Paradoxic Effect of Anti-CD4 Therapy on Lacrimal Gland Disease in MKL/Mp-lpr/lpr Mice Douglas A. Jabs,* William H. Burns,^ and Robert A.

Similar documents
Autoimmune disease, including lacrimal gland inflammation,

Ocular Inflammation in Autoimmune MRL/Mp Mice

J Jpn Coll Angiol, 2009, 49: collagen disease, genetic polymorphism, MRL mice, recombinant inbred strains, Cd72. MRL/Mp-lpr/lpr MRL/ lpr

Explain the laboratory diagnosis of Rabies?

Citation Acta Medica Nagasakiensia. 1992, 37

Gastric Carcinoma with Lymphoid Stroma: Association with Epstein Virus Genome demonstrated by PCR

What will we discuss today?

Supplementary Appendix

RECIPIENT: Lankenau Institute for Medical Research, Wynnewood, PA 19096

Deficiencies of Vitamins E and A in the Rat: Lipofuscin Accumulation in the Choroid

FOCiS. Lecture outline. The immunological equilibrium: balancing lymphocyte activation and control. Immunological tolerance and immune regulation -- 1

Testosterone-Induced Suppression of Autoimmune Diseose in Locrimoi Tissue of o Mouse Model (NZD/NZW F1) of Sjogren's Syndrome

SHORT COMMUNICATION. Human Papillomavirus Type 11 E1 Ú E4 and L1 Proteins Colocalize in the Mouse Xenograft System at Multiple Time Points

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

The pathogenesis of nervous distemper

Acute Retinal Necrosis Secondary to Varicella Zoster Virus in an Immunosuppressed Post-Kidney Transplant Patient

TSH Receptor Monoclonal Antibody (49) Catalog Number MA3-218 Product data sheet

LIST OF ORGANS FOR HISTOPATHOLOGICAL ANALYSIS:!! Neural!!!!!!Respiratory:! Brain : Cerebrum,!!! Lungs and trachea! Olfactory, Cerebellum!!!!Other:!

Anti-Ly9 (CD229) antibody treatment reduces marginal zone B cell numbers and salivary gland inflammation in a mouse model of Sjögren s Syndrome

Appendix 1. A. Procedure for preparing histopathology slides. The liver removed and stored immediately in buffered formalin 10 % for

DC were seeded into tissue culture dishes in IMDM 2% FCS, and added with PMN. (1:1; PMN: DC) for 16h also in the presence of DNAse (100 U/ml); DC were

Role of Tyk-2 in Th9 and Th17 cells in allergic asthma

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

(A) PCR primers (arrows) designed to distinguish wild type (P1+P2), targeted (P1+P2) and excised (P1+P3)14-

The Lymphatic System and Body Defenses

Quantitative Analysis of Major Histocompatibility Complex Class II-Positive Cells in Posterior Segment of Royal College of Surgeons Rat Eyes

SUPPLEMENTARY FIGURE 1

Distribution of type IV collagen, laminin, nidogen and fibronectin in the haemodynamically stressed vascular wall

Expression of acid base transporters in the kidney collecting duct in Slc2a7 -/-

[AUTOIMMUNITY] July 14, 2013

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.

T Cells in the Uninjected Eye after Anterior Chamber Inoculation of Herpes Simplex Virus Type 1

Autoimmunity. Autoimmunity arises because of defects in central or peripheral tolerance of lymphocytes to selfantigens

Immunology: an overview Lecture

HRP cytochemistry. Division of Radiooncology, Deutsches Krebsforschungszentrum, Heidelberg, Germany

Third line of Defense

Immunodeficiency. (2 of 2)

Supplemental materials

3. Lymphocyte proliferation (fig. 15.4): Clones of responder cells and memory cells are derived from B cells and T cells.

