Developmental Regulation of Chlamydia trachomatis Class I Accessible Protein 1, a CD8 + T Cell Antigen

Similar documents
Gamma Interferon Production by Cytotoxic T Lymphocytes Is Required for Resolution of Chlamydia trachomatis Infection

Coronaviruses cause acute, mild upper respiratory infection (common cold).

Laboratory diagnosis of congenital infections

Medical Virology Immunology. Dr. Sameer Naji, MB, BCh, PhD (UK) Head of Basic Medical Sciences Dept. Faculty of Medicine The Hashemite University

Determination of the temporal pattern and importance of BALF1 expression in Epstein-Barr viral infection

Jyotika Sharma, Feng Dong, Mustak Pirbhai, and Guangming Zhong*

Mechanismof interferon- -mediated anti-chlamydia infection

Influenza virus exploits tunneling nanotubes for cell-to-cell spread

Supplementary Figure 1 Role of Raf-1 in TLR2-Dectin-1-mediated cytokine expression

The toll-like receptor 4 ligands Mrp8 and Mrp14 play a critical role in the development of autoreactive CD8 + T cells

Hepatitis B Antiviral Drug Development Multi-Marker Screening Assay

The Case of the Spring Break Consequences

Chapter 6- An Introduction to Viruses*

SUPPLEMENTARY INFORMATION

VIRUSES. Biology Applications Control. David R. Harper. Garland Science Taylor & Francis Group NEW YORK AND LONDON

Principles of Adaptive Immunity

Sestrin2 and BNIP3 (Bcl-2/adenovirus E1B 19kDa-interacting. protein3) regulate autophagy and mitophagy in renal tubular cells in. acute kidney injury

Antigen Presentation to T lymphocytes

ALETA : MODE OF ACTION

IMMUNOLOGIC REACTIVITY IN HUMAN BREAST CANCER AGAINST CULTURED HUMAN BREAST TUMOR CELLS

The Infectious Cycle. Lecture 2 Biology W3310/4310 Virology Spring You know my methods, Watson --SIR ARTHUR CONAN DOYLE

PBMC from each patient were suspended in AIM V medium (Invitrogen) with 5% human

DNA codes for RNA, which guides protein synthesis.

I. Critical Vocabulary

Cytotoxic-T-Lymphocyte-Mediated Cytolysis of L Cells Persistently Infected with Chlamydia spp.

Graduate Survey - May 2014 (Human Services)

IL-13 Augments Compressive Stress-induced Tissue Factor Expression in Human Airway Epithelial Cells

Zheng, BJ; Du, LY; Zhao, GY; Lin, YP; Sui, HY; Chan, C; Ma, S; Guan, Y; Yuen, KY. Citation Hong Kong Medical Journal, 2008, v. 14 suppl. 4, p.

Figure S1. Schematic presentation of genomic replication of idsiv after transfection and infection. After transfection of idsiv plasmid DNA into 293T

Montgomery County Community College BIO 241 Clinical Microbiology II 4-3-3

Supplemental Table I.

Superior Fluorescent Labeling Dyes Spanning the Full Visible Spectrum...1. Trademarks: HiLyte Fluor (AnaSpec, Inc.)

Supplementary Information

Lecture 2: Virology. I. Background

HLA and antigen presentation. Department of Immunology Charles University, 2nd Medical School University Hospital Motol

Received 12 July 2011/Returned for modification 29 July 2011/Accepted 29 August 2011

Islet viability assay and Glucose Stimulated Insulin Secretion assay RT-PCR and Western Blot

Instructions for Use. APO-AB Annexin V-Biotin Apoptosis Detection Kit 100 tests

C H A R A C T E R I Z A T I O N O F T H E N O V E L D O M A I N W I T H N O N A M E G E N E I N C O L O N C A N C E R

MicroRNA and Male Infertility: A Potential for Diagnosis

Supplementary Figure 1 Cytokine receptors on developing thymocytes that can potentially signal Runx3d expression.

cure research HIV & AIDS

Chapter 6. Antigen Presentation to T lymphocytes

Chlamydia trachomatis Co-opts the FGF2 Signaling Pathway to Enhance Infection

Supplementary Data 1. Alanine substitutions and position variants of APNCYGNIPL. Applied in

Supplementary information

Chlamydia. By Madhuri Reddy

LESSON 4.4 WORKBOOK. How viruses make us sick: Viral Replication

Ali Alabbadi. Bann. Bann. Dr. Belal

Completing the CIBMTR Confirmation of HLA Typing Form (Form 2005)

CELL BIOLOGY - CLUTCH CH THE IMMUNE SYSTEM.

