cytokines such as T N F a (Figure 2.1). 30 o o Q Q TAP1 TAP2 Figure 2.1. Cytokine treatment resulted in increased expression of the transporters involved in antigen presentation, TAP1 (ABCB2) and TAP2 (ABCB3). C D l l c + dendritic cells were purified from C57BL/6 mouse spleens (>98% pure) and incubated at 37° overnight in the absence or presence of the maturation stimulus TNFa to induce DC maturation. Total R N A was extracted and gene expression was evaluated using RT-PCR. Many other genes located in the MHC-locus (chromosome 6p21 in humans and 17,4 in mice) are involved in antigen presentation and are IFN-responsive, such as the immunoproteasome subunits LMP2 and 7, and tapasin (York, Goldberg et al. 1999). Other genes outside of this M H C locus are involved in the immune response and also show this IFN-responsiveness, such as P 2 - m or ER-proteases. Initially, we wanted to see whether A B C 31 transporters were induced in APCs by pulsing with IFNy or TNFa. Having chosen gene-specific primers for all 46 available mouse A B C transporter sequences (shown in Table 2.1), we evaluated gene expression in APCs both untreated and treated with cytokine. We observed several genes to be upregulated, and these provided a starting point from which to examine potential antigen presentation function (see RT-PCR example in Figure 2.2 and full results in Table 2.2). O T ~ T - o i n ^ - N c o i - ^ o o T - r - T -0 < < < < < < m m o o o a c o . o m - o O O O O O O O O O O O O - a ^ Q Q D Q f j Q f j Q f j Q m f j Q D Q Q Q D Q f X l C Q \" D i 5 < < < < < < < < < < < < i 5 cs iLOQOT-c\\ ic \\ ir^-cr) O O O Q Q i i l i O O O O O O O O O O O O O O O O O ^ O O O O O o T - - t f T - CO O Q UJ Q OO CM CO co a> 00 CO CO 3 exon4 J 3' 5' - D - T Z Exon I II IV V VI VII VIII IX X XI XII KO region Mus muscuius chromosome 5 66 Figure 3.1. A B C B 9 gene knockout construct and expression profile. RT-PCR of A B C B 9 expression in C57BL/6 mouse (a) tissues or (b) purified bone marrow derived (bm)DCs in an immature state, and matured with TNFa, including TAP1 and TAP2 expression levels, (c) Diagram of the A B C B 9 genomic D N A knockout region. To assess the role of A B C B 9 in immune function, we derived the A B C B 9 \" mouse (summarized in Figure 3.1c) and observed that this mouse does not express A B C B 9 m R N A (Figure 3.2a). The A B C B 9 ~~ mice are normal in appearance, fertility, and growth rate compared to wild-type mice. To determine whether A B C B 9 affected leukocyte development, we examined leukocyte sub-populations in lymphoid tissues using flow cytometry. The thymus and lymph nodes of the A B C B 9 ~~ mice show a sharp reduction in total cellularity (-50%). Examination of the total numbers of different cell types in these organs revealed half as many B- and T-cells as wild-type (Figure 3.2b,c). The total numbers of DCs and M 0 in these organs were normal, however, as were the splenocytes, with the exception of slightly elevated numbers of B-cells (Figure 3.2b,c). This decrease in L N B- and T-cells, accompanied by a decrease in numbers of thymic M H C - I and MHC- I1 expressing cells, clearly suggests a role for A B C B 9 in T-cell development. 67 Figure 3.2. Cellular profiles of A B C B 9 \" mice show decreased B-cells and T-cells in lymph node and thymus, and a fewer M H C class 1+ or 11+ thymocytes, (a) RT -PCR of A B C B 9 expression in wild-type and A B C B 9 ~~ mouse brain, (b-c) Total organ cellularity of various cellular subtypes in (b) spleen and thymus and (c) lymph nodes as measured by flow cytometry, multiplied by total cell counts. The profiles depicted represent mean +/-SE of 3 mice. We investigated whether the loss of A B C B 9 resulted in a phenotype similar to the TAP1\" \" mouse (van Kaer, Ashton-Rickardt et al. 1992), with no CD8+ T-cells nor K b expression, and defective endogenous antigen presentation. In response to infection with vesicular stomatitus virus (VSV), A B C B 9 m i c e produced normal numbers of total K b/VSV-N ( 5 2 -59) (K b /VSV) specific CTLs, as determined directly by staining with K b /VSV tetramers (Figure 3.3a,b). A B C B 9 7 \" mice were also able to generate a functional V S V C T L response, measured by 5 1 C r -release assay (Figure 3.3c). In contrast to the TAP1\" 7\" mouse, A B C B 9 7 \" mice were fully capable of priming a T-cell response by presenting peptides from phagoctyosed infected cells. A B C B 9 therefore does not appear to function analogously to the T A P molecule because a live virus infection results in a robust C T L response. 69 25.0 wt 69 nai.'.:;;wt..'B9 nai - - D a y 0--— -Day 4~- -100 75 at c • J 50 25 } ^ A B C B 9 - / - ( - p e p ) j wt(\"pep) : j wt (+pep) j ABCB9/-{+pep)[ \"iObr-i \"• \"33:1 • 11;1 EffectorTarget Ratio 70 Figure 3.3. ABCB9\" mice generate a normal CTL response to live virus, (a) Flow cytometric tetramer analysis of splenocytes from mice infected 7 days with VSV. Percent Kb/VSV-N(5 2-59) specific CTLs from total splenocytes on (top) day 0 and (bottom) day 4 ex-vivo. (b) Quantity of Kb/VSV-N(5 2-59) specific CTLs from total wild-type (wt), ABCB97\" (B9), or naive (nai) splenocytes, N=3 mice, (c) 51Cr-release CTL assay of VSV primed effector splenocytes against targets pulsed + or - VSV peptide. To further evaluate the role of ABCB9 in viral cross-presentation, we injected mice with heat-inactivated (HI) Sendai virus, an exogenous antigen cross-presented on K b molecules in a 'TAP-independent' manner. The immunodominant peptide NP(324-332) is processed via the endocytic pathway and loaded onto K b molecules in endolysosomal compartments. We found that ABCB9\"~ mice were unable to mount an effective CTL response as measured by 5lCr-release assay. CTL function appears to be reduced approximately 10-fold, requiring lOx more CTL to achieve the same amount of killing as wild-type cells (Figure 3.4). As DCs are the most relevant APC involved in cross-presentation (Banchereau and Steinman 1998; Mellman and Steinman 2001), we asked whether the ABCB9\"7\" mouse CTL deficiency to Hl-Sendai was due to a specific defect in DC cross-presentation and priming of T-cells. We pulsed DCs with Hl-Sendai and injected these into syngeneic mice to prime a DC-specific CTL response (Inaba, Metlay et al. 1990; Brossart and Bevan 1997). Our results show that although antigen-pulsed DCs were able to more efficiently prime a CTL response than DCs stimulated by intra-peritoneal (i.p.) injection of antigen alone, the overall CTL response in the ABCB9\"7\" mice was still reduced 10-fold 71 compared to their wild-type counterparts (Figure 3.4b). This supports the hypothesis that loss of AJ3CB9 results in a defect in cross-presentation of Hl-Sendai antigen. a 30 b . -°-ABCB9-/-(-pep) ; -*-v*(-pep) : ~*-wt(+pep) -o-ABCB9-/-(+pep) 200:1 66:1 Effector Target Ratio 22:1 200:1 66:1 Effector.Target Ratio 22:1 Figure 3.4. ABCB9\"\" mice generate a deficient C T L response to killed virus. 5 1 C r -release C T L assay against Hl-Sendai, primed by (a) direct i.p. Hl-Sendai injection, or (b) i.p. injection of syngeneic DCs pulsed with Hl-Sendai. Mean+/-SE, triplicate samples. Similar results were obtained in three different experiments. This TAP-independent pathway of antigen presentation, typified by Hl-Sendai antigen, is known to be chloroquine sensitive, brefeldin-A resistant, and T A P independent (Liu, Zhou et al. 7 2 1995; L iu, Chambers et al. 1997). It is generally assumed that peptides from these endocytosed antigens are generated within the endolysosome by acidification and cathepsin activity. We show however that these TAP-independent antigens access the cytosol to undergo proteasome degradation into peptides. Treating DCs with lactacystin, a proteasome inhibitor, while pulsing with Hl-Sendai, abrogated C T L priming ability (Figure 3.5). 