Clinical Aspect and Application of Laboratory Test in Herpes Virus Infection. Masoud Mardani M.D,FIDSA

Chapter 16 Lymphatic System and Immunity. Lymphatic Pathways. Lymphatic Capillaries. network of vessels that assist in circulating fluids

and follicular helper T cells is Egr2-dependent. (a) Diagrammatic representation of the

Canberra, Australia). CD11c-DTR-OVA-GFP (B6.CD11c-OVA), B6.luc + and. Cancer Research Center, Germany). B6 or BALB/c.FoxP3-DTR-GFP mice were

SUPPLEMENTARY INFORMATION. Involvement of IL-21 in the epidermal hyperplasia of psoriasis

Reports. Spread of Murine Cytomegalovirus to Inner Ocular Structures Following Disruption of the Blood-Retina Barrier in Immunosuppressed BALB/c Mice

What is Autoimmunity?

What is Autoimmunity?

Nature Medicine: doi: /nm.4322

Ganciclovir-resistant CMV retinitis in a patient with wild-type plasma CMV. North Carolina School of Medicine, Chapel Hill, NC

Unit 5 The Human Immune Response to Infection

BIT 120. Copy of Cancer/HIV Lecture

Diseases of Immunity 2017 CL Davis General Pathology. Paul W. Snyder, DVM, PhD Experimental Pathology Laboratories, Inc.

Supporting Information

Primary Intraocular Lymphoma (PIOL)

During Murine Cytomegalovirus Infection

2014 SEVPAC Case #63 (Slide ID: #1)

Evaluation of Chromatin Clumping and Myelination of the Spinal Cord of Pigs with Congenital Tremor

Cells and viruses. Human isolates (A/Kawasaki/173/01 [H1N1], A/Yokohama/2057/03 [H3N2],

EBV and Infectious Mononucleosis. Infectious Disease Definitions. Infectious Diseases

7.012 Quiz 3 Answers

Effective activity of cytokine-induced killer cells against autologous metastatic melanoma including cells with stemness features

Potential Rebalancing of the Immune System by Anti-CD52 Therapy

Bachelor of Chinese Medicine ( ) AUTOIMMUNE DISEASES

Current Strategies in HIV-1 Vaccine Development Using Replication-Defective Adenovirus as a Case Study

NOTES: CH 43, part 2 Immunity; Immune Disruptions ( )

SUPPLEMENTARY INFORMATION

Campbell's Biology: Concepts and Connections, 7e (Reece et al.) Chapter 24 The Immune System Multiple-Choice Questions

COLLOID DROPLET FORMATION IN DOG THYROID IN VITRO

APPENDIX 1 ETHICAL CLEARANCE

Larger than Life. Angela Thomae Case Study CHEM 454

CONTENTS. STUDY DESIGN METHODS ELISA protocol for quantitation of mite (Dermatophagoides spp.) Der p 1 or Der f 1

SIV p27 ANTIGEN CAPTURE ASSAY

Molecular Pathology of Lymphoma (Part 1) Rex K.H. Au-Yeung Department of Pathology, HKU

react with these antibodies. Reports from different recent review Crocker and Burnett suggested that

Supplementary Appendix

The Role of CD4 + T Cells in Biphasic Hind Limb Paralysis Induced by the D Variant of Encephalomyocarditis Virus (EMC-D) in DBA/2 Mice

Determinants of Immunogenicity and Tolerance. Abul K. Abbas, MD Department of Pathology University of California San Francisco

Immunity to Viruses. Patricia Fitzgerald-Bocarsly September 25, 2008

Solution key Problem Set

. Autoimmune disease. Dr. Baha,Hamdi.AL-Amiedi Ph.D.Microbiology

Grave s Disease. of the endocrine system known as Grave s disease. This disease condition which affects the

7.013 Spring 2005 Problem Set 6

cure research HIV & AIDS

Post Kala-azar Dermal Leishmaniasis (PKDL) from the field to the cellular and the subcellular levels

Darwinian selection and Newtonian physics wrapped up in systems biology

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

2. The normal of the gut, and vagina keep the growth of pathogens in check. 3. in the respiratory tract sweep out bacteria and particles.