Microbiology of Atypical Pneumonia. Dr. Mohamed Medhat Ali

Production of Interferon Alpha by Dengue Virus-infected Human Monocytes

Controls for Chlamydia trachomatis and Neisseria gonorrhoea

HLA and antigen presentation. Department of Immunology Charles University, 2nd Medical School University Hospital Motol

Vasoactive intestinal peptide inhibits IFN-α secretion and modulates the immune function of plasmacytoid dendritic cells

Anti-infectious Immunity

Supplemental Information. T Cells Enhance Autoimmunity by Restraining Regulatory T Cell Responses via an Interleukin-23-Dependent Mechanism

Recombinant Protein Expression Retroviral system

Generating Spontaneous Copy Number Variants (CNVs) Jennifer Freeman Assistant Professor of Toxicology School of Health Sciences Purdue University

Persistent Infection of MDCK Cells by Influenza C Virus: Initiation and Characterization

PERSISTENT INFECTIONS WITH HUMAN PARAINFLUENZAVIRUS TYPE 3 IN TWO CELL LINES

Supporting Online Material for

7.012 Quiz 3 Answers

Live cell imaging of trafficking of the chaperone complex vaccine to the ER. BMDCs were incubated with ER-Tracker Red (1 M) in staining solution for

International Journal of Science, Environment and Technology, Vol. 7, No 1, 2018,

Chapter 35 Active Reading Guide The Immune System

Role of cyclooxygenase-2 in H5N1 viral pathogenesis and the potential use of its inhibitors

Supplementary Table; Supplementary Figures and legends S1-S21; Supplementary Materials and Methods

Viruses Tomasz Kordula, Ph.D.

Diagnostic Methods of HBV and HDV infections

Materials and Methods , The two-hybrid principle.

Proteomic profiling of small-molecule inhibitors reveals dispensability of MTH1 for cancer cell survival

Chlamydia muridarum-specific CD4 T-Cell Clones Recognize Infected Reproductive Tract Epithelial Cells in an Interferon-Dependent Fashion

Immunity to Murine Chlamydia trachomatis Genital Tract Reinfection Involves B Cells and CD4 T Cells but Not CD8 T Cells

SUPPLEMENTARY FIGURES AND TABLES

ab CytoPainter Golgi/ER Staining Kit

Effect of Azithromycin plus Rifampin versus That of Azithromycin Alone on the Eradication of Chlamydia pneumoniae

Immunology Lecture 4. Clinical Relevance of the Immune System

Interferon γ regulates idiopathic pneumonia syndrome, a. Th17 + CD4 + T-cell-mediated GvH disease

The Human Major Histocompatibility Complex

Innate Immunity & Inflammation

Basis and Clinical Applications of Interferon

Bihong Zhao, M.D, Ph.D Department of Pathology

The immune response against Chlamydia suis genital tract infection partially protects against re-infection

Antigen Presentation and T Lymphocyte Activation. Abul K. Abbas UCSF. FOCiS

Product Datasheet. HLA ABC Antibody (W6/32) NB Unit Size: 0.25 mg. Store at -20C. Avoid freeze-thaw cycles. Reviews: 1 Publications: 22

The Microimmunofluorescence Test for Chlamydia pneumoniae Infection: Technique and Interpretation

Department of Surgery, Duke Comprehensive Cancer Ctr. Duke University Medical Center, Durham, NC AVI BioPharma, Inc. Corvallis, OR

Section 6. Junaid Malek, M.D.

Gladstone Institutes, University of California (UCSF), San Francisco, USA

Towards an HIV Cure Pre-Conference Symposium 20 & 21 July 2012

chapter 17: specific/adaptable defenses of the host: the immune response

Adaptive Immunity to Bacteria. T cell subsets

Controls for Chlamydia trachomatis and Neisseria gonorrhoea

Chapter 1. Chapter 1 Concepts. MCMP422 Immunology and Biologics Immunology is important personally and professionally!

