DNA AND CELL BIOLOGY Volume 35, Number 3, 2016 ª Mary Ann Liebert, Inc. Pp. 1–9 DOI: 10.1089/dna.2016.3232

REVIEW ARTICLE

The Anatomy of a Career in Science Michael B.A. Oldstone

E

twenties, who had a fever, stiff neck, and fluctuated between stupor and coma. Stupor, as I learned, was a level of unconsciousness in which the patient was left until becoming conscious upon stimulation by pain. In contrast, coma was the next level down, indeed, the level of unconsciousness before death when a patient failed to respond to pain. Woodward informed the three of us, all unsophisticated and uneducated in medical practice or therapeutics, that if we did not make the correct diagnosis and begin the appropriate therapy, the patient would be dead by morning. The resident medical officer did a spinal tap on the patient, withdrew an aliquot of the purulent material from the spinal space, placed it sterilely in a test tube, and gave it to Woodward. He then marched the three of us to a laboratory adjacent to ward 3B, which was set up for students and house officers to test patients’ samples. Following Woodward’s directions, we first readied part of the collected spinal fluid for culture and, second, smeared what remained on glass slides for a Gram stain. As we then examined the specimen under a microscope, we were instructed that we were looking at Grampositive pneumococci. Woodward then lectured the three of us about Pasteur, van Leeuwenhoek, Ehrlich, and Koch, all of whom were among the swashbuckling adventurers I read about earlier in Microbe Hunters. This was the first, but not the last tutorial, I received from Woodward. The next afternoon the three of us and Woodward returned to ward 3B and observed the patient sitting up in bed and talking. His initial diagnosis was pneumococcal meningitis, confirmed by culture and treated with the antibacterial drug chloramphenicol. For me, this sequence of clinical observations, laboratory confirmation, and good therapeutic result resembled a miracle, the patient, a Lazarus returning from the dead. I was now hooked as a lifer in the field of infectious disease and pathogenesis. However, why was I selected by Woodward for this tutorial? The opportunity turned out to be totally serendipitous. Only one of the students was actually so anointed because of his family’s professional connections. He had previously flunked out of medical school and spent 2 years in the military (he had been drafted as was mandatory then). After completing his military commitment, he was readmitted to medical school on a trial basis. Because his father was a faculty member at the medical school, Woodward felt obliged to take a personal interest in tutoring him. However, to make that selection seem unbiased, Woodward chose two other students whose last names began with the letter ‘‘O,’’

arly in medical school and postgraduate training, I became fascinated with the history and intellectual advances of immunology and virology. I continued in postdoctoral work at the bench to delve more deeply into both disciplines. This article examines the dialogue between immunology and virology that I explored and encompasses my life’s work. Eventual melding of both fields led to the modern discipline of viral pathogenesis, which I, along with the late Bernard Fields, was involved in developing and championing. The central challenge of viral pathogenesis in the latter half of the last century and now, in the 21st century, is understanding the molecular and genetic entities that enable viruses to cause disease and how individual hosts and populations resist and control viral infections. Respectively, these issues refer to virus virulence versus host resistance. This reminiscence is a traveler’s story of how, from an early age, then through medical and graduate school and during a long career, I journeyed along the road of discovering where viral pathogenesis would lead, from the start of that adventure up to the present. As an only child in a very caring family living in New York City, I was often immersed in books and given abundant time for reading. My tastes were in adventure stories. My favorite menu included Percival Christopher Wren’s Beau Geste and the other two volumes of that trilogy, Arthur Conan Doyle’s Sherlock Holmes, as well as tales of the navigators and explorers of the 14th through 19th centuries. However, all those tales played second fiddle to the incredible adventures and conquests of scientists described in Paul de Kruif’s Microbe Hunters. By the tender age of 12 or so, I knew what I wanted to be; it was a career Microbe Hunter. After college where I majored in English and History, with exposure to chemistry, biology, and genetics, I entered medical school. On my third day there (the University of Maryland, Baltimore), while beginning the study of tissues, nerves, and blood vessels in a cadaver, I was summoned to medical ward 3B at the University Hospital to join Theodore Woodward, the professor and Chairman of Medicine. I had never met Dr. Woodward, but that summons was nothing less than a command. I had no idea if I had done anything wrong or why I was selected for this meeting. At that time in the late 1950s, the medical wards at University Hospital were segregated and in 3B resided African-American males. Two other freshman classmates whom I now met for the first time joined me as I arrived at ward 3B late in the afternoon. The patient we saw with Dr. Woodward was a muscular male in his mid-

Viral-Immunobiology Laboratory, The Scripps Research Institute, La Jolla, California.

1

2

also, the first letter of that wayward student’s last name. This justified the choice of Oldstone and my alphabetical partner whose names flanked that of the readmitted student. Woodward then followed up our personal education by taking the three of us on medical rounds with him continuously on Saturday afternoons throughout our first year in medical school. Medical school classes routinely ran from Monday through Saturday morning with no assigned class responsibilities on Saturday afternoons. Woodward was a superb clinician and a wonderful teacher specializing in infectious diseases. Over that year, I saw many patients hospitalized for a wide array of bacterial and viral infections. As a result, I became engaged, motivated, and passionate about the subject of infectious disease. In contrast, and I will not mention my other two classmates by name, the son of the faculty member was a poor student and despite Woodward’s tutorials, flunked out of school. The other student had limited interest in infectious diseases, desired to be a surgeon, and resented having to give up his formerly free Saturday afternoons. During the Second World War, Woodward worked closely with Joseph Smadel, one of the deans of American virology and rickettsiology. Smadel, then head of virology and virologic research at Walter Reed Army Medical Hospital, arranged as Woodward had requested for me to spend the summer after my first year of medical school working in his laboratory with him, Betsie Jackson, and Hank Fuller. At that time, and through Smadel, I was introduced to John Enders and Carlton Gajdusek, both famous virologists and Nobel Laureates. During my second year in medical school, Woodward situated me in the laboratory of Sheldon Greisman, where I learned about the biochemistry and biology of endotoxin and endotoxin shock. My laboratory experience with Smadel and Greisman led me toward a PhD degree program. With classmate and good friend, Bill Wood, I became one of the first two medical students at Maryland to attempt an MD/PhD program. At that time, no formal MD/ PhD program was in place, so Bill and I took a formal leave of absence from the medical school. My graduate training then continued at the Homewood campus of Johns Hopkins in the Department of Biochemistry working on fireflies and luciferase, and in the Microbiology Department at Maryland researching toxins from rickettsia. My purpose and drive to enter a basic science environment came from a desire to understand the mechanism(s) of infectious diseases I had seen, but about which neither I nor, indeed, the clinical medical faculty possessed much knowledge. That year and a half in the PhD program solidified my choice to make my life’s work basic biomedical research, so I returned to first obtain my MD. Based on experiments I published during work with Greisman on endotoxin (Oldstone, 1959), I was offered a position by Jacob Fine at Beth Israel and Harvard and spent my third summer and a few additional months working on endotoxin in his laboratory. Fine was one of the world’s experts on endotoxin shock. Sheldon Greisman’s influence on me far exceeded the scientific arena. His wife Janet had roomed with my future wife, Elizabeth (Betsy) Hoster, in college. The Greismans introduced us, and 55 years later we are still happily married, having produced three wonderful children, Jennifer, Beau, and Chris, and six granddaughters, Caroline, Aileen, Madeleine, Faye, Raina, and Marilee. While in Boston and through Fine, I again met Enders who was to play an im-