Reviewers' comments: Reviewer #1 Expert in EAE and IL-17a (Remarks to the Author):

Immunohistochemical Localization For Aldose Reductase in Diabetic Lenses

Morphological and functional state of immune organs in rats with experimental type 1 diabetes mellitus (DM-1)

Identification of Microbes Lecture: 12

ISOLATION OF A SARCOMA VIRUS FROM A SPONTANEOUS CHICKEN TUMOR

Introduction. Acute sodium overload produces renal tubulointerstitial inflammation in normal rats

Animal hosts Natural host Laboratory animals Rabbits Mice Rats Hamsters Newborn or suckling rodents Animal models for viral pathogenesis 4 Growth of v

Physiology Unit 3. ADAPTIVE IMMUNITY The Specific Immune Response

Herpesviruses. Virion. Genome. Genes and proteins. Viruses and hosts. Diseases. Distinctive characteristics

Overview of the Lymphoid System

Downloaded by on April 28, Publication Date: April 24, 1984 doi: /bk

all of the above the ability to impart long term memory adaptive immunity all of the above bone marrow none of the above

Transcription:

Paradoxic Effect of Anti-CD4 Therapy on Lacrimal Gland Disease in MKL/Mp-lpr/lpr Mice Douglas A. Jabs,* William H. Burns,^ and Robert A. Prendergast* Purpose. MKL/Mp-lpr/lpr mice (MRL/lpr) spontaneously develop lacrimal gland inflammatory lesions and are a model for the human disease Sjogren's syndrome. Therapy with monoclonal antibodies (mab) to CD4 ameliorates the autoimmune renal, vasculitic, and intraocular inflammatory lesions in MRL/lpr mice. The effect of anti-cd4 mab therapy on lacrimal gland immunopathology was evaluated. Methods. From 1 to 5 mondis of age, MRL/lpr mice were treated with weekly intraperitoneal injections of 2 mg anti-cd4 mab, after which they were killed and their lacrimal glands were removed for histologic evaluation and immunocytochemistry. Control mice were administered weekly intraperitoneal injections of either saline or normal rat immunoglobulin. Results. Anti-CD4 mab treatment produced no reduction in lacrimal gland inflammation but did change its morphology. In control mice, there were multiple sharply delineated foci of inflammatory cells in the lacrimal gland, whereas in anti-cd4 mab-treated mice, there was a more diffuse inflammation surrounding illdefined foci that spread throughout the gland. Immunocytochemistry revealed that in control mice, lesions were composed predominandy of CD4+ T cells, but in anti-cd4 mab-treated mice, CD8+ T cells predominated. Conclusions. Aldiough anti-cd4 mab therapy of MRL/ lpr mice eliminated autoimmune renal disease, autoantibody formation, and ocular inflammatory disease, it had a paradoxic effect on lacrimal gland lesions. Lacrimal gland lesions in the anti-cd4 mab-treated mice were not decreased, but they had a different morphology and a different immunocytochemical profile. Invest Ophthalmol Vis Sci. 1996; 37:246-250. WLKL/Mp-lpr/lpr (MRL/lpr) mice spontaneously develop an autoimmune disease characterized by vasculitis, lymphadenopathy, glomerulonephritis, and autoantibody production. 