200:1 66:1 EffectorTarget ratio 22:1 •Lactacystin and HI Sendai (targets w/ peptide) •Lactacystin and HI Sendai (targets w/o peptide) •Lactacystin and peptide (targets w/ peptide) - Lactacystin and peptide (targets w/o peptide) • HI Sendai (targets w/ peptide) • HI Sendai (targets w/o peptide) Figure 3.5. HI Sendai antigen processing is proteasome-dependent. 5 1Cr-release C T L assay against HI Sendai in wild-type mice. DCs were purified from C57BL/6 mouse spleens and pulsed +/- the proteasome inhibitor lactacystin prior to incubation with HI Sendai antigen, or Sendai(324-332) peptide. Effector CTLs were primed in-vivo by i.p. injection of these +/- lactacystin, +/- HI Sendai/peptide pulsed DCs into syngeneic 73 animals on day 0 and 7. On day 14, spleens were collected from primed animals and restimulated in-vitro with 1 u M Sendai(324-332) peptide. Splenocytes were used as effectors in a standard 5 1Cr-release assay against chromium-labelled RMA-S target cells. Fil led shapes indicate target cells pulsed with Sendai(324-332) peptide, open shapes indicate target cells with irrelevant peptide bound (negative controls). Results represent mean of triplicate samples. In order to assess whether loss of A B C B 9 also affects presentation of exogenous non-viral antigens, we examined the C T L response to ovalbumin (OVA). DCs were pulsed with soluble O V A and injected into syngeneic mice. To determine whether cross-presentation of O V A protein in ABCB9\" 7 \" mice affected K b/OVA ( 2 57-264) (rC b/OVA) peptide-specific C T L production in-vivo, we measured the generation of splenic CTLs directly with K b /OVA tetramer (Figure 3.6a). A B C B 9 7 \" mice on average produced 10-fold fewer K b /OVA CTLs than wild-type animals (Figure 3.6b). K b /OVA C T L function was further examined by 5 1Cr-release assay. Similar to the response to Hl-Sendai, ABCB9\" 7 \" splenocytes were lOx less effective at ki l l ing K b /OVA target cells (Figure 3.6c). 74 a naive wild type (nai) DayO H < 8 Dav 4 a . • O o 0.12% 10\" 10 • I04 10° l(T 1 • 0.08% wild type (wt) 0.05%) *- 'ft''\"\"\"' 1 \" ' ' \" ' V ' ' ' 1 ' \" \" ' a 10U I01 1VN ER-phagosome <*~ TAP\" ER y MHC-I a ^ / ^ ^ P r o t e a s o m e ABCB9 V ^ _ ^ Endosomes \\ Exogenous antigens (ovalbumin, HI Sendai virus) Figure 3.10. Proposed models of A B C B 9 as an endosomal or endolysosomal antigen transporter in cross-presentation, (a) Exogenous antigens are taken up through the endosomal pathway, processed through A B C B 9 into the cytosol to be degraded by the proteasome. Alternatively, (b) following proteasome degradation, antigens from both the phagocytic and endocytic pathways pass through the substrate specific A B C B 9 or T A P into the E R or ER-phagosome / endolysosome to access M H C - I in this antigen presenting compartment. 83 Barrier-protected organs such as brain and testis are considered to be immune-privileged sites. The cross-presentation of self antigens by DCs has been shown to be the main mechanism of induction of tolerance to self (Bachmann, Lutz et al. 1996; Steinman, Turley et al. 2000; Heath and Carbone 2001; Steinman and Nussenzweig 2002). The high levels of A B C B 9 in brain microglia (unpublished observation) and testis Sertoli cells (Zhang, Zhang et al. 2000) may provide a high constitutive level of exogenous self-antigens in order to promote tolerance in these immunologically sensitive sites. Our data suggests a possible role for A B C B 9 in thymic T-cell selection and peripheral self-tolerance, in addition to immune priming. A role for A B C B 9 in exogenous antigen movement through the endolysosome may begin to reveal the molecular processes involved in this crucial immunological event. 84 3.6 Bibliography Ackerman, A. L., C. Kyritsis, et al. (2003). \"Early phagosomes in dendritic cells form a cellular compartment sufficient for cross presentation of exogenous antigens.\" Proc Natl Acad Sci U S A 100(22): 12889-94. Amigorena, S., J. R. Drake, et al. (1994). \"Transient accumulation of new class II M H C molecules in a novel endocytic compartment in B lymphocytes.\" Nature 369(6476): 113-20. Bachmann, M. F., M . B. Lutz, et al. (1996). \"Dendritic cells process exogenous viral proteins and virus-like particles for class I presentation to CD8+ cytotoxic T lymphocytes.\" Eur J Immunol 26(11): 2595-600. Bachmann, M. F., A. Oxenius, et al. (1995). \"TAP 1-independent loading of class I molecules by exogenous viral proteins.\" Eur J Immunol 25(6): 1739-43. Banchereau, J. and R. M . Steinman (1998). \"Dendritic cells and the control of immunity.\" Nature 392(6673): 245-52. Bevan, M. J. (1987). \"Antigen recognition. Class discrimination in the world of immunology rnewsl.\" Nature 325(6101): 192-4. Brossart, P. and M . J. Bevan (1997). \"Presentation of exogenous protein antigens on major histocompatibility complex class I molecules by dendritic cells: pathway of presentation and regulation by cytokines.\" Blood 90(4): 1594-9. Gromme, M . and J. Neefjes (2002). \"Antigen degradation or presentation by M H C class I molecules via classical and non-classical pathways.\" Mo l Immunol 39(3-4): 181-202. Guermonprez, P., L. Saveanu, et al. (2003). \"ER-phagosome fusion defines an M H C class I cross-presentation compartment in dendritic cells.\" Nature 425(6956): 397-402. Heath, W. R. and F. R. Carbone (2001). \"Cross-presentation in viral immunity and self-tolerance.\" Nat Rev Immunol 1(2): 126-34. Houde, M., S. Bertholet, et al. (2003). \"Phagosomes are competent organelles for antigen cross-presentation.\" Nature 425(6956): 402-6. Inaba, K., J. P. Metlay, et al. (1990). \"Dendritic cells pulsed with protein antigens in vitro can prime antigen-specific, MHC-restricted T cells in situ.\" J Exp Med 172(2): 631-40. Kelly, A., S. H. Powis, et al. (1992). \"Assembly and function of the two A B C transporter proteins encoded in the human major histocompatibility complex.\" Nature 355(6361): 641-4. Kovacsovics-Bankowski, M . and K. L. Rock (1995). \" A phagosome-to-cytosol pathway for exogenous antigens presented on M H C class 1 molecules.\" Science 267(5195): 243-6. Liu, T., B. Chambers, et al. (1997). \" TAP peptide transporter-independent presentation of heat-killed Sendai virus antigen on M H C class I molecules by splenic antigen-presenting cells.\" J Immunol 159(11): 5364-71. Liu, T., X. Zhou, et al. (1995). \"Heat-inactivated Sendai virus can enter multiple M H C class I processing pathways and generate cytotoxic T lymphocyte responses in vivo.\" J Immunol 154(7): 3147-55. Lizee, G., G. Basha, et al. (2003). \"Control of dendritic cell cross-presentation by the major histocompatibility complex class I cytoplasmic domain.\" Nat Immunol 4(11): 1065-73. 85 Ljunggren, H. G., S. Paabo, et al. (1989). \"Molecular analysis of H-2-deficient lymphoma lines. Distinct defects in biosynthesis and association of M H C class I heavy chains and beta 2-microglobulin observed in cells with increased sensitivity to N K cell lysis.\" J Immunol 142(8): 2911-7. Mellman, I., P. Pierre, et al. (1995). \"Lonely M H C molecules seeking immunogenic peptides for meaningful relationships.\" Curr Opin Cell B io l 7(4): 564-72. Mellman, 1. and R. M . Steinman (2001). \"Dendritic cells: specialized and regulated antigen processing machines.\" Ce l l 106(3): 255-8. Moore, M . W., F. R. Carbone, et al. (1988). \"Introduction of soluble protein into the class I pathway of antigen processing and presentation.\" Cel l 54(6): 777-85. Porgador, A., J. W. Yewdell, et al. (1997). \"Localization, quantitation, and in situ detection of specific peptide-MHC class I complexes using a monoclonal antibody.\" Immunity 6(6): 715-26. Schmitt, L. and R. Tampe (2002). \"Structure and mechanism of A B C transporters.