OLDSTONE

portant role in the early phase of my career. After finishing medical school and residency, I talked to Enders about future training. He told me that if he was looking for the best place to do postdoctoral work in a research laboratory, at this time in 1966, he would seek a position at the interface of virology and immunology and recommended I train with Frank Dixon in La Jolla, California. I recall him telling me that, since I knew how to plaque vesicular stomatitis virus and grow it in culture, I knew sufficient virology to get started. Now, according to Enders, was the appropriate time to learn immunology and use it to study viruses for a future research career. Frank Dixon was a major figure in immunobiology and immunopathology and the founder of Scripps Clinic and Research Foundation (now The Scripps Research Institute). Because I had experience working with viruses, Dixon accepted my application and suggested I investigate the reported chronic kidney disease (Hotchin, 1962) associated with lymphocytic choriomeningitis virus (LCMV). To learn how to handle LCMV, I went with Dixon’s referral for a short tutorial with Wally Rowe at the NIH. Rowe worked initially with LCMV and immunity to the virus (Rowe, 1954) before his stellar studies of retroviruses. I joined the Dixon group near the end of 1966. His laboratory was filled with eight energetic and flashy postdoctoral fellows. Sparks flew everywhere. My contemporary postdoctoral fellows were truly amazing, considering that, five of the six Americans, including me, were later elected to the National Academy of Sciences, The National Academy of Medicine, or both (five of the six), and one received the Lasker Award. Furthermore, the faculty recruited by Dixon was remarkable, carefully selected, and small in number: Hans Mu¨ller-Eberhard, whose interest was protein chemistry became involved in dissecting the complement effector system; Bill Weigle studied B- and T-cell tolerance and autoimmune diseases; and Charlie Cochrane’s interest was effector protein and peptide release by inflammatory cells. Three years later, Karl Habel, an exceptional senior virologist and former head of the Virology Laboratory at the NIH, was recruited to Scripps, and I spent time in his laboratory. He influenced and polished my knowledge and approaches in virology. However, it was also Dixon’s constant stream of expert visitors, who he made sure spent oneon-one time with me and his other postdoctoral fellows. Those experiences were incredible and left an indelible impression. I had the opportunity to discuss my scientific program, plans, and results, while receiving advice from seminal figures in immunology and virology. These included Macfarlane Burnet, Gus Nossal, Baruj Benacerraf, John Humphrey, Henry Kunkel, Jim Gowens, Herman Eisen, Byron Waksman, Hilary Koprowski, Carlton Gajdusek, Jacques Monod, Renato Dulbecco, Astrid Fagraeus, George Klein, Frank Fenner, Tom Weller, Harry Ginsburg, Ed Kilbourne, Wally Rowe, and Purnell Choppin. What a bonus for a young fellow growing up in science! From the beginning, I focused my research on what I perceived as important questions in biology and intellectual issues to address rather than concentrating on one infectious agent. As such, I explored many aspects of immunologic tolerance, autoimmunity, immunosuppression, and acute and persistent infections. Even so, I had and continue to pursue a long-term attachment and relationship to LCMV and its

THE ANATOMY OF A CAREER IN SCIENCE

murine model. Because LCMV itself is not cytotoxic, the injury and disease following its infection is primarily a reflection of an immune response against the virus. As for LCMV itself, we cloned, characterized, and sequenced it, established its genomic order, discerned its profile by electron microscopy, identified and located its cellular receptor, and determined how the receptor functions (reviewed in Oldstone, 2002; Oldstone and Campbell, 2011). In addition to our work using LCMV, I utilized cytomegalovirus to ask how virus was activated and what cells became involved during transplantation rejection (Olding et al., 1975; Rice et al., 1984; Schrier et al., 1985). Measles virus was studied to address its ability to cause immunosuppression and how, in a subset of patients, measles virus caused a chronic progressive lethal disease of the central nervous system (CNS), so-called subacute sclerosing panencephalitis (SSPE) (reviewed in Naniche and Oldstone, 2000; Oldstone, 2009). More recently, I became interested in and analyzed how influenza virus causes injury and disease, especially the role played by cytokine storm and interferon (IFN), that is, role of immunopathology in this virus infection (Marsolais et al., 2009; Teijaro et al., 2011; Walsh et al., 2011). One of the rewards of this investigation was a close collaboration with Hugh Rosen, a fellow professor and superb scientist also at Scripps. Most recently, I have wondered why natural selection and evolution have yielded 14 type 1 IFNs that signal through the same heterodimeric receptor; that question prompted the mapping of precise roles for IFN type 1a and b (Teijaro et al., 2013; Ng et al., 2015). My early work investigating LCMV and associated kidney involvement provided the seminal observation that chronic viral infection initiated early in life can induce an antibody response resulting in virus-antibody immune complexes (VAbIC) and V-AbIC-mediated disease (Oldstone and Dixon, 1967). This was a paradigm-shifting observation at that time since the work of Ray Owens and Macfarlane Burnet led to the common belief that neonatal exposure to a virus would result in immunologic tolerance and no antibody would be produced against the virus. To the contrary, our findings not only addressed the fundamental immunologic question of tolerance but also demonstrated the pathogenic potential of VAbICs that could deposit in tissues and cause arteritis, choroiditis, and glomerulonephritis (reviewed in Oldstone, 1975). Interestingly, a few weeks after the publication of our article in Science (Oldstone and Dixon, 1967), I received a letter from Enders saying I was on the right track. Now, studies begun with LCMV were expanded to murine models to show there was no immunologic tolerance in chronic retroviral infection (Oldstone et al., 1972, 1976). A few years after, we reported that B-cell tolerance was not caused by infection initiated in utero or at birth; Rafi Ahmed, a former postdoctoral fellow, who advanced from my laboratory to an initial faculty position at UCLA, documented that the absence of virus-specific T cells in adult mice persistently infected with LCMV, and importantly that infection either in utero or at birth did not result in clonal deletion of LCMV-specific T cells ( Jamieson and Ahmed, 1988). At the time of my initial studies with LCMV and VAbIC, I met Abner Notkins, a young NIH scientist who was also investigating V-AbICs, while working with lactic dehydrogenase virus (Notkins et al., 1966). Abner and I became and continue to be lifelong friends and together initiated a viral pathogenesis–viral immunity for an annual one-evening workshop at the