1 Among the autoimmune le- From the Departments of * Ophthalmology and f Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland. Supported by National Institutes of Health grants EY05912 and EY01 765 and by an unrestricted grant from Research to Prevent Blindness. Submitted for publication April 24, 1995; revised August 15, 1995; accepted September 13, 1995. Proprietary interest category: N. Reprint requests: Douglas A. Jabs, Wilmer Eye Institute, 550 N. Broadway, Suite 700, Baltimore, MD 21205. sions are lacrimal gland inflammatory infiltrates, which are a model for the human disorder, Sjogren's syndrome. 2 These lacrimal gland lesions are composed largely of CD4+ T cells, as are the other autoimmune lesions (including ocular and vasculitic), whereas the massively enlarged lymph nodes are composed primarily of Thy 1.2+, CD4-, and CD8- TCR a//3+ "double-negative" T cells. 2 ' 3 MRL/lpr mice are congenic with MRL/Mp +/+ (MRL/+) mice, which develop a later onset and milder systemic autoimmune disease. 1 The lpr mutation results in a defective Fas protein and defective apoptosis of lymphocytes. 4 The consequences of this defective apoptosis include the accumulation of double-negative T cells, resultant massive lymphadenopathy, and acceleration of the autoimmune disease inherent in the MRL/Mp strain. 1 Previous studies 5 ' 6 have demonstrated that treatment of MRL/lpr mice with monoclonal antibodies (mab) to the CD4 molecule resulted in a marked improvement in systemic autoimmune disease, including reduction in lymphadenopathy and autoantibody formation, and the elimination of renal and ocular disease. In this report, we describe the effect of anti-cd4 mab therapy on the lacrimal gland lesions in MRL/lpr mice. MATERIALS AND METHODS. Monoclonal Antibodies. Monoclonal antibodies were prepared as previously described. 5 ' 6 Hybridoma cells secreting mab of rat origin to the murine CD4 antigen L3T4 (GK 1.5) were obtained from ATCC (Rockville, MD). 7 One million cells were injected into the peritoneal cavity of severe combined immunodeficiency mice obtained from the Jackson Laboratories (Bar Harbor, ME). Antibody was harvested from the severe combined immunodeficiency peritoneal exudate by paracentesis starting 8 days after cell transfer. Immunoglobulin (Ig) was purified using 50% saturated ammonium sulfate precipitation, followed by solubilization and dialysis against phosphate-buffered saline (PBS). The partially purified mab was then standardized for protein content using spectrophotometric absorbance at 280 nm. Treatment Protocol. Female MRL/lpr mice were obtained from the Jackson Laboratories and kept under standard conditions in the animal facilities of the Woods Research Building of The Johns Hopkins Hospital. Animals were treated as previously described 5 ' 6 with one initial intravenous and one intraperitoneal injection of 2 mg each, starting at 1 month of age, followed by weekly intraperitoneal injections of 2 mg of anti-cd4 mab. Two control groups were used: The first was given injections of normal saline, and the second was given injections of normal rat Ig. The rat Ig 246 Investigative Ophthalmology & Visual Science, January 1996, Vol. 37, No. 1 Copyright Association for Research in Vision and Ophthalmology