\" Curr Opin Struct B io l 12(6): 754-60. Shastri, N., G. Gammon, et al. (1986). \"Ia molecule-associated selectivity in T cell recognition of a 23-amino-acid peptide of lysozyme.\" J Exp Med 164(3): 882-96. Shastri, N. and F. Gonzalez (1993). \"Endogenous generation and presentation of the ovalbumin peptide/Kb complex to T cells.\" J Immunol 150(7): 2724-36. Sigal, L., S. Crotty, et al. (1999). \"Cytotoxic T-cell immunity to virus-infected non-haematopoietic cells requires presentation of exogenous antigen.\" Nature 398(6722): 77-80. Steinman, R. M . and M. C. Nussenzweig (2002). \"Avoiding horror autotoxicus: the importance of dendritic cells in peripheral T cell tolerance.\" Proc Natl Acad Sci U S A 99(1): 351-8. Steinman, R. M., S. Turley, et al. (2000). \"The induction of tolerance by dendritic cells that have captured apoptotic cells.\" J Exp Med 191(3): 411-6. Valdez, Y., W. Mah, et al. (2002). \"Major histocompatibility complex class II presentation of cell-associated antigen is mediated by CD8alpha+ dendritic cells in vivo.\" J Exp Med 195(6): 683-94. van Kaer, L., P. G. Ashton-Rickardt, et al. (1992). \"TAP1 mutant mice are deficient in antigen presentation, surface class I molecules, and CD4-8 + T cells.\" Cel l 71: 1205. Yewdell, J. W. and J. R. Bennink (2001). \"Cut and trim: generating M H C class I peptide ligands.\" Curr Opin Immunol 13(1): 13-8. Zhang, F., W. Zhang, et al. (2000). \"Characterization of A B C B 9 , an A T P binding cassette protein associated with lysosomes.\" J B io l Chem 275(30): 23287-94. 86 C H A P T E R IV E X O G E N O U S CROSS-PR IM ING A N D T U M O U R I M M U N O L O G Y C H A P T E R IV. E X O G E N O U S CROSS-PR IMING A N D T U M O U R I M M U N O L O G Y 4.1 Overview Cancer arises in the body when genetic mutations accumulate and allow cells to proliferate in an unchecked manner. The cellular immune response regularly surveys for such cells by testing for tumour associated antigens (TAA) bound to class I M H C molecules on the cell surface. When such a cancerous cell is found by a dendritic cell (DC), it w i l l be processed by the D C in order to cross-prime a T-cell response. Primed cytotoxic T-lymphocytes (CTLs) w i l l then proliferate and circulate through the body sampling for these same MHC- I bound TAAs. Upon encountering the tumour, i f they display their TAA s on the surface, CTLs can recognize and destroy the cancerous cells. Normally this process is very efficient, but tumours may arise whereby their antigen presentation is abrogated. The transporter associated with antigen presentation (TAP) has been shown to be integral to this process. Here we investigate the role of another TAP- l ike protein (ABCB9) in tumour immunology. A B C B 9 ~~ mice are unable to mount an effective immune response to exogenous cross-presented antigens. We wanted to evaluate whether exogenous cross-presentation plays a major role in the anti-tumour immune response. We injected A B C B 9 A mice with the B16 melanoma, CMT.64 or CMT.1-4 small cell lung carcinoma and followed tumour progression and animal survival. Further, we immunized mice with CMT.64 or CMT.1-4 prior to injecting them with live tumour cells. Our results show no significant effect of A B C B 9 on survivorship of mice injected with antigen-expressing tumours, whether immunized or not. ABCB9~'~ mice do show a slight increase in survival and reduction of tumour growth when injected with non-antigen presenting tumour cells, 88 and this trend is reversed i f they are pre-immunized. This suggests a possible supportive, although not primary role for A B C B 9 and exogenous antigen cross-presentation in tumour immunology and a possible function in induction of tolerance. 4.2 Introduction Cancerous cells arise spontaneously in the body in response to any number of external or internal stimuli. Cancer-inducing agents are often mutagenic agents and include such omnipresent environmental factors as ultraviolet radiation from the sun's rays, cigarette smoke breathed in the air, and naturally occurring or man-made radioactive materials that emit alpha, beta or gamma irradiation. A cancer is essentially a cell that proliferates unchecked and does not die, or one in which the rate of proliferation is greater than that of elimination (Laiho and Latonen 2003). Any number of genetic mutations can contribute to this, and indeed it generally requires several such mutations so that the cell 's natural cycle checkpoints can be overridden (Wright, Egan et al. 1990; Laiho and Latonen 2003; Sarasin 2003; Malaguarnera 2004). This requirement for an accumulation of mutations explains why cancers tend to be more prevalent as people age, after increased years of exposure to environmental factors (Sarasin 2003). Cancers also appear in clusters of individuals who are exposed to large amounts of mutagens, such as the incidence of mouth cancer in early watch dial painters, who used radium-infused paint to illuminate the numbers on watch faces, and habitually licked the brush tip to a point (McGrayne 1996). 89 Cancerous cells arise frequently, but are eliminated by the cellular immune system, which has evolved a very intricate recognition mechanism to eliminate diseased cells. Expression of tumour-associated antigens (TAA) on cellular class I M H C molecules allows the body to recognize them as dangerous and destroy them with cytotoxic T-lymphoctyes (CTLs). Generating an immune response to tumours requires two steps of antigen presentation. First, the antigen presenting cells (APCs), particularly dendritic cells (DCs), must encounter the tumour cell and determine that it is abnormal. Once that occurs the cell itself is phagocytosed and the T A A s are processed and cross-presented on the D C class I MHC . The DCs must then migrate to the lymphoid organs (spleen, lymph nodes) to encounter naive T-cells and prime them to activate, proliferate and destroy the tumour target cells. Secondly, for the purposes of immunosurveillance, the tumour cells themselves must express the TAAs on their own surface MHC- I molecules, so that activated T-cells can identify and destroy them. A major method of tumour immune evasion involves the elimination of T A A s on MHC- I of tumour cells. This can be accomplished by mutation or down-regulation of any number of genes and proteins involved in the antigen presentation pathway. Many tumours that do not express T A A s have been examined and found to be defective in their expression of antigen processing transporter TAP1 and/or TAP2, or the proteasome subunits LMP2 and/or LMP7 (Tanaka, Isselbacher et al. 1985; Singal, Ye et al. 1996; Garrido, Ruiz-Cabello et al. 1997). It has been shown that re-introducing these genes by gene therapy is sufficient to partially restore T A A and MHC-I surface expression and allows the cells to be recognized and killed as C T L targets (Tanaka, Isselbacher et al. 1985; Wallich, Bulbuc et al. 1985; Garrido, Ruiz-Cabello et al. 1997; Seliger, Maeurer et al. 1997; Alimonti, Zhang et al. 2000). 