6/11/15. BACTERIAL STDs IN A POST- HIV WORLD. Learning Objectives. How big a problem are STIs in the U.S.?

Supplementary materials and methods.

What is the immune system? Types of Immunity. Pasteur and rabies vaccine. Historical Role of smallpox. Recognition Response

Page 32 AP Biology: 2013 Exam Review CONCEPT 6 REGULATION

Transcription:

BRIEF REPORT Developmental Regulation of Chlamydia trachomatis Class I Accessible Protein 1, a CD8 + T Cell Antigen Zarine R. Balsara, Nadia R. Roan, Lisa N. Steele, and Michael N. Starnbach Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts In this report, we show that the Chlamydia trachomatis specific CD8 + T cell antigen class I accessible protein 1 (Cap1) is expressed during the early stages of the C. trachomatis developmental cycle. We provide additional evidence suggestingthat Cap1 may be important in early immune recognition of the organism. Understanding the temporal and spatial expression of CD8 + T cell antigens such as Cap1 may be beneficial in designing multisubunit vaccines to stimulate a vigorous immune response against C. trachomatis. Chlamydia trachomatis is an obligate intracellular bacterium that causes a wide range of diseases in humans and animals. C. trachomatis has a unique biphasic developmental cycle [1]. The elementary body, the infectious form of the bacterium, shows no metabolic activity. Once elementary bodies enter the host cell, they begin to convert to noninfectious reticulate bodies that are metabolically active and that replicate within a specialized vacuole known as an inclusion. Approximately 20 h after infection, reticulate bodies begin to convert back to elementary bodies. By 36 h after C. trachomatis infection, the host cell lyses, and elementary bodies are released into the extracellular environment to begin further rounds of infection and replication. The resolution of C. trachomatis infection and resistance to reinfection depends on the coordinated activity of B cells, CD4 + Received 25 September 2005; accepted 13 December 2005; electronically published 12 April 2006. Potential conflicts of interest: none reported. Financial support: National Institutes of Health (grants AI-039558 and AI-055900). Reprints or correspondence: Dr. Michael Starnbach, Dept. of Microbiology and Molecular Genetics, Harvard Medical School, 200 Longwood Ave., Boston, MA 02115 (starnbach@ hms.harvard.edu). The Journal of Infectious Diseases 2006; 193:1459 63 2006 by the Infectious Diseases Society of America. All rights reserved. 0022-1899/2006/19310-0017$15.00 T cells, and CD8 + T cells (reviewed in [2]). We and others have been particularly interested in understanding how CD8 + T cells control C. trachomatis infection. The recent discovery of a number of C. trachomatis specific CD8 + T cell antigens [3 5] has begun to facilitate these studies. One of the first C. trachomatis specific CD8 + T cell antigens to be identified was class I accessible protein 1 (Cap1), a 31- kda protein of unknown function [4]. Fling et al. [4] initially demonstrated that Cap1 is expressed in fibroblasts at the C. trachomatis inclusion membrane 24 h after infection. More recently, we have begun to use Cap1 as a tool to study the initiation of the C. trachomatis specific CD8 + T cell response [6]. Once activated, CD8 + T cells differentiate into effector cells and acquire the important task of recognizing C. trachomatis infected cells and targeting them for destruction. However, because C. trachomatis has a strictly coordinated developmental cycle, many antigens may be regulated in their expression and may not always be available for recognition by CD8 + T cells. Therefore, to determine which classes of C. trachomatis proteins may contribute most to CD8 + T cell mediated clearance of this pathogen, it is essential to understand the temporal and spatial expression of a diversity of these antigens in their host cells. Here, we characterize the subcellular compartmentalization and temporal expression of Cap1 in C. trachomatis infected host cells in vitro. Using a combination of different techniques, we address the questions of when Cap1 is initially expressed at both the transcript and protein level, whether the protein remains localized to the inclusion membrane throughout the C. trachomatis developmental cycle, and when infected host cells are first able to be recognized by Cap1-specific CD8 + T cells. Materials and methods. McCoy, HeLa, and BALB 3T3 cells (American Type Culture Collection) were infected with C. trachomatis serovar L2 434/Bu in a sucrose-phosphate-glutamate media by centrifugation of bacteria with cells at 1200 g for 1 hat37 C. After centrifugation, the inoculum was removed and replaced with Dulbecco s modified Eagle medium (HeLa), MEM (McCoy), or RPMI 1640 medium (BALB/3T3) containing 10% fetal bovine serum. For real-time polymerase chain reaction (PCR) analysis, RNA was isolated from cells using the Ultraspec RNA system (Biotecx Laboratories). One microgram of total RNA from each sample was reverse-transcribed using 3 -specific primers to generate cdna specific for cap1, inca, and 16s rrna. Real-time PCR was then performed on each cdna sample, using the ABI Prism 7700 Sequence Detection System (Applied Biosystems). The primers and probes used in these studies were designed using BRIEF REPORT JID 2006:193 (15 May) 1459