Reports 247 used in these experiments was produced by subjecting normal rat serum to an ammonium sulfate precipitation and dialysis identical to that used from the preparation of the anti-cd4 mab. Animals were killed by exsanguination at 5 months of age. One lacrimal gland was removed, fixed in 4% buffered formaldehyde, embedded in paraffin, sectioned at 5 //m, and stained with hematoxylin and eosin. Histopathologic evaluation was performed by a masked reader (RAP) unaware of the treatment for each animal. In 10 control and 9 anti-cd4 mabtreated mice, the other lacrimal gland was removed, embedded in OCT compound (Miles, Elkhart, IN), frozen in liquid nitrogen, sectioned at 6 //m on a cryostat, and stained as outlined below. These experiments conformed to the ARVO Statement for the Use of Animals in Ophthalmic and Vision Research. Immunocytochemistry. Analysis of frozen sections of lacrimal glands was performed using a panel of monoclonal antibodies to cell surface markers and the avidin-biotin-peroxidase complex (ABC) technique. 2 ' 3 Briefly, frozen sections were fixed in chilled (4 C) acetone, air-dried, rehydrated in PBS, and incubated with the appropriate blocking agent (Vector Laboratories, Burlingame, CA) for 20 minutes. The primary antibody was applied, and the slides were incubated for 60 minutes. Slides were washed in PBS, incubated with a biotinylated secondary antibody for 30 minutes, rinsed in PBS, incubated with the ABC agent for 45 minutes, washed again in PBS, developed with a 0.1% nitrogen peroxide and 3-amino-9-ethyl-carbazole containing acetate buffer, and counterstained with Harris' hematoxylin (Sigma, St. Louis, MO). The percentage of positive mononuclear inflammatory cell staining with a monoclonal antibody was enumerated using a 10 X 10 grid net micrometer disc that covered an area of 0.16 mm 2 using a X25 objective and X10 ocular mounted in a standard Zeiss binocular microscope (Carl Zeiss, Oberkochen, Germany). 2 The mabs used for immunocytochemistry were rat anti-thy 1.2 for T cells (Becton-Dickinson, Mountain View, CA), rat anti-l3t4 (GK 1.5) for CD4+ T cells (Becton-Dickinson), rat anti-lyt 2 for CD8+ T cells (Becton-Dickinson), and a pan-b cell (pre-b, B cells, and plasma cells) mab (RA3-LC2/1, ATCC, Rockville, MD) for B cells. Virology. Frozen lacrimal glands were thawed and homogenized at 10% (wt/vol) in culture medium and assayed for infectious virus on primary mouse embryo cultures. Cultures were examined every few days for cytopathic effect and were discarded after 3 weeks if they remained negative. Frozen tissue samples were thawed in TE9 buffer and digested overnight at 37 C in proteinase K (100 //g/ml) and 1% sodium dodecyl sulfate. DNA was extracted with phenol.chloroform, precipitated from the aqueous phase, and resuspended in water. The extracted DNA was assayed by polymerase chain reaction for a unique region of the immediate early region of murine cytomegalovirus as performed by Henry and Hamilton, 8 using the primers designated by them as CHI6 and CHI7 and a probe using oligomer CHI5. The expected 310-bp amplified fragment was detected in these same DNA samples after "spiking" with DNA from plasmids containing this viral gene, and these DNA samples were positive for a fragment of the actin gene using standard polymerase chain reaction primers for actin. It was estimated that 150 copies of the viral genome could be detected in 1 fig of the sample DNA. RESULTS. Lacrimal Gland Histology. Lacrimal glands were obtained from nine saline-treated and nine rat Ig-treated control mice for histologic evaluation. All lacrimal glands from control mice developed the typical multifocal, well-demarcated inflammatory lesions previously described in MRL/lpr mice and graded either grade 3 or 4 using the previously described modified focus score scale. 2 In contrast, the lacrimal glands from the nine anti-cd4 mab-treated mice presented a distinct histopathology (Fig. 1), in which there was a more diffuse inflammatory infiltrate surrounding ill-defined foci that spread throughout the lacrimal gland parenchyma. Thus, there was no diminution in the amount of inflammation after anti- CD4 mab treatment, but there was a change in the morphology of the inflammation. Lacrimal Gland Immunocvtochemistry. Lacrimal glands from five saline-treated, five rat Ig-treated, and nine anti-cd4 mab-treated mice were stained for cell surface markers (Table 1). Results of the saline-treated and rat Ig-treated control groups were similar, and both control groups were combined for an analysis compared to anti-cd4 mab-treated mice. In both control groups, the infiltrate was predominately a CD4+ T cell infiltrate (mean, 55%), with lesser numbers of CD8+ T cells (mean, 19%) and B-cells (mean, 15%). In contrast, although the infiltrate in anti-cd4 mabtreated mice was largely a T-cell infiltrate, it was composed primarily of CD8+ T cells (Fig. 2); 11% of the infiltrating cells in lacrimal glands of anti-cd4 mabtreated mice were CD4+ cells (P = 0.003 versus controls), whereas 63% were CD8+ T cells (P = 0.003 versus controls). The percentage of B cells in the lacrimal glands of anti-cd4 mab-treated mice also was decreased when compared to control animals (mean, 4% versus 15%; P = 0.007). Lacrimal Gland Virology. Because a viral infection that required CD4+ T cells for control might produce lacrimal gland damage in anti-cd4 mab-treated mice, lacrimal gland sections were evaluated for the presence of murine cytomegalovirus, the virus most likely to infect the salivary and lacrimal glands. Histology of