90 Small-cell lung carcinomas (SCLC) are highly malignant in both humans and mice and are generally fatal. The CMT.64 cell line is a murine S C L C that arose spontaneously in a C57BL/6 mouse (Franks, Carbonell et al. 1976) and shows extremely reduced endogenous MHC-I restricted T A A expression at the cell surface (Jefferies, Kolaitis et al. 1993; Mandelboim, Bar-Haim et al. 1997). CMT.64 cells are defective in their expression of proteasome subunits L M P 2 and LMP7, and transporter associated with antigen presentation (TAP) subunits TAP1 and TAP2 (Gabathuler, Reid et al. 1994). However, antigen presentation can be rescued in these cells by the addition of TAP1 alone (Gabathuler, Reid et al. 1994; Alimonti, Zhang et al. 2000). This demonstrates that even in tumours where many components are disrupted, it is possible to reimplement immunosurveillance by introducing only one integral gene. Having established that A B C B 9 plays a role in cross-priming, and that A B C B 9 7 \" mice cannot cross-prime exogenous antigens effectively, we wanted to investigate whether exogenous cross-priming has any effect on cancer progression in-vivo. This was done using the A B C B 9 ~~ mouse model to evaluate tumour growth and disease progression. As A B C B 9 is normally expressed in activated DCs, expression of A B C B 9 in the tumours themselves would not be expected to affect immune response. However, a lack of A B C B 9 in APCs may create a more permissive environment for tumour growth in-vivo by decreasing or eliminating the activation of anti-tumour T-cells by DCs. Several H-2K b restricted murine tumour cell lines are available for this purpose, such as the melanoma B16/F10 (B16) (Fidler, Gersten et al. 1976) and small-cell lung carcinoma CMT.64 and CMT.1-4 (CMT.64 transfected with TAP1, expressing surface H-2K b molecules) (Alimonti, Zhang et al. 2000). It has been established (Alimonti, Zhang et al. 2000; Thompson, 91 Timme et al. 2000) that certain H-2K b expressing tumour cell lines can be destroyed by an anti-tumour cellular immune response, i f animals are immunized first with irradiated tumour cells. Our laboratory has previously established that expressing T A P in tumour cells that have down regulated antigen presentation machinery is sufficient to re-implement partial H-2K b surface expression and incite an anti-tumour immune response (Gabathuler, Reid et al. 1994; Al imonti, Zhang et al. 2000). To test this hypothesis, we separately injected each of the non-H-2K b expressing tumour cell lines above (B16/F10, CMT.64) and the C M T tumour cell line stably transfected with TAP1 (CMT.1-4) into wild-type (C57BL/6) and A B C B 9 7 \" mice deficient in exogenous antigen cross-priming. For both the H-2K b expressing (CMT.1-4) and non-H-2K b tumour cells, we injected live cells or injected irradiated tumour cells followed by injection of live tumour cells, to determine if there was any differential anti-tumour response between our A B C B 9 ~~ mice and wild-type controls, and whether this was influenced by prior immunization with the tumour antigen. 4.3 Materials & Methods Models Mouse models C57B1/6 and A B C B 9 7 (on C57BL/6 background) mice (both H-2K b haplotype). 92 Tumour cell lines: B16, a non-K b expressing H-2K b haplotype murine melanoma; CMT.64, a non-K b expressing, H-2K b haplotype murine small-cell lung carcinoma; CMT.1-4 (CMT.64 stably transfected with TAP1 to reconstitute partial K b expression). Tumour injections For B16 cells, l .SxlO 3 were injected sub-cutaneously (s.c.) in 100 uL of PBS with a 25G needle on the right hind leg. B16 were separately injected s.c. on matched cohorts of mice from the wild-type and A B C B 9 7 \" groups. Tumour growth was monitored until any single tumour was estimated at ~ 1 0 % body weight, at which point all animals were sacrificed, the tumours excised and weighed. For C M T cells, 3.5xl0 5 were injected intra-peritoneally (i.p.) in 500 uL of PBS with a 25G needle. CMT.64 or CMT.1-4 (an adenocarcinoma) was separately injected intraperitoneally (i.p.) into wild-type and A B C B 9 7 \" groups of mice. To immunize animals, C M T cells were irradiated with 10 4 rads before injecting 3.5xl0 5 cells per mouse i.p.. 14 days later the animals were challenged with 3.5x105 live cells per mouse i.p.. The animals were monitored for signs of morbidity, at which time each individual morbid animal was euthanized, and a survivorship record was kept in graphical form. Statistics A l l experimental results were evaluated for statistical significance by the paired, 2-tailed T-test. 93 4.4 Results Very little is known about the specific details of antigen processing pathways for initiation of an anti-tumour response, other than that D C cross-priming appears to be involved. We were interested to evaluate whether exogenous cross-presentation was the primary mechanism of anti-tumour T-cell cross-priming. Since the A B C B 9 7 \" mouse model is deficient in exogenous cross-presentation (Chapter III), it provides a method to evaluate the involvement of exogenous cross-priming in tumour responses in-vivo. We challenged these animals by injecting them with tumour cells of a shared haplotype background (B6). Normally tumours such as B16 and CMT.64 that do not present M H C antigens succeed in proliferating and metastasizing and result in mortality of the mice by evading CTL-mediated immunosurveillance and kil l ing (Franks, Carbonell et al. 1976; Gabathuler, Alimonti et al. 1998; Alimonti, Zhang et al. 2000). These non-MHC expressing tumour cells can be targeted for destruction by natural killer (NK) cells (Trinchieri 1989; Papamichail, Perez et al. 2004). As attested to by the prevalence of aggressive cancers however, the N K response alone is often not sufficient to eliminate the disease. Those tumours that do express M H C antigens however, are capable of being destroyed in an immuno-competent mouse. If exogenous cross-presentation is involved in anti-tumour cross-priming, it would be expected that those tumours that express little or no M H C (and therefore do not present antigens at the cell surface) w i l l grow similarly in both mouse genotypes. It would also be expected that those tumours presenting antigens on the cell surface, such as CMT.1-4 should show a differential growth rate between the two animal groups, with the A B C B 9 7 \" mice being more susceptible to tumour progression due to a lack of immune response to TAAs . As the type of tumour and route of injection can result in a differential induction of the 94 immune response, we chose two tumour types to evaluate initially: an adenocarcinoma small-cell lung cancer, CMT.64 to be injected intraperitoneally (i.p.) and B16, a melanoma to be injected sub-cutaneously (s.c). Our experimental results from the B16 tumour cell injections show that wild-type and A B C B 9 ~~ mice do not show statistically significant differences in growth of these non-H-2K b expressing tumours, as shown in Figure 4.1 below. While there was a trend towards smaller tumours in the A B C B 9 7 \" mice, the size differences were not statistically significant (p>0.25). 95 9& 1.860g V V 0.369g 0.166g 0.587g C57BI/6 wild-type 1.171g 1 028g 9 0.886g 0.208g A B C B 9 - / -0.213g 2.5 B16 Tumour progression 2.0 § 1.5 | 1.0 3 0.5 0.0 O O O O O o o o Wild-type ABCB9'-Figure 4.1. B16 tumour progression. Wild-type and A B C B 9 \" mice do not show differential growth of non-H-2K b expressing B16 tumours. 4 x l 0 5 B16 tumour cells were injected sub-cutaneously on day 0 and excised on day 13. (a) B16 tumours excised from mice on day 13 post-injection, with corresponding tumour weights, (b) Graphical plot of tumour sizes for each group of mice, each circle represents one mouse (N = 5). Statistically not significant (T-test, p>0.25). With no apparent effect of the loss of A B C B 9 \" \" on B16 tumour growth, mice were injected i.p. with CMT.64 S C L C and monitored for survivorship for 90 days (Figure 4.2). Initially, the A B C B 9 ~~ mice were seen to survive on average approximately 5 days longer than the wild-type, with all wild-type mice dying by day 35 post-injection. 