3

Federation meetings then held routinely in Atlantic City, New Jersey. Our meetings soon attracted a large cadre of senior, intermediate, and beginning virologists and immunologists. It was at one of those meetings that we came in contact with Bernie Fields. Bernie’s interest and mine focused on studies in viral pathogenesis and, as a result of work from students we both trained and others who were attracted, the field of viral pathogenesis developed. From Bernie’s laboratory and mine, a foundation developed upon which the current field of viral immunity and viral pathogenesis grew. It was in this milieu that a new and larger set of colleagues from virology and immunology joined together. Unfortunately, Bernie, my great friend, colleague, and supporter, had his life cut short by pancreatic cancer at the young age of 52. However, his trainees continue to do elegant studies in both these fields, thus keeping his memory alive. Our early analysis of the pathogenesis of chronic virusinduced disease led to a new paradigm of disease. That is, the continuous replication of a noncytolytic virus in differentiated cells in vitro and in vivo produced viruses (foreign genes) that could alter the differentiation function of cells without killing them (Oldstone, 1989, 1993; de la Torre et al., 1991; Oldstone, 2002). This was a dramatic departure from the existing dogma that viral diseases reflected the destruction of infected cells. The importance was the realization that persisting virus infection could cause a biochemical alteration in cells whose appearance remained anatomically normal. The initial observations were in vitro with alterations of acetylcholine enzymes in neuroblastoma cells persistently infected with LCMV (Oldstone et al., 1977) and LCMV infection in vivo of growth hormone (GH)-producing cells located in the anterior pituitary gland (Oldstone et al., 1982). The result of GH cell infection was a twofold to fivefold reduction in GH transcription, a 50% reduction in GH protein levels, and, consequently, aborted growth of the infected host (Oldstone et al., 1982, 1984; Oldstone, 1989; Oldstone 1993). Growth was restored by adoptive transfer of GH or uninfected cells that made GH. In contrast, however, even when GH production was diminished, there were no effects on another pituitary hormone, prolactin. Interestingly, infection in a culture of cloned pituitary cells that made both GH and prolactin reduced only GH transcription, but not prolactin transcription (de la Torre and Oldstone, 1992). Making and mapping chimeras with deletions in GH and prolactin promoters localized the defect in the GH promoter to the Pit-1 transcription factor. Binding of the COO- terminal part of the LCMV nucleoprotein was involved. These observations with GH were rapidly extended to other endocrine systems, that is, to b cells in the islets of Langerhans with low insulin and abnormal glucose tolerance (Oldstone et al., 1984); the thyroid gland and low thyroglobulin output (Klavinskis and Oldstone, 1987; Klavinskis et al., 1988); and virusinfected neurons in the brain that displayed a loss of GAP43 protein both in vitro and in vivo (Oldstone, 1989, 1993; de la Torre et al., 1996). The loss of GAP43 in vivo is associated with learning and cognitive defects. GAP43 is involved in synaptic remodeling, memory, and learning. Similarly, as shown by Ian Lipkin during his postdoctoral fellowship, content of another neuronal transmitter, like somatostatin mRNA, was decreased (Lipkin et al., 1988). In contrast, amounts of other neuronal proteins such as amyloid precursor protein,

4

C-fos, and cholecystokinin were not diminished (reviewed in Oldstone, 1989; Oldstone, 1993, 2002). These observations led us to explore why so many observers reported that a wide array of RNA and DNA viruses, including lytic viruses, could persistently infect neurons that escaped immune destruction. That is, why would viruses preferentially select neurons for residency instead of other CNS cell types. Since virus-specific cytotoxic CD8 T cells (CTLs) were responsible for the recognition and clearance of virus-infected cells elsewhere in the body, we wondered if neurons possibly possessed a defect in their ability to present viral antigen (peptides) in the context of major histocompatibility complex (MHC) molecules to CTLs or if CTLs were restricted from entering the CNS, that is, did not cross the blood–brain barrier and migrate into and through the brain. We reasoned that, since neuronal cells for the most part are not replaceable, but are essential for life of the organism, there must be a strong selection process to maintain their survival. Furthermore, we deduced that viruses might/ likely take advantage of this selection process to find a safe haven for hiding without being confronted by the host’s immune system. Our initial studies revealed that virus-specific CD8 and CD4 T cells or, indeed, any activated T cell easily crossed the blood–brain barrier, thereby leaving the systemic circulation to enter the brain. Kinetic calculations indicated that, if there were no specific infectious material in the brain for the T cell to recognize, T cells exited generally within 5–10 days. However, if there were recognizable antigens, the transit time increased to 21–40 days for most activated T cells with a substantial population staying behind longer. Yet, neurons infected in vivo and expressing specific viral proteins were not immunologically damaged by corresponding virus-specific CTLs (Oldstone et al., 1986). This observation led postdoctoral fellows Etienne Joly and Lennart Mucke to evaluate MHC-viral antigen (peptide) presentation of neurons to virus-specific T cells in both cultured neuronal cell line and in the whole animal ( Joly et al., 1991; Mucke and Oldstone, 1992). They found that 5 · 106 or more (up to 3 · 107) LCMV-specific CTLs did not lyse infected neurons in vitro or cause immunopathologic injury of neurons in persistently infected mice following adoptive transfer. However, in contrast, intracerebral injection of adults with as few as 100 PFU of virus caused immunopathologicmediated disease and death in animals with acute LCMV infection. The difference was that, in acute infection, virus replicated in cells of the leptomeninges and choroid plexus, but not neurons. In the persistent infection, however, in contrast, virus was present in neurons. Analysis indicated that neurons had few to barely detectable MHC molecules on their surfaces compared to leptomeningeal and choroid plexus cells, which express high levels of MHC molecules on their surfaces (Mucke and Oldstone, 1992). Studies of neuronal cells in culture disclosed a number of significant defects in MHC-antigen presentation, including little to almost no MHC-heavy chain transcription, whereas light chain b2-microglobulin mRNA and protein were made normally ( Joly et al., 1991). Furthermore, neurons had even less mRNA of the endosomal peptide transporters HAM1 and HAM2, which are needed to bring viral peptides to the MHC groove ( Joly and Oldstone, 1992). Transfection with a fusion gene encoding a different and functional MHC class I mol-