248 Investigative Ophthalmology 8c Visual Science, January 1996, Vol. 37, No. 1 FIGURE l. Lacrimal gland histology of MRL/lpr mice. (A, top) Saline-treated mouse, showing typical well-demarcated multifocal inflammatory cell infiltrate. (B, bottom) Anti-CD4 mab-treated mouse, showing ill-defined infiltrate with diffuse extension into the parenchyma Hematoxylin and eosin; magnification, X160. FIGURE 2. Immunohistology of anti-cd4 mab-treated MRL/ lpr mice. (A, top) Staining for CD4+ cells, showing relative paucity of CD4+ T cells. (B, bottom) Staining for CD8+ cells, showing predominance of CD8+ T cells. Magnification, X250. the lacrimal glands showed no evidence of cytomegalic inclusions, and both culture and polymerase chain reaction were negative for cytomegalovirus. Hence, there was no evidence that the lacrimal gland disease in the anti-cd4 mab-treated mice was caused by cytomegalovirus infection. B cells (up to 28% of the total) at 18 months of age in MRL/+ mice. Double-negative T cells, if present in the lacrimal gland lesions of MRL/lpr mice, constitute only a small percentage of the infiltrate.2'3 The target organ lesions in the eye and kidney in MRL/lpr mice are, like lacrimal gland lesions, composed largely of CD4+ T cells. 23 We have reported 5 ' 6 that anti-cd4 mab treatment reduces the percentage of CD4+ T cells in the spleen by 97% and largely eliminates ocular and renal disease and autoantibody formation in MRL/lpr mice. Therefore, we initially hypothesized that such treatment would eliminate lacrimal gland lesions. However, the response seen was unanticipated. Instead of improving the lacrimal gland lesions, anti-cd4 mab that treatment resulted in persistent inflammation with a different morphologic picture than seen in controls. In addition, the DISCUSSION. Lacrimal gland inflammatory lesions are characteristic of several autoimmune mouse strains, including MRL/lpr mice, congenic MRL/ + mice, and (NZBxNZW) Fl hybrid (NZB/W) mice. The lacrimal gland lesions in MRL/lpr and MRL/ + are composed largely of CD4+ T cells, although there is a greater percentage of CD8+ T cells in MRL/ + mice than in MRL/lpr mice.2 In both MRL/lpr and MRL/+ mice, B cells constitute a small percentage of the infiltrate, although there is a late accumulation of