Primer Express software (Applied Biosystems). The initial amount of cap1 or 16s rrna cdna template in each sample was calculated by comparison with a standard curve in which known dilutions of C. trachomatis L2 genomic DNA were amplified using the same gene-specific primers and probe. Cells analyzed by fluorescent in situ hybridization (FISH) were grown and infected on glass coverslips. Cells were fixed and processed for FISH, essentially as described elsewhere [7, 8]. Cells were hybridized at 35 C with a Cy5-labeled probe complementary to 18 nt of the cap1 mrna sequence, counterstained with the nucleic acid stain 4,6-diamidino-2-phenylindole dihydrochloride (DAPI), and visualized with a Nikon E1000 automated upright epifluorescence microscope (Nikon Imaging Facility, Harvard Medical School, Boston). For immunofluorescence studies, cells were grown and infected on glass coverslips. Cells were fixed in 100% methanol for 10 min at room temperature, washed in PBS, and blocked with 10% donkey serum and 2% bovine serum albumin. Cells were then costained with a mouse monoclonal antibody against the C. trachomatis major outer membrane protein, MOMP (Accurate Chemical and Scientific), and a rabbit polyclonal antibody against Cap1 [4]. The anti-momp antibody was visualized in combination with a fluorescein isothiocyanate conjugated donkey anti-mouse IgG antibody, whereas the anti-cap1 antibody was visualized in combination with a rhodamine red X conjugated donkey anti-rabbit IgG (Jackson ImmunoResearch Laboratories). To evaluate processing and presentation of Cap1 antigen, BALB/3T3 cells were fixed at different times after infection with 1% paraformaldehyde for 15 min at room temperature before being cultured with the Cap1-specific CD8 + T cell line 69 [9]. Line 69 was maintained and expanded by weekly stimulation on C. trachomatis infected cells, as described elsewhere [9]. Supernatants from the cocultured cells were analyzed for levels of interferon (IFN) g, using an IFN-g Minikit (Pierce). Results. To examine the transcriptional regulation of cap1, we performed real-time PCR analysis on C. trachomatis infected cells. McCoy cells, a murine fibroblast cell line, were infected with C. trachomatis at an MOI of 1:1 in the presence of 1.5 mg/ml cycloheximide. At various times after infection, total RNA was isolated and reverse-transcribed to generate cdna specific for cap1, inca, and 16s rrna. Real-time PCR was then performed on each cdna sample. Because production of 16s rrna transcripts by individual organisms remains relatively constant during infection [10], the amount of cap1 or inca in each sample was divided by the amount of 16s rrna in the sample, to normalize for the increase in the number of C. trachomatis organisms over time. For each time point, the fold change in cap1 mrna levels relative to that present in the initial inoculum (0 h) is plotted in figure 1A. As early as 4 h after infection, cap1 transcript levels were up-regulated. cap1 Figure 1. Transcription of cap1 early in the Chlamydia trachomatis developmental cycle. A, Infection of McCoy cells with C. trachomatis for various lengths of time. Total RNA was isolated at each time point and was used to generate cdna specific for cap1 and 16s rrna. Real-time polymerase chain reaction was then performed in duplicate on the cdna to determine the amount of cap1 and 16s rrna present in each sample. cap1 levels were normalized against the levels of 16s rrna in each sample, to control for the increase in the total number of C. trachomatis organisms present over time. Bars represent the fold change in cap1 transcript levels for each time point relative to the amount of cap1 mrna present in the initial inoculum (0 h after infection). The graph depicts data from 1 of 3 representative experiments. Error bars represent the range of data collected for each time point in this particular experiment. B, Infection of HeLa cells with C. trachomatis foreither20hor42h. Cells were fixed and hybridized with a Cy5-labeled oligomer that recognizes cap1 mrna. Cells were also counterstained with the nucleic acid stain DAPI and then visualized by epifluorescence microscopy. White stars are included to depict the location of the C. trachomatis inclusion. mrna levels remained elevated until 20 h after infection, after which time transcript levels dropped below that present in the initial elementary body inoculum. cap1 transcript levels modestly rose again between 24 and 36 h after infection. This profile of cap1 transcription was in contrast to that observed for inca, which encodes an inclusion membrane protein previously shown to be expressed at the middle of the C. trachomatis developmental 1460 JID 2006:193 (15 May) BRIEF REPORT