OLDSTONE

ecule into these neuronal cells subsequently allowed virusspecific CTLs to function by recognizing the new MHC class I molecules that contained appropriate viral peptide and could, then, lyse the neuronal cells in vitro ( Joly et al., 1991). This observation was made in the early 1990s, before the fall of the Berlin Wall. Because of that timing, Etienne, who came from the Pasteur Institute and was recommended by Jacob to join our laboratory, titled our report for Science describing why infected neurons were not lysed as ‘‘Better Red than Dead.’’ As humorous as that title was, I felt obliged to change it to ‘‘Viral persistence in neurons explained by lack of major histocompatibility class I expression’’ ( Joly et al., 1991). Another postdoctoral fellow, Glenn Rall, with Lennart Mucke, extended these findings to a novel in vivo model using transgenic technology to insert a functioning MHC molecule into neurons of mice (Rall et al., 1995). Although expression of the new MHC class I molecule was now detected in neurons, and neurons were infectable with virus, virus-specific CTLs still could not lyse such ‘‘healthy’’ neurons. However, when neurons from these transgenic mice were isolated from the CNS and brought to over 95% purity by using a panning technique in which Petri plates used for culturing were coated with antibody specific to MHC molecules expressed on the surfaces of those neurons, the neuronal cells could now be lysed. However, CNS neurons are not lysed by CTLs in vivo unless they are severely damaged (lose electrical activity). ‘‘Healthy’’ neurons, as defined by retention of electrical activity, are protected from immunologicmediated injury. Thus, the CNS milieu is important since healthy neurons removed from the CNS are amenable to lysis by CTLs in vitro (Rall et al., 1995). These observations speak to the presence of multiple selective host factors that preserve neurons from attack by killer cells likely for the purpose of selecting and preserving these cells, which are irreplaceable and essential for host survival. Earlier, my laboratory team was engaged in studies of fluidity and movement of host and viral molecules in cell surface membranes (Lampert et al., 1975; Oldstone et al., 1980, 1983). Having discussions with John Singer and John Holland down the road from TSRI at nearby UCSD helped guide this work. Studies we did with George Klein using Epstein-Barr virus (EBV) tumor cell lines collected in Uganda and specific reagents made at Scripps in Hans Mu¨ller-Eberhard’s laboratory indicated that the third component of complement (C3) migrated in the cells’ membranes in tandem with EBV surface molecules. These results were the first to suggest that C3 was the EBV receptor ( Jondal et al., 1976; Yefenof et al., 1976). Later, experiments done in the laboratories of Strominger and Fearon and of Nemerow and Cooper definitively proved that the C3 molecule was the receptor for EBV (Fingeroth et al., 1984; Nemerow et al., 1985). We went on to identify, utilize, and study the receptors for both measles virus (Dorig et al., 1993; Manchester et al., 1994, 1997) and LCMV. Mari Manchester in our laboratory, working with John Atkinson at Washington University, St. Louis, and the independent research of Richardson in Canada showed that human CD46, a complement regulatory protein, was the actual receptor for Edmonston strain measles virus (Manchester et al., 1994). Measles is a human pathogen and does not naturally infect mice, although it can be adapted by forced passages to infect rodent hosts. To obtain a small mouse model in which the Edmonston strain measles virus, itself, or mutations and gene swaps of the virus

THE ANATOMY OF A CAREER IN SCIENCE

could be studied in vivo, while manipulating the known genetics and immunologic profiles of the mouse, we (Rall et al., 1997; Oldstone et al., 1999) and others utilized the CD46 receptor with cell-specific transgenes to engineer mice that expressed the CD46 measles virus receptor in cells of the immune or CNS systems. By such engineering and manipulation of the host and virus, a more careful and thorough dissection became possible, enabling us to better understand aspects of measles virus pathogenesis than possible when studying humans. In the 1990s and early 2000s, despite an effective and efficient vaccine, measles virus was still infecting millions of humans and killing slightly less than a million individuals each year, primarily by suppressing the immune system and affecting the CNS. To study the measles-induced persistent and fatal CNS disease, SSPE, John Patterson applied reverse genetics to swap the biased hypermutated measles virus M gene obtained from a patient with SSPE as a replacement for the normal matrix (M) gene or in other studies disabled C and V genes of the measles virus genome (Patterson et al., 2000, 2001). We found that inclusion of the hypermutated M protein from the SSPE patient actively contributed to the length of time the resulting infection lasted; thus, the hallmark M protein mutations were not passive events in the pathogenesis of SSPE (Patterson et al., 2001). When the measles virus genome contained a biased hypermutated M gene, the virus was infectious in vivo and prolonged the survival of its murine host by 50–60 days over CNS infection by the native virus that contained a normal M gene. With nonmutated Edmonston measles virus in the transgenic mouse model, death occurred 2 weeks following inoculation. Other studies found that the V and C proteins of measles virus functioned as virulence factors in vivo (Patterson et al., 2000). Although wild-type measles, measles C-, or measles V- had growth and cytopathic effects in vitro similar to those of the native Edmonston measles virus, the mutant virus displayed major differences in vivo. Inoculation of 5 · 102 to 1 · 103 PFU of Edmonston strain measles infected all transgenic mice bearing the measles virus receptor resulting in illness and death by 2 weeks postinfection. In contrast, at least 2–3 logs more measles virus containing the C- or V- mutations were required to kill 50% of infected mice. Spread of the wild-type Edmonston and Edmonston C- was similar throughout the CNS, but in direct contrast, measles virus with a disordered V gene had a restricted spread (Patterson et al., 2000). Viral load and transcription were markedly reduced for V-, but not C-, throughout the CNS. Levels of multiple cytokines and chemokines were equivalent for V-, C-, and wild-type viruses. Importantly, infection with wild-type Edmonston strain measles virus in our transgenic CD46 model led to biased hypermutations of the viral genome, affecting primarily the M gene with the conversion of U to C and A to G bases, high titers of antibodies to measles virus, and infiltration of B and T cells into the CNS followed by death, thus recapitulating the SSPE disease of humans. The observation of predominant M gene-biased hypermutations was first described in human SSPE brains by Cattaneo and Billeter (Cattaneo et al., 1988). All these diagnostic markers just named define the progressive, fatal neurodegenerative disease SSPE attributable to measles virus infection of the CNS. To achieve a model of SSPE with prolonged CNS infection following native nonmutated measles virus infection, initially we had to immunosuppress transgenic mice bearing the measles virus receptor in their neurons, followed several days later with measles virus infection (Oldstone