Reports 249 TABLE i. Immunohistology of Anti-CD4 Monoclonal Antibody-Treated and Control MRL/lpr Mice Group Number of Animals Thy-1.2 Mean ± SD (median) CD4 % Cells Staining Positive for: CD8 Pan-B Saline Ratlg Controls Anti-CD4 P value* 5 5 10 9 87 ± 3 (87) 92 ± 2 (92) 90 ± 4 (90) 87 ± 3 (87) 0.11 60 ± 10 (59) 51 ± 11 (48) 55 ± 11 (51) 11 ± 9 (8) 0.0003 21 ± 4 (19) 18 ± 7(16) 19 ± 5 (18) 63 ± 11 (66) 0.0003 15 ± 11 (12) 15 ± 13 (9) 15 ± 11 (10) 4 ± 6(1) 0.0007 SD = standard deviation; Ig = immunoglobulin; Controls = saline-treated and rat Ig-treated groups combined. * P value determined by Wilcoxon rank-sum test for control group vs. anti-cd4 monoclonal antibody-treated group. immunocytochemical profile in the lacrimal glands was altered markedly by anti-cd4 mab treatment. CD4+ T cells largely were absent, and the infiltrate was composed largely of CD8+ T cells. Although immunocytochemistry evaluates the percentage of cells staining positively and not the absolute number of cells per unit of tissue, the fact that the overall infiltrate was not diminished suggests that there was an absolute increase in the number of CD8+ T cells in the lacrimal glands of anti-cd4 mab-treated mice compared to control mice. The explanation for the paradoxic response of the lacrimal gland inflammation after anti-cd4 mab treatment remains uncertain. There was no evidence of a viral infection that might have been worsened by immunosuppression after anti-cd4 mab-treatment. However, the striking increase in CD8+ T cells in anti- CD4 mab-treated mice indicates that CD8+ effector cells can be generated without CD4+ helper T cell participation, a result similar to that seen after the genetic deletion of CD4 by Rahemtulla et al. 9 These investigators demonstrated that CD8+ T cells were increased in mice genetically incapable of making CD4+ T cells. Because CD8+ T cells do not require CD4+ T cells for normal development, our data suggest that the lacrimal gland disease intrinsic to the MRL/Mp strain is mediated by both CD4+ and CD8+ T cells and that one effect of the Ipr mutation is a shift to a more aggressive CD4+ T cell-mediated autoimmune disease as seen in MRL/lpr mice. Elimination of CD4+ T cells by anti-cd4 mab treatment does not eliminate lacrimal gland disease but does permit the CD8+ T cell response to predominate. Because ocular and vasculitic lesions are seen primarily in MRL/lpr mice and not in MRL/+ mice 1 ' 5 ' 6 and because these lesions are mediated primarily by CD4+ T cells, they are suppressed by anti-cd4 mab therapy. A corresponding Sjogren's-like clinical syndrome exists in humans. Patients infected with human immune deficiency virus (HIV) have an absolute loss of CD4+ T cells as a consequence of HIV-mediated damage to this T-cell subset. A syndrome known as diffuse infiltrative lymphocytosis syndrome may develop in these patients; lacrimal gland and salivary gland infiltration are primary components of the disorder. 10 A clinical syndrome similar to Sjogren's syndrome, including dry eyes and dry mouth, develops in patients with diffuse infiltrative lymphocytosis syndrome, but histologic evaluation of minor salivary gland biopsies in these patients has demonstrated that they are largely composed of CD8+ T cells, not CD4+ T cells. Conversely, minor salivary gland biopsies from patients with Sjogren's syndrome have revealed that the lesions are composed largely of CD4+ T cells. Hence, patients with diffuse infiltrative lymphocytosis, in whom CD4+ T cells are depleted, contract Sjogren's-like syndrome with CD8+ T-cell mediated disease, a situation analogous to that seen in our anti- CD4 mab-treated MRL/lpr mice. KeyWords autoimmune response, immunopathology, lacrimal gland, monoclonal antibody therapy, Sjogren's syndrome References 1. Theofilopoulos AN, Dixon FJ. Murine models of systemic lupus erythematosus. Adv Immunol. 1985; 37: 269-390. 2. Jabs DA, Prendergast RA. Murine models of Sjogren's syndrome: Immunohistologic analysis of different strains. Invest Ophthalmol Vis Sri. 1988; 29:1437-1443. 3. Jabs DA, Prendergast RA. Reactive lymphocytes in lacrimal gland and renal vasculitic lesions of autoimmune MRL/lpr mice express L3T4. / Exp Med. 1987; 166:1198-1203. 4. Watanabe-Fukunaga R, Brannan CI, Copeland NG, Jenkins A, Nagata S. Lymphoproliferative disorder in mice explained by defects in Fas antigen that mediates apoptosis. Nature. 1992; 314-317. 5. Jabs DA, Prendergast RA. Autoimmune ocular disease in MRL/Mp-//>r//jfrrmice is suppressed by anti-cd4 antibody. Invest Ophthalmol Vis Sri. 1991;32:27l8-2722. 6. Jabs DA, Burek CL, Hu Q, Kuppers RC, Lee B, Prendergast RA. Anti-CD4 monoclonal antibody therapy suppresses autoimmune disease in MKL/Mp-lpr/lpr mice. Cell Immunol. 1992; 141:496-507.