Figure 2. Expression of Cap1 protein at significant levels within an infected cell as early as 8 10 h after infection. A, Infection of HeLa cells with Chlamydia trachomatis for 4 h (i iii), 10 h (iv vi), 14 h (vii ix), 18 h (x xii), 24 h (xiii xv), or 36 h (xvi xviii). Cells were fixed and costained with a mouse monoclonal antibody against the C. trachomatis major outer membrane protein (MOMP) (fluorescein isothiocyanate [FITC] conjugated secondary antibody; i, iv, vii, x, xiii, and xvi) and a rabbit polyclonal antibody against Cap1 (rhodamine red X [RRX] conjugated secondary antibody; ii, v, viii, xi, xiv, and xvii) and then visualized by epifluorescence microscopy. FITC (green) and RRX (red) images were overlaid to determine the relative localization of MOMP and Cap1 (iii, vi, ix, xii, xv, and xviii). B, Infection of BALB/3T3 epithelial cells with C. trachomatis for the indicated times. Cells were fixed and coincubated with a Cap1-specific CD8 + T cell line. After 16 h of coincubation, the amount of interferon (IFN) g secreted into the supernatant was measured by ELISA. Detection of IFN-g in the cultures indicates the recognition of infected BALB/3T3 cells by the T cell line. The data shown are representative of 3 separate experiments. cycle [11]. Similar to what was described by Scidmore-Carlson et al. [11], we observed that inca mrna was first expressed between 12 and 14 h after infection (data not shown). inca levels peaked by 20 h after infection (1100-fold up-regulation), declined significantly by 24 h after infection, and then rose again by 36 h after infection. These real-time PCR studies suggest that there is early transcription of cap1 after C. trachomatis infection. This finding is consistent with existing data generated by microarray analysis in which Belland et al. [12] identified the cap1 homologue from C. trachomatis serovar D as an immediate-early gene. However, during later stages of infection, when C. trachomatis inclusions become more asynchronous in their development, it becomes more difficult to use real-time PCR or microarray analysis to accurately evaluate the transcriptional regulation of cap1. The overall changes in cap1 mrna levels detected by either of these methods could result from transcriptional regulation within individual reticulate bodies or could simply reflect the conversion of a number of reticulate bodies to metabolically inactive elementary bodies. Therefore, to begin to address this concern, we used FISH to examine cap1 mrna expression at the level of the individual inclusion. HeLa cells, a human cervical epithelial cell line, were infected with C. trachomatis at an MOI of 3:1 in the presence of 1.5 mg/ml cycloheximide. At various times after infection, cells were fixed and stained with a fluorescently labeled probe recognizing cap1 mrna. To confirm that our fluorescent probe was specifically binding cap1 mrna and not genomic DNA, FISH analysis was also performed on infected cells using a fluorescently labeled oligomer complementary to the cap1 probe. Minimal signal was detected using this oligomer (data not shown). Unfortunately, FISH proved far less sensitive than realtime PCR in detecting cap1 transcripts in infected cells. In fact, cap1 mrna was not detected by FISH until 18 h after C. trachomatis infection (data not shown). Furthermore, because we were unable to visually distinguish reticulate bodies from elementary bodies within an inclusion, the transcriptional regulation of cap1 by individual organisms could not be compared. Nevertheless, we observed that although cap1 mrna was present within the inclusion at 20 h after infection, transcripts were no longer detectable within large inclusions at 42 h after infection (figure 1B). Our FISH analysis suggests that transcription of cap1 most likely declines in individual inclusions late in the C. trachomatis developmental cycle and remains undetectable for the duration of the cycle. The subsequent increase in cap1 transcription observed in our real-time PCR studies at 36 h after infection (figure 1A) most likely reflects the asynchrony of C. trachomatis infection. By 36 h, many inclusions have already lysed, and organisms have begun new rounds of infection and transcriptional up-regulation of early genes such as cap1. These studies suggest that a combination of real-time PCR and FISH analysis may BRIEF REPORT JID 2006:193 (15 May) 1461