4 0 % (2 mice) from the A B C B 9 A group survived until day 60, and 2 0 % (1 mouse) of wild-type survived throughout the 90-day study period. Although this difference was considered to be statistically significant (p<0.05) based upon a paired T-test, due to the small sample size, no conclusion can be drawn until larger populations are evaluated. The time until 5 0 % mortality (TD50) was reached for wild-type and ABCB9~~ mice were 25 and 31 days respectively. Because the sample size was small (N-5), the differences observed may not be truly representative of an A B C B 9 ~~ population differential survival rate. 97 CMT.64 survivorship 100 0 10 20 30 40 50 60 70 Days post tumour injection • Wild-type ABCB9-/-80 90 Days survival post CMT.64 injection Wild-type ABCB9^ 98 Figure 4.2. Survivorship of CMT.64 injected mice. 3.5xl0 5 CMT.64 cells (non- H-2K b expressing, transfected with empty vector) were injected i.p. for each mouse, (a) Survivorship curve, (b) Chart marking days survival for each mouse per group, each circle represents one mouse (N = 5). Statistically significant (T-test, p<0.05). Mice were injected i.p. with the K b expressing CMT.1-4 (CMT.64+TAP) and monitored for survivorship for 90 days (Figure 4.3). Both wild-type and A B C B 9 ~~ mice showed a similar survival rate, with T D 5 0 being 68 and 70 days respectively. This was significantly longer than for mice injected with CMT.64 tumours, with TD50 at 25 and 31 days respectively for wild-type and ABCB9\" 7 \" mice (Figure 4.2). 99 CMT1-4 survivorship 100 80 en | 60 » 40 •Wild-type • ABCB9-/-20 T 1 1 r n , ! 1 1 1 ! 1 1 1 1 1 |T\" 0 10 20 30 40 50 60 70 Days post tumour injection 80 90 Days survival post CMT.1-4 injection 90 f 80 70 O ro 60 1 50 (0 & 4 0 n Q * 30 20 10 0 o o Wild-type ABCB9\" / _ 100 Figure 4.3. Survivorship of CMT.1-4 injected mice. 3.5xl0 5 CMT.1-4 cells (transfected with TAP1 to restore H-2K b expression) were injected i.p. for each mouse, (a) Survivorship curve, (b) Chart marking days survival for each mouse per group, each circle represents one mouse (N = 5). Statistically not significant (T-test, p>0.05). Mice were then injected with irradiated C M T cells (either CMT.64 or CMT.1-4) and given 14 days to allow them to generate a robust cellular immune response. On day 14 following immunization, the animals were injected with live C M T cells corresponding to those with which they had been originally immunized. In the CMT.64 immunized mice, both A B C B 9 / _ and wi ld-type mice showed an increased survival compared to non-immunized CMT.64 challenge (Figure 4.4). This indicates that even the reduced M H C on the cell surface was sufficient to induce a cellular immune response. In the first 40 days, there was an increase in the morbidity of the ABCB9\" 7 \" mice compared with wild-type, however beyond 40 days, both groups showed a similar response with 70-80% long-term survival. 101 Immunized CMT.64 tumour progression re > 3 CO 100 80 60 40 20 0 i ' \" 1 r — — i 1 -| 1 0 10 20 30 40 50 60 70 Days post live tumour injection •Wild-type CMT.64 AE3CB9 CMT.64 80 90 Days survival post live CMT.64 injection 90 80 70 75 60 -I ,> | 50 vt % 40 re Q * 30 10 0 r o o - o • o Wild-type ABCB97\" 102 Figure 4.4. Survivorship of pre-immunized CMT.64 injected.mice. Mice were injected i.p. with 3.5xl0 5 irradiated CMT.64 cells 14 days prior to challenge by i.p. injection of 3.5x105 live CMT.64 cells, (a) Survivorship curve, (b) Chart marking days survival for each mouse per group, each circle represents one mouse (N - 10). Statistically not significant (T-test, p>0.05). For both A B C B 9 \" \" and wild-type mice, when animals were pre-immunized, survival was also increased remarkably when challenged with the CMT.1-4 cells, as 90-100% of both groups survived the entire study period (Figure 4.5), compared to only 4 0 % survival in non-immunized CMT.1-4 injected mice from both groups (Figure 4.3a). This demonstrates the potential of the cellular immune response to eliminate M H C expressing tumour cells. Since the A B C B 9 7 \" mice generated a similar anti-tumour response compared to wild-type mice, this demonstrates that exogenous antigen presentation does not make a major contribution to this type of anti-tumour immunity. 103 100 80 5 60 CO 40 20 Immunized CMT.1-4 tumour progression •Wild-type CMT.1-4 • ABCB9 CMT.1-4 0 10 20 30 40 50 60 70 80 90 Days post live tumour injection Days survival post live CMT.1-4 injection 90 80 70 75 60 > § 50 w & 4 0 re Q * 30 20 10 0 Wild-type ABCB9\"'\" 104 Figure 4.5. Survivorship of pre-immunized CMT.1-4 injected mice. Mice were injected i.p. with 3.5xl0 5 irradiated CMT.1-4 cells 14 days prior to challenge by i.p. injection of 3.5xl0 5 live CMT.1-4 cells, (a) Survivorship curve, (b) Chart marking days survival for each mouse per group, each circle represents one mouse (N - 10). Statistically not significant (T-test, p>0.05). 4.5 Discussion Wild-type and ABCB9~'~ mice that were injected with the live B16 tumour cells showed statistically similar tumour growth, albeit with A B C B 9 ~\" showing a tendency toward smaller tumours. Wild-type mice injected with C M T tumour cells showed the expected survival curve, with CMT.1-4 (+TAP) resulting in a much longer survival (Figure 4.2, 4.3), as shown previously (Alimonti, Zhang et al. 2000). The ABCB9\"~ mice did not show an increased susceptibility to tumour proliferation after injection with CMT.64 cells, and surprisingly a sub-group of ABCB9~'~ mice even showed increased longevity compared to wild-type mice (Figure 4.2). In the initial stages of the CMT.64 tumours, the survival of the knockout mice and wild-type animals differed slightly, with A B C B 9 m i c e surviving 5 days longer to the TD 5o point. In the period following 35 days post tumour injection, there were two A B C B 9 m i c e that survived to day 60, and one to the end of the study at 90 days, while none of the wild-type mice survived past 35 days. While this may be a true representation of the population, the sensitivity of the assay must be 105 considered. For this size of group (N=5), having one or two mice survive could merely represent difference in the original injection, or simply animal to animal variation. In the immunized mice injected with the H-2K b expressing CMT.1-4 (CMT.64+TAP), the survivorship of both wild-type and A B C B 9 ' A mice was the same (Figure 4.5). This indicates that exogenous antigen cross-priming has little or no effect on the progression of MHC-I antigen expressing tumour cells in the body. Those immunized mice injected with CMT.64 however, showed an initial difference between the survivorship of ABCB9~~ animals and wild-type, although later in the study, the susceptibility became equal between the two groups (Figure 4.4). In the early stages, more knockout mice succumbed to the tumours than wild-type. By day 30 post tumour challenge, two out of ten (20%) of the A B C B 9 7 \" mice had succumbed, while all wild-type mice survived until day 40, at which point one animal of each type succumbed. When comparing non-immunized to immunized mice, immunized mice show increased survival in all cases, indicating the initiation of a cellular immune response (Figures 4.2-4.5). The immunization response to CMT.1-4, which expresses M H C plus bound antigens at the cell surface, provided almost complete protection, with 9 0 % and 100% of wild-type and A B C B 9 ~\" mice, respectively, surviving beyond the study period (Figure 4.5). In comparison, the immunization response against CMT.64 was also protective, although to a lesser degree (Figure 4.4), as would be expected based upon the significantly reduced M H C surface expression compared to CMT.1-4 (Jefferies, Kolaitis et al. 1993; Alimonti, Zhang et al. 2000). This shows that the cellular immune response is important even in those cancers with reduced M H C surface expression. 