5

et al., 2005). Thus two hits, transient suppression of the host’s immune system followed within a few days by virus infection, were required to cause SSPE. Postdoctoral fellow Simone Ward followed up this observation and, in collaboration with Chuck Samuel’s laboratory, showed that the RNA editing the enzyme adenosine deaminase is a restriction factor for controlling measles virus replication in vitro (Ward et al., 2011). Yusuke Yanagi, who earlier cloned the T-cell receptor in Tak Mak’s group, came to my laboratory as an eager postdoctoral fellow to learn virology. After returning to Japan, Yusuke isolated and described the ‘‘SLAM’’ receptor for circulating wild-type measles viruses that infect humans (Tatsuo et al., 2000). Yusuke has gone on to do brilliant work with the structure and fusion of measles virus hemagglutinin protein. In terms of the immune system and measles virus infection, our transgenic models proved valuable by showing that measles virus interacts with SLAM receptors on dendritic cells (DCs) to cause immunosuppression (Hahm et al., 2004, 2007). Bumsuk Hahm showed that the mechanism was inhibition of innate immunity caused by selective signaling through TLR4mediated suppression of IL-12 synthesis. Other studies revealed that CD4 T cells were an important factor in controlling primary measles virus infection of the CNS (Tishon et al., 2006). CD4 T-cell control is dependent on combined activity with either CTLs or B cells. Neither CD4, CD8, B cells alone, nor CTLs with B cells was effective. IFN-g also played a role in CNS clearance since the absence of an IFN-g response led to the persistence of virus infection. More recently and up to the present, we have focused on the molecular dissection of how viruses persist. To address this issue, we turned our attention back to LCMV. First, postdoctoral fellow Wei Cao, following up work by another postdoctoral fellow, Persephone Borrow, uncovered the information that alpha-dystroglycan (a-DG) is the receptor for LCMV, the other Old World arenavirus Lassa fever virus (LASV) and clade C New World arenaviruses (Cao et al., 1998). Second, working in collaboration with Kevin Campbell, postdoctoral fellow Stefan Kunz discovered that, for a-DG to act as a functional receptor, it first needed to react with the glycosyltransferase, LARGE (Kanagawa et al., 2004; Kunz et al., 2005). LARGE added two O-linked sugars and changed the conformation of a-DG allowing the receptor to transit to the cell’s surface (Yoshida-Moriguchi et al., 2010). Kunz mapped the binding site for LCMV and LASV to 17 amino acids. This 17-amino-acid area was also the binding site for extracellular matrix molecules (ECMs) like laminin and agrin. For LCMV or LASV to bind to its functional receptor, it must initially push ECMs away. This is accomplished as the viruses bind three- to five-times more strongly to the receptor than laminin and easily displace it (Kunz et al., 2001). Third, postdoctoral fellows Noemi Sevilla and Stefan Kunz documented that a-DG is preferentially located in the immune system on DCs (Sevilla et al., 2000). DCs contain over 98% of the total a-DG expressed in the immune system. Fourth, Sevilla and Kunz, joined by Dorian McGavern, Brian Sullivan, Andrew Lee, Dan Popkin, David Brooks, Elina Zuniga, and Cherie Ng studied aDG-mediated arenavirus infection of DCs (Sevilla et al., 2004; Zuniga et al., 2004, 2008; Brooks et al., 2006; Popkin et al., 2011; Sullivan et al., 2011; Ng and Oldstone, 2012; Lee et al., 2013). LCMV is basically a nonlytic virus and by a noncytolytic mechanism of continuous replication in DCs

6

impairs the ability of these cells to act as antigen-presenting cells. The result is an inability of DCs to arm and expand functional virus-specific T cells. Virus-specific T cells are an absolute requirement to clear an acute virus infection. The result was/is the development of a persistent infection. Fifth, Rafi Ahmed, during his postdoctoral fellowship, showed that LCMV variants isolated from lymphoid cells initiate immunosuppression in adult immunocompetent hosts (Ahmed et al., 1984; Ahmed and Oldstone, 1988). In contrast, virus variants isolated from neurons in the CNS ably generate robust antiviral T-cell responses that terminate an acute infection. Sevilla and Kunz then showed that immunosuppressive variants bind at 2–2.5 logs higher affinity to a-DG and infect greater numbers of DCs (30% to 50% of splenic total) than LCMV isolates from nonlymphoid tissues (brain) that by contrast infect less than 5% of DCs (Sevilla et al., 2000, 2004; Kunz et al., 2001; Sullivan et al., 2011). Sixth, Maria Salvato, Persephone Borrow, and Juan Carlos de la Torre cloned and sequenced one such immunosuppressive variant, LCMV Clone 13 (Salvato et al., 1991), originally isolated by Rafi Ahmed from its parental CTL-inducing virus LCMV ARM 53b. ARM 53b clones were initially isolated by Frank Dutko (Dutko and Oldstone, 1983). We subsequently utilized reverse genetics to document that, of the 3356 amino acids contained by Cl 13 and ARM 53b, remarkably only two amino acids determine whether viral-induced immunosuppression and persistence or alternatively a robust antiviral Tcell response and clearance of the acute infection occur (Sullivan et al., 2011; Lee et al., 2013). Of the two amino acid mutations, one is in the spike glycoprotein and responsible for virus binding to its receptor and entry into the cell (Cl 13/ ARM 53b GP aa 260 Leu/Phe) (Sullivan et al., 2011), whereas the second is in the viral polymerase (Cl 13/ARM 53b L aa 1079 Glu/Lys). This mutation in the polymerase (L gene) enhanced transcription and replication in DCs by 1.5–2 logs during Cl 13 infection compared to ARM 53b infection (Lee et al., 2013). Furthermore and importantly, Andrew Lee showed that the differences in virus replication and yield are restricted to DCs and not found in several other conventional laboratory cell lines (Lee et al., 2013). The reverse genetics system used to swap the differing amino acids between Cl 13 and ARM 53b was worked out and reported by de la Torre’s laboratory (Emonet et al., 2009). de la Torre is now a Professor at Scripps. Seventh, virus-specific epitopes recognized by virusspecific T cells were mapped in our laboratory primarily by J. Lindsay Whitton and several other individuals (Oldstone et al., 1988; Whitton et al., 1988, 1989; Klavinskis et al., 1990; Lewicki et al., 1995a, 1995b). Cl 13 and ARM 53b LCMV and the recognition T-cell epitopes are now the primary model used worldwide in multiple laboratories to study basic mechanisms, kinetics, and activities of acute virus infection and of viral persistence. Eighth, David Brooks (Brooks et al., 2006) in our laboratory and workers in those of Rafi Ahmed (Barber et al., 2006) and John Wherry (Wherry, 2011) found that immunosuppressive variants co-op the host’s negative regulators of the immune response (NIR), which are generated and function to keep the immune response in check, so it does not harm healthy tissues (i.e., immunopathology). By virus hijacking immunodominant NIR, IL-10, and PD-1/PD-L1, T cells become exhausted. This is a common phenotype for the persistent infections of mice and humans and in many or