allow for enhanced sensitivity in measuring transcriptional regulation of C. trachomatis genes throughout the entire developmental cycle of the organism. To determine whether this early transcription of cap1 also correlates with early protein expression, we then used indirect immunofluorescence microscopy to study the temporal and spatial expression of Cap1 protein. HeLa cells were infected with C. trachomatis at an MOI of 10:1. At different times after infection, cells were fixed and costained with antibodies specific for the Cap1 protein [4] and MOMP. Antibodies against MOMP detect individual elementary bodies and reticulate bodies within a Chlamydia inclusion. As shown in figure 2A, Cap1 protein was initially detected at the C. trachomatis inclusion membrane at 8 10 h after infection, when single reticulate bodies are first detected within the inclusion. Because immunofluorescence analysis is somewhat limited in its sensitivity, it is also possible that Cap1 is expressed earlier within infected cells. Cap1 protein levels continued to increase by 14 18 h after infection and remained stably expressed as late as 36 h after infection. Cap1 appeared to localize predominantly at the inclusion membrane throughout the course of the C. trachomatis developmental cycle. Our analysis of cap1 expression indicates that this gene is transcribed and translated soon after C. trachomatis infects nonprofessional antigen-presenting cells. Cap1 protein may, therefore, be available for processing and presentation on major histocompatibility complex (MHC) class I relatively early in the C. trachomatis developmental cycle. To test this hypothesis, a murine epithelial cell line, BALB/3T3, was infected with C. trachomatis at an MOI of 100:1. At various times after infection, cells were fixed and cocultured with the Cap1-specific CD8 + T cell line 69 [4]. Recognition of infected BALB/3T3 cells by this T cell line was measured using an IFN-g ELISA (figure 2B). These T cells recognized Cap1 on the surface of infected BALB/3T3 cells as early as 8 h after C. trachomatis infection and continued to recognize infected cells as late as 20 h after infection. In evaluating these findings, it is important to consider that recognition of infected cells by T cells is more reliable for evaluating initial protein expression than for measuring the decline in protein expression, because of the substantial half-life of MHC-peptide complexes at the cell surface [13, 14]. Discussion. In this study, we examined the temporal and spatial expression pattern of the C. trachomatis specific CD8 + T cell antigen Cap1. Using a variety of techniques, we determined that Cap1 is expressed at the early stages of infection. The fact that, by 8 h after infection, sufficient Cap1 protein is expressed to stimulate CD8 + T cells suggests that Cap1 expression may be critical in early immune detection of C. trachomatis infected cells. Cap1 is one of a growing number of proteins that are being described as CD8 + T cell antigens. The interest in identifying and characterizing C. trachomatis specific CD8 + T cell antigens stems, in part, from a desire to develop an effective vaccine against this pathogen. Past studies have focused on boosting antibody titers and the CD4 + T cell response against Chlamydia (reviewed in [15]). However, the additional inclusion of a number of different CD8 + T cell antigens into a vaccine may prove to be a promising way of inducing long-lasting immunity against C. trachomatis. The temporal and spatial expression studies we performed with Cap1 can be expanded to other C. trachomatis specific T cell antigens. Choosing a diversity of antigens that overlap in their temporal expression would ensure that there is no period during the C. trachomatis developmental cycle when the organism could escape recognition by the host immune system. This approach may increase the possibility of developing a vaccine that achieves sterilizing immunity against C. trachomatis. In addition, because genetic manipulation of C. trachomatis remains elusive, it has been difficult to attribute functions to most C. trachomatis specific proteins. However, knowing the developmental regulation and subcellular compartmentalization of C. trachomatis proteins allows one to hypothesize a range of putative functions for these proteins. This information, together with insights into the cellular biology of C. trachomatis infection and protein-protein interactions, may help to elucidate the role of specific proteins in the pathogenesis of C. trachomatis. Acknowledgments We thank James Nicholas Lafave and David Regan, for technical support, and Sarah D Orazio and Adrianus van der Velden, for critical manuscript reading. References 1. Stephens RS. Chlamydia: intracellular biology, pathogenesis, and immunity. Washington, DC: ASM Press, 1999. 2. Loomis WP, Starnbach MN. T cell responses to Chlamydia trachomatis. Curr Opin Microbiol 2002; 5:87 91. 3. Starnbach MN, Loomis WP, Ovendale P, et al. An inclusion membrane protein from Chlamydia trachomatis enters the MHC class I pathway and stimulates a CD8 + T cell response. J Immunol 2003; 171:4742 9. 4. Fling SP, Sutherland RA, Steele LN, et al. CD8 + T cells recognize an inclusion membrane-associated protein from the vacuolar pathogen Chlamydia trachomatis. Proc Natl Acad Sci USA 2001; 98:1160 5. 5. Kim SK, Angevine M, Demick K, et al. Induction of HLA class I- restricted CD8 + CTLs specific for the major outer membrane protein of Chlamydia trachomatis in human genital tract infections. J Immunol 1999; 162:6855 66. 6. Steele LN, Balsara ZR, Starnbach MN. Hematopoietic cells are required to initiate a Chlamydia trachomatis-specific CD8 + T cell response. J Immunol 2004; 173:6327 37. 7. Poppert S, Essig A, Marre R, Wagner M, Horn M. Detection and differentiation of chlamydiae by fluorescence in situ hybridization. Appl Environ Microbiol 2002; 68:4081 9. 8. Manz W, Amann R, Ludwid W, Wagner M, Schleifer KH. Phylogenetic 1462 JID 2006:193 (15 May) BRIEF REPORT