106 4.6 Conclusions Results from these experiments should be interpreted with caution, since the numbers of animals tested in each case was relatively small (N=5 or 10). Should these results reflect the typical response to tumour challenge in wild-type and A B C B 9 ~~ mice, it appears that exogenous antigen presentation is not the primary method by which a cellular anti-tumour response is primed in-vivo, as assessed by the A B C B 9 / _ mouse model. Animals deficient in exogenous antigen presenation, but with intact phagocytic and endogenous antigen presentation can mount a normal anti-tumour response to s.c. B16 melanoma, or to i.p. injected CMT-64 or CMT.1-4 (CMT.64 + TAP) SCLC. Pre-immunization with irradiated tumour cells resulted in a similarly increased survival in A B C B 9 ~~, as compared to wild-type mice. One notable difference was observed in non-immunized mice challenged with CMT.64. In this group, two of five A B C B 9 ~~ mice survived longer than their wild-type counterparts. This result is the opposite of what would be expected i f A B C B 9 played a role in priming the immuological T-cell response against tumours, suggesting a possible role for A B C B 9 in the generation of immune tolerance. 4.7 Future directions Further experiments should be conducted to determine whether this data is significant. If significant, then a potential explanation for the results observed here would be i f A B C B 9 also plays a role in immune tolerance. T-cell priming by DCs is a double-edged sword. On one hand, MHC-restricted antigen presentation is required to prime a C T L response to dangerous antigens. 107 On the other hand, the same MHC-restricted antigen presentation of self proteins is required to initiate tolerogenic T-cells into the anergic T-cell response. The mechanism by which the D C 'decides' to prime a C T L or tolerogenic response is still very much a mystery, although clues are beginning to emerge (Liu, lyoda et al. 2002). It has recently been shown that DCs purified from inflamed brain and central nervous system were incapable of priming a C T L response, and were only able to promote tolerance (Suter, Biollaz et al. 2003). As A B C B 9 A is highly expressed in the brain, a specific role in tolerance to self tissues should be considered. As DCs encounter tumour cells that do not present surface antigens, they may not be recognized as dangerous. Instead, a tolerogenic response to these cells could be produced, whereby CTLs specific for these antigens are destroyed. Such a role in tolerance would account for an increase in survival of non-immunized A B C B 9 ~~ mice compared with wild-type when challenged with CMT.64 tumour cells. It is also predictable that this response is not observed in immunized animals as the irradiated CMT.64 cells should provide sufficient danger signals through their apoptosis to override tolerance and promote a phagocytic-primed C T L response. Future investigation of T IL cytokines pruoduced in A B C B 9 ~~ mice should focus upon whether these animals do indeed produce fewer tolerogenic signals, such as IL-10 and tumour growth factor-P (TGFP), as well as evaluation of their levels of the Th-1 stimulating cytokines IL-2, IL-12 and IFNy, or Th-2 stimulating cytokine IL-4. 108 4.8 Bibliography Alimonti, J., Q. J. Zhang, et al. (2000). \" TAP expression provides a general method for improving the recognition of malignant cells in vivo.\" Nat Biotechnol 18(5): 515-20. Fidler, I. J., D. M. Gersten, et al. (1976). \"Characterization in vivo and in vitro of tumor cells selected for resistance to syngeneic lymphocyte-mediated cytotoxicity.\" Cancer Res 36(9 pt.l): 3160-5. Franks, L. M., A. W. Carbonell, et al. (1976). \"Metastasizing tumors from serum-supplemented and serum-free cell lines from a C57BL mouse lung tumor.\" Cancer Res 36(3): 1049-55. Gabathuler, R., J. Alimonti, et al. (1998). \"Surrogate antigen processing mediated by TAP -dependent antigenic peptide secretion.\" J Cel l B io l 140(1): 17-27. Gabathuler, R., G. Reid, et al. (1994). \"Comparison of cell lines deficient in antigen presentation reveals a functional role for TAP-1 alone in antigen processing.\" J Exp Med 180(4): 1415-25. Garrido, F., F. Ruiz-Cabello, et al. (1997). \"Implications for immunosurveillance of altered H L A class I phenotypes in human tumours.\" Immunol Today 18(2): 89-95. Jefferies, W. A., G. Kolaitis, et al. (1993). \"IFN-gamma-induced recognition of the antigen-processing variant CMT.64 by cytolytic T cells can be replaced by sequential addition of beta 2 microglobulin and antigenic peptides.\" J Immunol 151(6): 2974-85. Laiho, M. and L. Latonen (2003). \"Cel l cycle control, D N A damage checkpoints and cancer.\" Ann Med 35(6): 391-7. L iu, K., T. lyoda, et al. (2002). \"Immune tolerance after delivery of dying cells to dendritic cells in situ.\" J Exp Med 196(8): 1091-7. Malaguarnera, L. (2004). \"Implications of apoptosis regulators in tumorigenesis.\" Cancer Metastasis Rev 23(3-4): 367-87. Mandelboim, O., E. Bar-Haim, et al. (1997). \"Identification of shared tumor-associated antigen peptides between two spontaneous lung carcinomas.\" J Immunol 159(12): 6030-6. McGrayne, S. B. (1996). Nobel prize women in science: Their lives, struggles and momentous discoveries. New York, Carol Publishing Group. Papamichail, M., S. A. Perez, et al. (2004). \"Natural killer lymphocytes: biology, development, and function.\" Cancer Immunol lmmunother 53(3): 176-86. Sarasin, A. (2003). \"An overview of the mechanisms of mutagenesis and carcinogenesis.\" Mutat Res 544(2-3): 99-106. Seliger, B., M. J. Maeurer, et al. (1997). \" TAP off-tumors on.\" Immunol Today 18(6): 292-9. Singal, D. P., M. Ye, et al. (1996). \"Molecular basis for lack of expression of H L A class I antigens in human small-cell lung carcinoma cell lines.\" Int J Cancer 68(5): 629-36. Suter, T., G. Biollaz, et al. (2003). \"The brain as an immune privileged site: dendritic cells of the central nervous system inhibit T cell activation.\" Eur J Immunol 33(11): 2998-3006. Tanaka, K., K. J. Isselbacher, et al. (1985). \"Reversal of oncogenesis by the expression of a major histocompatibility complex class I gene.\" Science 228(4695): 26-30. Thompson, T. C , T. L. Timme, et al. (2000). \"Mouse prostate reconstitution model system: A series of in vivo and in vitro models for benign and malignant prostatic disease.\" Prostate 43(4): 248-54. Trinchieri, G. (1989). \"Biology of natural killer cells.\" Adv Immunol 47: 187-376. 109 Wallich, R., N. Bulbuc, et al. (1985). \"Abrogation of metastatic properties of tumour cells by de novo expression of H-2K antigens following H-2 gene transfection.\" Nature 315(6017): 301-5. Wright, J. A., S. E. Egan, et al. (1990). \"Genetic regulation of metastatic progression.\" Anticancer Res 10(5A): 1247-55. 110 C H A P T E R V A V I EW TO T H E F U T U R E C H A P T E R V. A V I EW TO T H E F U T U R E 5.1 In review With the initial aims of this thesis completed, it is time to evaluate what has been done, how this may impact the field, and where these discoveries may lead. The identification of several novel candidate antigen presenting genes should provide a starting place from which to further evaluate the mechanisms of antigen transport. The characterization of A B C B 9 as a transporter involved in dendritic cell cross-presentation should lead to further evaluation of its ability to modulate T-cell priming. O f particular interest, having defined a new step in the exogenous antigen cross-presentation pathway, this information has clearly delineated a difference between viral antigen cross-priming through the phagosome, and endocytic exogenous antigen cross-priming. The model shown in Figure 3.10 of Chapter III adds to the mechanistic model of antigen cross-presentation built over recent years (Brossart and Bevan 1997; Yewdell, Norbury et al. 1999; Sigal and Rock 2000; Heath and Carbone 2001; Yewdell and Bennink 2001), and adds an additional level of refinement to the most recent models put forth elucidating the ER-phagolysosome (Ackerman, Kyritsis et al. 2003; Guermonprez, Saveanu et al. 2003; Houde, Bertholet et al. 2003; Lizee, Basha et al. 2003). In addition, the unexpected finding that HI Sendai antigen accesses the cytosolic antigen processing pathway redefines a previously poorly understood mechanism and provides biologically relevant information that can be used to illuminate the details of TAP-independent antigen cross-presentation (Liu, Chambers et al. 1997). Finally, having shown a limited effect of exogenous antigen cross-priming on cancer immunology, future immunotherapy can focus on cross-presentation of endogenous 112 phagocytosed antigens instead. However, the potential for A B C B 9 to act in tolerance as well as C T L priming should be evaluated for possible treatment of autoimmune disease. 