OLDSTONE

likely all cancers. Recently, postdoctoral fellows John Teijaro, Cherie Ng, and Brian Sullivan (Teijaro et al., 2013) showed that signaling by the IFN type 1 receptor induced during the first 16–24 h of infection by the immunosuppressive LCMV variant Cl 13 is the master regulator of IL-10 and PD-1/PD-L1 expression: IL-10 and PD-1/PD-L1 being immunodominant NIRs. Furthermore, these investigators documented that IFN1b receptor signaling, not IFN-a, is the culprit (Ng et al., 2015). Blockade of IFN-b or of IFN type 1 ab receptors diminished NIR, maintained normal secondary lymphoid architecture, restored T-cell function, and reduced virus titers, thus controlling the persistent infection (Teijaro et al., 2013; Ng et al., 2015). Our findings uncovered by experimentation with immune-mediated virus injury, virus-induced cell injury, virus-induced immunosuppression, and persistence in the LCMV murine model have been extended to many other RNA and DNA viruses, as well as to infections of humans and other animals, and to some human cancers. Currently our laboratory’s work focuses on three interrelated areas. First, a current postdoctoral fellow, Brett Marro, is seeking by high-throughput screening the isolation of molecules that block NIRs and certain IFN type 1 pathways as potential therapeutic targets for persistent viral infections, cancers, and autoimmune disorders. Included in this project is learning why so many IFN-1 molecules have been selected and retained since all of them signal through the same heterodimeric type 1 IFN receptor. Our task is to dissect the individual and independent biologic functions of all 14 IFN-a and 1 IFN-b molecules. Second, we are extending our findings and knowledge derived from LCMV in a murine model to the other Old World arenavirus, LASV, and its infection in humans. This exploration of LASV pathogenesis is in collaboration with Brian Sullivan, Juan Carlos de la Torre, and Kristian Andersen at Scripps, Kevin Campbell at the University of Iowa, Pardis Sabeti at Broad Institute and Harvard, and Bob Garry at Tulane University, with a focus on the innate and adoptive T-cell responses made to LASV by infected individuals who survive or succumb to the infection. We are also analyzing the role played by mutations in LARGE. LARGE is a basic component of the functional a-DG receptor that LASV uses (Cao et al., 1998; Kunz et al., 2005). Mutations in LARGE are found in *20% of individuals residing in endemic LASV areas (Andersen et al., 2012). Third, we continue our assessment of immunopathologic injury and the role of cytokine storm in viral infections with Hugh Rosen and John Teijaro at Scripps (Marsolais et al., 2009; Teijaro et al., 2011; Walsh et al., 2011) and autoimmune diseases. To sail a good ship into such rough and uncharted waters requires both a superb complement of colleagues and a formidable crew. Throughout my four decades in science, I have found good friends and colleagues not only at my institution but also nationally and internationally. I must highlight that, in addition to our shared pleasure in speaking the language of science and experiments, I have bonded with several coworkers for their intellectual exchanges, love of opera, Broadway shows, and trout fishing. Furthermore, I have maintained my love of history and literature and have written a book Viruses, Plagues, & History as a follow-up to de Kruif’s Microbe Hunters read in my childhood. Viruses, Plagues, & History was favorably reviewed by the New York Times Sunday Book Review and has now been

THE ANATOMY OF A CAREER IN SCIENCE

translated into six languages. I have been blessed to have such hearty and resourceful crewmates (fellows) all along my science journey and a superb secretary, Gay Wilkins-Blade, to keep the ship on an even keel. As for fellows, starting with the earliest, Raymond Welsh, Michael Buchmeier, Lars Olding, and Bob Fujinami, who set the bar high for passion and fine experimental work, 73 other fellows and still counting have followed. I take much pleasure and pride in that, of these 77 fellow travelers, over 95% are engaged in full-time basic research. Also among them, many now run their own top-of-thehill laboratories that are turning out exceptional F2 progeny. Disclosure Statement

No competing financial interests exist. References

Ahmed, R., and Oldstone, M.B.A. (1988). Organ-specific selection of viral variants during chronic infection. J Exp Med 167, 1719–1724. Ahmed, R., Salmi, A., Butler, L.D., Chiller, J.M., and Oldstone, M.B.A. (1984). Selection of genetic variants of lymphocytic choriomeningitis virus in spleens of persistently infected mice. Role in suppression of cytotoxic T lymphocyte response and viral persistence. J Exp Med 160, 521–540. Andersen, K.G., Shylakhter, I., Tabrizi, S., Grossman, S.R., Happi, C.T., and Sabeti, P.C. (2012). Genome-wide scans provide evidence for positive selection of genes implicated in Lassa fever. Philos Trans R Soc Lond B Biol Sci 367, 868–877. Barber, D.L., Wherry, E.J., Masopust, D., Zhu, B., Allison, J.P., Sharpe, A.H., Freeman, G.J., and Ahmed, R. (2006). Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439, 682–687. Brooks, D.G., Trifilo, M.J., Edelmann, K.H., Teyton, L., McGavern, D.B., and Oldstone, M.B.A. (2006). Interleukin10 determines viral clearance or persistence in vivo. Nat Med 12, 1301–1309. Cao, W., Henry, M.D., Borrow, P., Yamada, H., Elder, J.H., Ravkov, E.V., Nichol, S.T., Compans, R.W., Campbell, K.P., and Oldstone, M.B.A. (1998). Identification of alphadystroglycan as a receptor for lymphocytic choriomeningitis virus and Lassa fever virus. Science 282, 2079–2081. Cattaneo, R., Schmid, A., Eschle, D., Baczko, K., ter Meulen, V., and Billeter, M.A. (1988). Biased hypermutation and other genetic changes in defective measles viruses in human brain infections. Cell 55, 255–265. de la Torre, J.C., Borrow, P., and Oldstone, M.B.A. (1991). Viral persistence and disease: cytopathology in the absence of cytolysis. Br Med Bull 47, 838–851. de la Torre, J.C., Mallory, M., Brot, M., Gold, L., Koob, G., Oldstone, M.B.A., and Masliah, E. (1996). Viral persistence in neurons alters synaptic plasticity and cognitive functions without destruction of brain cells. Virology 220, 508–515. de la Torre, J.C., and Oldstone, M.B.A. (1992). Selective disruption of growth hormone transcription machinery by viral infection. Proc Natl Acad Sci USA 89, 9939–9943. Dorig, R.E., Marcil, A., Chopra, A., and Richardson, C.D. (1993). The human CD46 molecule is a receptor for measles virus (Edmonston strain). Cell 75, 295–305. Dutko, F.J., and Oldstone, M.B.A. (1983). Genomic and biological variation among commonly used lymphocytic choriomeningitis virus strains. J Gen Virol 64, 1689–1698.