oligodeoxynucleotide probes for the major subclass of proteobacteria: problems and solutions. Syst Appl Microbiol 1992; 15:593 600. 9. Starnbach MN, Bevan MJ, Lampe MF. Protective cytotoxic T lymphocytes are induced during murine infection with Chlamydia trachomatis. J Immunol 1994; 153:5183 9. 10. Mathews SA, Volp KM, Timms P. Development of a quantitative gene expression assay for Chlamydia trachomatis identified temporal expression of sigma factors. FEBS Lett 1999; 458:354 8. 11. Scidmore-Carlson MA, Shaw EI, Dooley CA, Fischer ER, Hackstadt T. Identification and characterization of a Chlamydia trachomatis early operon encoding four novel inclusion membrane proteins. Mol Microbiol 1999; 33:753 65. 12. Belland RJ, Zhong G, Crane DD, et al. Genomic transcriptional profiling of the developmental cycle of Chlamydia trachomatis. Proc Natl Acad Sci USA 2003; 100:8478 83. 13. Cerundolo V, Elliott T, Elvin J, Bastin J, Rammensee HG, Townsend A. The binding affinity and dissociation rates of peptides for class I major histocompatibility complex molecules. Eur J Immunol 1991; 21: 2069 75. 14. Luescher IF, Loez JA, Malissen B, Cerottini JC. Interaction of antigenic peptides with MHC class I molecules on living cells studied by photoaffinity labeling. J Immunol 1992; 148:1003 11. 15. Igietseme JU, Eko FO, Black CM. Contemporary approaches to designing and evaluating vaccines against Chlamydia. Expert Rev Vaccines 2003; 2:129 46. BRIEF REPORT JID 2006:193 (15 May) 1463