5.2 A B C B 9 Having established A B C B 9 as a transporter involved in dendritic cell cross-presentation, there are several directions from which to pursue future experiments. With the demonstration that A B C B 9 ~~ mice cannot cross-present the exogenous antigen ovalbumin, the next step w i l l be to determine why ovalbumin processing is deficient. It is possible that A B C B 9 transports this protein directly, or indirectly by transporting some intermediate substance that is involved in antigen processing. It wi l l be very important to determine the precise substrate specificity of A B C B 9 , in particular, whether it transports ovalbumin protein as intact protein, partially degraded as large antigenic epitopes, or as discrete specific peptides, similarly to TAP. A cross-presentation peptide transport assay would be a useful method by which to determine this (Heemels, Schumacher et al. 1993; Schumacher, Kantesaria et al. 1994). Such an assay could also provide information as to whether loss of A B C B 9 alters the peptide repertoire. This type of assay would have to be conducted in dendritic cells, purified from wild-type and A B C B 9 ~\" mice. We have identified that A B C B 9 7 \" mice can respond normally to endogenous viral antigens cross-presented by DCs, presumably through phagocytosis of infected cells. A B C B 9 ~~ mice do not respond normally to exogenous antigens endocytosed and cross-presented by DCs. An assay should be conducted to determine i f A B C B 9 DCs are able to present ovalbumin that has been phagocytosed as a coating on latex beads (Ackerman, Kyritsis et al. 2003; 113 Guermonprez, Saveanu et al. 2003; Houde, Bertholet et al. 2003). This can be done by pulsing DCs in-vitro with latex beads coated in ovalbumin, and evaluating H2-K b /OVA peptide at the cell surface with B3Z T-cells (Shastri and Gonzalez 1993), measuring IL-2 production, similarly to Figure 3.8 in Chapter III. Alternatively, DCs can be ' fed ' cells known to be phagocytosed (i.e. Listeria monocytogenes) (Paschen, Dittmar et al. 2000), or other irradiated non-APCs that have been transfected with ovalbumin. If H2-K b /OVA surface presentation is restored in A B C B 9 ~~ DCs following co-incubation with these OVA-transfected cells, it would show that phagocytic uptake of ovalbumin was sufficient to restore cross-presentation, contrasted with the endocytic antigen processing pathway. This w i l l allow for the differentiation between A B C B 9 as a transporter in the endosomal compartment (H2-K b /OVA presentation wi l l be increased), or as a substrate-specific transporter in the phagolysosome (H2-K b /OVA presentation wi l l not be increased). 5.3 Cross-presentation and cancer immunology For the cancer immunology studies, the differential tumour response based upon A B C B 9 \" / _ animal model indicates that further experiments should be conducted. These animals should be evaluated for presence of tumour infiltrating lymphocytes (TIL). These lymphocytes can be monitored for subtype (CD4+ versus CD8+) and production of immunological cytokines to determine whether loss of A B C B 9 7 \" results in release of regulatory or immunogenic cytokines. IL-2 and a high proportion of CD8+ T-cells would indicate a primarily Th-1 type C T L response. Conversely, predominantly IL-4 and CD4+ cells would indicate a Th-2 type helper T-cell 114 response. Production of cytokines such as IL-10 would indicate an immunosuppressive reaction, promoting immune tolerance (Alimonti, Zhang et al. 2000; Steinman, Hawiger et al. 2003). 5.4 New avenues for A B C B 9 Last but not least, the avenue of autoimmunity is well worth considering. Dendritic cells have lately been revealed as integral to generating tolerance by the mechanism of cross-presentation (Steinman and Nussenzweig 2002; Steinman, Hawiger et al. 2003), and as such, the possible effects of A B C B 9 on autoimmune disease should be fully evaluated. The role of A B C B 9 in antigen cross-presentation in DCs, and particularly the results of the tumour challenge experiments, suggests a possible role in creating tolerance and protecting against autoimmunity. There are readily available mouse models of autoimmune disease such as the non-obese diabetic (NOD) mouse, with an immune response against the pancreatic insulin-secreting islet cells (Gallegos and Bevan 2004). The A B C B 9 ~\" mouse should be crossed onto the N O D background mouse to determine whether the autoimmune diabetic pathology is accelerated or increased. Alternatively, young N O D mice could be irradiated and injected with syngeneic ex-vivo bone marrow that had been transduced to highly express A B C B 9 , to observe whether there was relief of the diabetic phenotype later in life. 5.5 Multiple RT-PCR screen for A B C transporter gene expression 115 The A B C transporter family-wide screen utilized in this thesis is a novel, although straightforward approach. Since the publication of the human (and mouse) genome, large-scale screening on microarrays has gained in popularity. These gene 'chips ' generally compare c D N A from several samples, and give a readout of increased or decreased gene expression for thousands of different genes (most still uncharacterized) (Goldsmith and Dhanasekaran 2004). While the semi-quantitative gene screen used here is based on a similar concept, it focuses on one specific gene family, and is limited to a manageable number of genes (N=46). Having identified several A B C transporter genes with potential antigen presenting functions, the next step wi l l be to characterize each one independently with regards to their effects on antigen presentation. Each one should be evaluated using loss-of function and gain-of function experiments. Since antigen presentation occurs in primary antigen presenting cells, the best model would be to work in-vivo, in mouse models which could be compared to previous work in this field, similarly to the TAP1 (van Kaer, Ashton-Rickardt et al. 1992) and A B C B 9 knockout mice (Chapter III). Any A B C s found to be involved in antigen presentation should also be examined for the potential for use in gene therapy for any variety of diseases that utilize cell-mediated immunity. This particular screening method of evaluating A B C expression could also be used as a tool to evaluate patterns of transporter expression in several other systems. In particular, it could be applied to evaluate A B C genes active in barrier-protected immune-privileged organs, such as the brain, testis, eye and at the fetal / maternal placental barrier. This should be useful in providing candidate transport proteins used to facilitate the movement of substrates across these otherwise impermeable barriers. There is precedent for the hypothesis that A B C transporters are 116 involved in these functions, as A B C B 1 (PGP/MDR1) was coincidentally found to play a role in blood-brain barrier biology in A B C B 1 7 \" mice (Schinkel, Smit et al. 1994; Schinkel, Mayer et al. 1997). The phenotype was discovered only when the mouse colony received an otherwise harmless spray of the insecticide vermectin, used to remove external parasites living on the mice. While normally harmless to vertebrates, the A B C B l \" \" mice died when subjected to this treatment, and the underlying cause was found to be passage through the otherwise impermeable blood-brain barrier, indicating an important role for A B C B 1 ~~ in regulation of components crossing into the brain. 