7

Emonet, S.F., Garidou, L., McGavern, D.B., and de la Torre, J.C. (2009). Generation of recombinant lymphocytic choriomeningitis viruses with trisegmented genomes stably expressing two additional genes of interest. Proc Natl Acad Sci U S A 106, 3473–3478. Fingeroth, J.D., Weis, J.J., Tedder, T.F., Strominger, J.L., Biro, P.A., and Fearon, D.T. (1984). Epstein-Barr virus receptor of human B lymphocytes is the C3d receptor CR2. Proc Natl Acad Sci U S A 81, 4510–4514. Hahm, B., Arbour, N., and Oldstone, M.B.A. (2004). Measles virus interacts with human SLAM receptor on dendritic cells to cause immunosuppression. Virology 323, 292–302. Hahm, B., Cho, J.H., and Oldstone, M.B.A. (2007). Measles virusdendritic cell interaction via SLAM inhibits innate immunity: selective signaling through TLR4 but not other TLRs mediates suppression of IL-12 synthesis. Virology 358, 251–251. Hotchin, J. (1962). The biology of lymphocytic choriomeningitis infection: virus-induced immune disease. Cold Spring Harb Symp Quant Biol 27, 479–499. Jamieson, B.D., and Ahmed, R. (1988). T-cell tolerance: Exposure to virus in utero does not cause a permanent deletion of specific T cells. Proc Natl Acad Sci U S A 85, 2265–2268. Joly, E., Mucke, L., and Oldstone, M.B.A. (1991). Viral persistence in neurons explained by lack of major histocompatibility class I expression. Science 253, 1283–1285. Joly, E., and Oldstone, M.B.A. (1992). Neuronal cells are deficient in loading peptides onto MHC class I molecules. Neuron 8, 1185–1190. Jondal, M., Klein, G., Oldstone, M.B.A., Bokish, V., and Yefenof, E. (1976). Surface markers on human B and T lymphocytes. VIII. Association between complement and Epstein-Barr virus receptors on human lymphoid cells. Scand J Immunol 5, 401–410. Kanagawa, M., Saito, F., Kunz, S., Yoshida-Moriguchi, T., Barresi, R., Kobayashi, Y.M., Muschler, J., Dumanski, J.P., Michele, D.E., Oldstone, M.B.A., and Campbell, K.P. (2004). Molecular recognition by LARGE is essential for expression of functional dystroglycan. Cell 117, 953–964. Klavinskis, L.S., Notkins, A.L., and Oldstone, M.B.A. (1988). Persistent viral infection of the thyroid gland: alteration of thyroid function in the absence of tissue injury. Endocrinology 122, 567–575. Klavinskis, L.S., and Oldstone, M.B.A. (1987). Lymphocytic choriomeningitis virus can persistently infect thyroid epithelial cells and perturb thyroid hormone production. J Gen Virol 68, 1867–1873. Klavinskis, L.S., Whitton, J.L., Joly, E., and Oldstone, M.B.A. (1990). Vaccination and protection from a lethal viral infection: identification, incorporation, and use of a cytotoxic T lymphocyte glycoprotein epitope. Virology 178, 393–400. Kunz, S., Rojek, J.M., Kanagawa, M., Spiropoulou, C.F., Barresi, R., Campbell, K.P., and Oldstone, M.B.A. (2005). Posttranslational modification of alpha-dystroglycan, the cellular receptor for arenaviruses, by the glycosyltransferase LARGE is critical for virus binding. J Virol 79, 14282–14296. Kunz, S., Sevilla, N., McGavern, D.B., Campbell, K.P., and Oldstone, M.B.A. (2001). Molecular analysis of the interaction of LCMV with its cellular receptor [alpha]-dystroglycan. J Cell Biol 155, 301–310. Lampert, P.W., Joseph, B.S., and Oldstone, M.B.A. (1975). Antibody-induced capping of measles virus antigens on plasma membrane studied by electron microscopy. J Virol 15, 1248– 1255. Lee, A.M., Cruite, J., Welch, M.J., Sullivan, B.M., and Oldstone, M.B.A. (2013). Pathogenesis of Lassa fever virus infection.

8

I. Susceptibility of mice to recombinant Lassa GP/LCMV chimeric virus. Virology 442, 114–121. Lewicki, H., Tishon, A., Borrow, P., Evans, C.F., Hahn, K.M., Jewell, D.A., Wilson, I.A., and Oldstone, M.B.A. (1995a). CTL escape viral variants. I. Generation and molecular characterization. Virology 210, 29–40. Lewicki, H.A., von Herrath, M.G., Evans, C.F., Whitton, J.L., and Oldstone, M.B.A. (1995b). CTL escape viral variants. II. Biologic activity in vivo. Virology 211, 443–450. Lipkin, W.I., Battenberg, E.L., Bloom, F.E., and Oldstone, M.B.A. (1988). Viral infection of neurons can depress neurotransmitter mRNA levels without histologic injury. Brain Res 451, 333–339. Manchester, M., Gairin, J.E., Patterson, J.B., Alvarez, J., Liszewski, M.K., Eto, D.S., Atkinson, J.P., and Oldstone, M.B.A. (1997). Measles virus recognizes its receptor, CD46, via two distinct binding domains within SCR1-2. Virology 233, 174– 184. Manchester, M., Liszewski, M.K., Atkinson, J.P., and Oldstone, M.B.A. (1994). Multiple isoforms of CD46 (membrane cofactor protein) serve as receptors for measles virus. Proc Natl Acad Sci U S A 91, 2161–2165. Marsolais, D., Hahm, B., Walsh, K.B., Edelmann, K.H., McGavern, D., Hatta, Y., Kawaoka, Y., Rosen, H., and Oldstone, M.B.A. (2009). A critical role for the sphingosine analog AAL-R in dampening the cytokine response during influenza virus infection. Proc Natl Acad Sci U S A 106, 1560–1565. Mucke, L., and Oldstone, M.B.A. (1992). The expression of major histocompatibility complex (MHC) class I antigens in the brain differs markedly in acute and persistent infections with lymphocytic choriomeningitis virus (LCMV). J Neuroimmunol 36, 193–198. Naniche, D., and Oldstone, M.B.A. (2000). Generalized immunosuppression: how viruses undermine the immune response. Cell Mol Life Sci 57, 1399–1407. Nemerow, G.R., Wolfert, R., McNaughton, M.E., and Cooper, N.R. (1985). Identification and characterization of the Epstein-Barr virus receptor on human B lymphocytes and its relationship to the C3d complement receptor (CR2). J Virol 55, 347–351. Ng, C.T., and Oldstone, M.B.A. (2012). Infected CD8a- dendritic cells are the predominant source of IL-10 during establishment of persistent viral infection. Proc Natl Acad Sci U S A 109, 14116–14121. Ng, C.T., Sullivan, B.M., Teijaro, J.R., Lee, A.M., Welch, M., Rice, S., Sheehan, K.C., Schreiber, R.D., and Oldstone, M.B.A. (2015). Blockade of interferon beta, but not interferon alpha, signaling controls persistent viral infection. Cell Host Microbe 17, 653–661. Notkins, A.L., Mahar, S., Scheele, C., and Goffman, J. (1966). Infectious virus-antibody complex in the blood of chronically infected mice. J Exp Med 124, 81–97. Olding, L.B., Jensen, F.C., and Oldstone, M.B.A. (1975). Pathogenesis of cytomegalovirus infection. I. Activation of virus from bone marrow-derived lymphocytes by in vitro allogenic reaction. J Exp Med 141, 561–572. Oldstone, M.B.A. (1959). Altered reactivity to Escherichia coli endotoxin of mice subjected to sublethal tourniquet treatment. Proc Soc Exp Biol Med 102, 256–258. Oldstone, M.B.A. (1975). Virus neutralization and virusinduced immune complex disease. Virus-antibody union resulting in immunoprotection or immunologic injury—two sides of the same coin. Prog Med Virol 19, 84–119.