250 Investigative Ophthalmology & Visual Science, January 1996, Vol. 37, No. 1 7. Dialynas DP, Quan ZS, Wall KA, et al. Characterization of murine T cell surface molecule, designated L3T4, identified by monoclonal antibody GK-1.5: Similarity of L3T4 to the human Leu-3/T4 molecule. JImmunol. 1983; 131:2445-2451. 8. Henry SC, Hamilton JD. Detection of murine cytomegalovirus immediate early 1 transcripts in the spleens of latently infected mice. / Infect Dis. 1993; 167:950-954. 9. Rahemtulla A, Fung-Leung WP, Schilham MW, et al. Normal development and function of CD8+ cells but markedly decreased helper cell activity in mice lacking CD4. Nature. 1991;353:180-184. 10. Itescu S, Brancato LJ, Buxbaum J, et al. A diffuse infiltrative CD8 lymphocytosis syndrome in human immunodeficiency virus (HIV) infection: A host immune response associated with HLA-DR5. Ann Intern-Med. Genetic Predisposition to Coronavirus-Induced Retinal Disease Yun Wang,* Miguel Burnier,\ Barbara Detrick,% and John J. Hooks* Purpose. Retinal inflammatory and degenerative processes in humans and animals frequently are associated with genetic factors. The murine coronavirus, mouse hepatitis virus (MHV), JHM strain, induces a biphasic retinal disease in adult BALB/c mice. The genetic constitution of the host and the virus serotype can be critical factors in determining the outcome of a virus infection. The purpose of this study was to evaluate the possible role of host genetics in murine coronavirus-induced retinal disease. Methods. JHM virus was inoculated by the intravitreal route into BALB/c, CD-I, and A/J mice. At varying times after inoculation, eyetissueswere evaluated histologically. Antibody responses to the virus were evaluated by neutralization assays. Results. JHM virus induces a biphasic retinal disease in BALB/c mice. In the early phase, 1 to 7 days after inoculation, retinal vasculitis is observed. The second phase, characterized by retinal degeneration in the absence of inflammation, is seen by day 10 and progresses for several months. There is a similar biphasic disease process in JHM virus-infected A/J mice. However, retinal changes are less severe than those seen in BALB/c mice. Retinal tissue damage induced by JHM virus in CD-I mice is different. Only the early phase of the disease, consisting of retinal vasculitis, was observed. These CD-I mice do not develop the retinal degenerative disease. In fact, after day 10, the retina has a normal appearance. These differences in retinal tissue damage are seen over a wide range of infectivity of the virus From the * Immunology and Virology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD; the ^Department of Ophthalmology, McCill University, Montreal, Quebec, Canada; and the XDepartment of Pathology, The George Washington University Medical Center, Washington, DC. Submitted for publication February 14, 1995; revised fuly 28, 1995; accepted September 5, 1995. Proprietary interest category: N. Reprint requests: John J. Hooks, National Eye Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 6N228, Bethesda, MD 20892. inocula. Virus concentrations ranging from 10 1 ' 4 to 10 4 ' 4 TCID 50 /5 (A were capable of inducing both inflammation and degeneration in BALB/c mice, whereas, the highest concentration of virus (10 44 TCID 50 /5 //I) in CD-I mice resulted in only the early inflammatory changes. Conclusions. The authors show that the genetics of the host can profoundly affect the nature of retinal tissue damage. These studies substantiate the concept that a virus can indeed trigger retinal degenerative processes in genetically susceptible hosts. Invest Ophthalmol Vis Sci. 1996; 37:250-254. L-toronaviruses induce a number of intriguing pathologic processes. The mouse hepatitis virus (MHV), JHM strain, induces central nervous system disease characterized by acute infections and chronic demyelinating diseases. JHM virus also can induce a biphasic retinal disease in BALB/c mice. 1 ' 2 In the early phase, 1 to 7 days after inoculation, mild retinal vasculitis is observed. This initial phase is associated with the presence of viral proteins and the detection of infectious virus within the retina. The second stage in seen by day 10 and progresses for several months. This stage is characterized by a retinal degeneration in the absence of vasculitis or inflammation. This degenerative process is associated with a reduction of the photoreceptor layer, loss of interphotoreceptor binding protein, abnormality in the retinal pigment epithelium, and retinal detachment. In this later phase, infectious virus and viral proteins are not detected. However, autoantibodies directed against the retinal pigment epithelial (RPE) cell and the retina are observed. 3 Murine coronavirus infections can be modified or regulated by various factors, such as virus serotype, genetic constitution of the host, age or developmental stage of the host, and route of inoculation. Genetic factors of the host can be a critical factor in determining the outcome of a viral infection. 4 In fact, the genetic background can influence the type and intensity of the host immune response, the presence of cell membrane surface molecules used for viral adsorption