5.6 Conclusion In conclusion, 1 feel that the goals of this thesis, namely identifying A B C transporters involved in exogenous antigen presentation, have been attained. Having characterized the first A B C protein (ABCB9 ) specifically involved in cross-presentation has been a major accomplishment in both the cell biology of antigen presentation as well as illumination of the mechanism of cross-priming in the immune response. Evaluation of the A B C B 9 \" \" mouse when challenged with various antigens has revealed differential phagocytic processing of cells infected with endogenous antigens versus direct processing of endocytosed exogenous antigens, which are specifically regulated by the A B C B 9 transporter protein. A tumour model has revealed that exogenous antigen presentation may play a supportive, although not principal role in tumour immunology. Having identified several additional (N=17) A B C transporters with potential functions in antigen presentation has opened the door to elucidating the mechanisms of 117 exogenous antigen presentation, through the participation of antigen transporters. This should contribute to both the fields of cellular biology and immunology. 118 5.7 Bibliography Ackerman, A. L., C. tvyritsis, et al. (2003). \"Early phagosomes in dendritic cells form a cellular compartment sufficient for cross presentation of exogenous antigens.\" Proc Natl Acad Sci U S A 100(22): 12889-94. Alimonti, J., Q. J. Zhang, et al. (2000). \" TAP expression provides a general method for improving the recognition of malignant cells in vivo.\" Nat Biotechnol 18(5): 515-20. Brossart, P. and M. J. Bevan (1997). \"Presentation of exogenous protein antigens on major histocompatibility complex class I molecules by dendritic cells: pathway of presentation and regulation by cytokines.\" Blood 90(4): 1594-9. Gallegos, A. M. and M. J. Bevan (2004). \"Driven to autoimmunity: the nod mouse.\" Cel l 117(2): 149-51. Goldsmith, Z. G. and N. Dhanasekaran (2004). \"The microrevolution: applications and impacts of microarray technology on molecular biology and medicine (review).\" Int J Mo l Med 13(4): 483-95. Guermonprez, P., L. Saveanu, et al. (2003). \"ER-phagosome fusion defines an M H C class I cross-presentation compartment in dendritic cells.\" Nature 425(6956): 397-402. Heath, W. R. and F. R. Carbone (2001). \"Cross-presentation, dendritic cells, tolerance and immunity.\" Annu Rev Immunol 19: 47-64. Heemels, M . T., T. N. Schumacher, et al. (1993). \"Peptide translocation by variants of the transporter associated with antigen processing.\" Science 262(5142): 2059-63. Houde, M., S. Bertholet, et al. (2003). \"Phagosomes are competent organelles for antigen cross-presentation.\" Nature 425(6956): 402-6. Liu, T., B. Chambers, et al. (1997). \" TAP peptide transporter-independent presentation of heat-killed Sendai virus antigen on M H C class I molecules by splenic antigen-presenting cells.\" J Immunol 159(11): 5364-71. Lizee, G., G. Basha, et al. (2003). \"Control of dendritic cell cross-presentation by the major histocompatibility complex class I cytoplasmic domain.\" Nat Immunol 4(11): 1065-73. Paschen, A., K. E. Dittmar, et al. (2000). \"Human dendritic cells infected by Listeria monocytogenes: induction of maturation, requirements for phagolysosomal escape and antigen presentation capacity.\" Eur J Immunol 30(12): 3447-56. Schinkel, A. H., U. Mayer, et al. (1997). \"Normal viability and altered pharmacokinetics in mice lacking mdrl-type (drug-transporting) P-glycoproteins.\" Proc Natl Acad Sci U S A 94(8): 4028-33. Schinkel, A. H., J. J. Smit, et al. (1994). \"Disruption of the mouse mdr la P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs.\" Cel l 77(4): 491-502. Schumacher, T. N., D. V. Kantesaria, et al. (1994). \"Peptide length and sequence specificity of the mouse TAP1/TAP2 translocator.\" J Exp Med 179(2): 533-40. Shastri, N. and F. Gonzalez (1993). \"Endogenous generation and presentation of the ovalbumin peptide/Kb complex to T cells.\" J Immunol 150(7): 2724-36. Sigal, L. J. and K. L. Rock (2000). \"Bone marrow-derived antigen-presenting cells are required for the generation of cytotoxic T lymphocyte responses to viruses and use transporter 119 associated with antigen presentation (TAP)-dependent and -independent pathways of antigen presentation.\" J Exp Med 192(8): 1143-50. Steinman, R. M., D. Hawiger, et al. (2003). \"Tolerogenic dendritic cells.\" Annu Rev Immunol 21:685-711. Steinman, R. M . and M. C. Nussenzweig (2002). \"Avoiding horror autotoxicus: the importance of dendritic cells in peripheral T cell tolerance.\" Proc Natl Acad Sci U S A 99(1): 351-8. van Kaer, L., P. G. Ashton-Rickardt, et al. (1992). \"TAP1 mutant mice are deficient in antigen presentation, surface class 1 molecules, and CD4-8 + T cells.\" Cel l 71: 1205. Yewdell, J. W. and J. R. Bennink (2001). \"Cut and trim: generating M H C class I peptide ligands.\" Curr Opin Immunol 13(1): 13-8. Yewdell, J. W., C. C. Norbury, et al. (1999). \"Mechanisms of exogenous antigen presentation by M H C class I molecules in vitro and in vivo: implications for generating CD8+ T cell responses to infectious agents, tumors, transplants, and vaccines.\" Adv Immunol 73: 1-77. 120 A P P E N D I X I M U L T I P L E A B C T R A N S P O R T E R RT-PCR D A T A Appendix I. M U L T I P L E A B C T R A N S P O R T E R R T - P C R D A T A R T - P C R data that comprised Table 2.1 is included below: N T = no treatment, IFN = interferon-y, HI Sen = heat inactivated Sendai virus, O V A = ovalbumin, H E L = hen egg lysozyme Splenic D C s NT IFN HI Sen OVA 12345678 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 31 32 33 E . t E r-- r ~ f 5 f r -1=ABCA1, 2=A2, 3=A3, 4=A4, 5=A7, 6=A8, 7=B1, 8=B4, 9=B8, 10=B10, 11=B11, 12=C1, 13=C2, 14=C5, 15=C8, 16=D1, 17=D2, 18=F2, 19=F3, 20=G1, 21=G2, 22=G3, 23=ABC7, 24=C9, 25=C10, 26=D4, 27=E1, 28=D3, 29=F1, 31=B2(TAP1), 32=B3(TAP2), 33=B9(TAPL). 122 Splenic macrophages 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 31 32 33 NT IFN HI Sen OVA 1 = A B C A 1 , 2=A2, 3=A3, 4=A4, 5=A7, 6=A8, 7=B1, 8=B4, 9=B8, 10=B10, 11=B11, 12=C1, 13=C2, 14=C5, 15=C8, 16=D1, 17=D2, 18=F2, 19=F3, 20=G1, 21=G2, 22=G3, 2 3 = A B C 7 , 24=C9, 25=C10, 26=D4, 27=E1, 28=D3, 29=F1, 31=B2(TAP1) , 32=B3(TAP2) , 33=B9(TAPL). 123 Splenic B-lvmphocytes 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 31 32 33 NT IFN HI Sen OVA 1 = A B C A 1 , 2=A2, 3=A3, 4=A4, 5=A7, 6=A8, 7=B1, 8=B4, 9=B8, 10=B10, 11=B11, 12=C1, 13=C2, 14=C5, 15=C8, 16=D1, 17=D2, 18=F2, 19=F3, 20=G1, 21=G2, 22=G3, 2 3 = A B C 7 , 24=C9, 25=C10, 26=D4, 27=E1, 28=D3, 29=F1, 31=B2(TAP1), 32=B3(TAP2), 3 3 = B 9 ( T A P L ) . 124 "@en ; edm:hasType "Thesis/Dissertation"@en ; vivo:dateIssued "2005-05"@en ; edm:isShownAt "10.14288/1.0099838"@en ; dcterms:language "eng"@en ; ns0:degreeDiscipline "Zoology"@en ; edm:provider "Vancouver : University of British Columbia Library"@en ; dcterms:publisher "University of British Columbia"@en ; dcterms:rights "For non-commercial purposes only, such as research, private study and education. Additional conditions apply, see Terms of Use https://open.library.ubc.ca/terms_of_use."@en ; ns0:scholarLevel "Graduate"@en ; dcterms:title "The ABCs of antigen presentation : defining antigen processing pathways"@en ; dcterms:type "Text"@en ; ns0:identifierURI "http://hdl.handle.net/2429/17320"@en .