OLDSTONE

Oldstone, M.B.A. (1989). Viral alteration of cell function. Sci Am 261, 42–48. Oldstone, M.B.A. (1993). Rous-whipple award lecture. Viruses and diseases of the twenty-first century. Am J Pathol 143, 1241–1249. Oldstone, M.B.A. (2002). Biology and pathogenesis of lymphocytic choriomeningitis virus infection. Curr Top Microbiol Immunol 263, 83–117. Oldstone, M.B.A. (2009). Modeling subacute sclerosing panencephalitis in a transgenic mouse system: uncoding pathogenesis of disease and illuminating components of immune control. Curr Top Microbiol Immunol 330, 31–54. Oldstone, M.B.A., Aoki, T., and Dixon, F.J. (1972). The antibody response of mice to murine leukemia virus in spontaneous infection: absence of classical immunologic tolerance (AKR mice-complement-fixing antibodies-lymphocytic choriomeningitis virus-immunofluorescence-glomerular deposits of antigen-antibody complexes). Proc Natl Acad Sci U S A 69, 134–138. Oldstone, M.B.A., Blount, P., Southern, P.J., and Lampert, P.W. (1986). Cytoimmunotherapy for persistent virus infection: unique clearance pattern from the central nervous system. Nature 321, 239–243. Oldstone, M.B.A., and Campbell, K.P. (2011). Decoding arenavirus pathogenesis: essential roles for alpha-dystroglycan-virus interactions and the immune response. Virology 411, 170–179. Oldstone, M.B.A., Dales, S., Tishon, A., Lewicki, H., and Martin, L. (2005). A role for dual viral hits in causation of subacute sclerosing panencephalitis. J Exp Med 202, 1185–1190. Oldstone, M.B.A., Del Villano, B.C., and Dixon, F.J. (1976). Autologous immune responses to the major oncornavirus polypeptides in unmanipulated AKR/J mice. J Virol 18, 176–181. Oldstone, M.B.A., and Dixon, F.J. (1967). Lymphocytic choriomeningitis: production of anti-LCM antibody by ‘‘tolerant’’ LCM-infected mice. Science 158, 1193–1195. Oldstone, M.B.A., Fujinami, R.S., and Lampert, P.W. (1980). Membrane and cytoplasmic changes in virus-infected cells induced by interactions of antiviral antibody with surface viral antigen. Prog Med Virol 26, 45–93. Oldstone, M.B.A., Fujinami, R.S., Tishon, A., Finney, D., Powell, H.C., and Lampert, P.W. (1983). Mapping of the major histocompatibility complex and viral antigens on the plasma membrane of a measles virus-infected cell. Virology 127, 426–437. Oldstone, M.B.A., Holmstoen, J., and Welsh, R.M. (1977). Alterations of acetylcholine enzymes in neuroblastoma cells persistently infected with lymphocytic choriomeningitis virus. J Cell Physiol 91, 459–472. Oldstone, M.B.A., Lewicki, H., Thomas, D., Tishon, A., Dales, S., Patterson, J., Manchester, M., Homann, D., Naniche, D., and Holz, A. (1999). Measles virus infection in a transgenic model: virus-induced immunosuppression and central nervous system disease. Cell 98, 629–640. Oldstone, M.B.A., Rodriguez, M., Daughaday, W.H., and Lampert, P.W. (1984). Viral perturbation of endocrine function: disordered cell function leads to disturbed homeostasis and disease. Nature 307, 278–281. Oldstone, M.B.A., Sinha, Y.N., Blount, P., Tishon, A., Rodriguez, M., von Wedel, R., and Lampert, P.W. (1982). Virus-induced alterations in homeostasis: alterations in differentiated functions of infected cells in vivo. Science 218, 1125–1127. Oldstone, M.B.A., Southern, P., Rodriguez, M., and Lampert, P. (1984). Virus persists in beta cells of islets of Langerhans and is associated with chemical manifestations of diabetes. Science 224, 1440–1443.

THE ANATOMY OF A CAREER IN SCIENCE

Oldstone, M.B.A., Whitton, J.L., Lewicki, H., and Tishon, A. (1988). Fine dissection of a nine amino acid glycoprotein epitope, a major determinant recognized by lymphocytic choriomeningitis virus-specific class I-restricted H-2Db cytotoxic T lymphocytes. J Exp Med 168, 559–570. Patterson, J.B., Cornu, T.I., Redwine, J., Dales, S., Lewicki, H., Holz, A., Thomas, D., Billeter, M.A., and Oldstone, M.B.A. (2001). Evidence that the hypermutated M protein of a subacute sclerosing panencephalitis measles virus actively contributes to the chronic progressive CNS disease. Virology 291, 215–225. Patterson, J.B., Thomas, D., Lewicki, H., Billeter, M.A., and Oldstone, M.B.A. (2000). V and C proteins of measles virus function as virulence factors in vivo. Virology 267, 80–89. Popkin, D.L., Teijaro, J.R., Sullivan, B.M., Urata, S., Rutschmann, S., de la Torre, J.C., Kunz, S., Beutler, B., and Oldstone, M.B.A. (2011). Hypomorphic mutation in the site-1 protease Mbtps1 endows resistance to persistent viral infection in a cell-specific manner. Cell Host Microbe 9, 212–222. Rall, G.F., Manchester, M., Daniels, L.R., Callahan, E.M., Belman, A.R., and Oldstone, M.B.A. (1997). A transgenic mouse model for measles virus infection of the brain. Proc Natl Acad Sci U S A 94, 4659–4663. Rall, G.F., Mucke, L., and Oldstone, M.B.A. (1995). Consequences of cytotoxic T lymphocyte interaction with major histocompatibility complex class I-expressing neurons in vivo. J Exp Med 182, 1201–1212. Rice, G.P., Schrier, R.D., and Oldstone, M.B.A. (1984). Cytomegalovirus infects human lymphocytes and monocytes: virus expression is restricted to immediate-early gene products. Proc Natl Acad Sci U S A 81, 6134–6138. Rowe, W.P. (1954). Studies on pathogenesis and immunity in lymphocytic choriomeningitis infection of the mouse. Res Rept Naval Med Res Inst 12, 167. Salvato, M., Borrow, P., Shimomaye, E., and Oldstone, M.B.A. (1991). Molecular basis of viral persistence: a single amino acid change in the glycoprotein of lymphocytic choriomeningitis virus is associated with suppression of the antiviral cytotoxic T-lymphocyte response and establishment of persistence. J Virol 65, 1863–1869. Schrier, R.D., Nelson, J.A., and Oldstone, M.B.A. (1985). Detection of human cytomegalovirus in peripheral blood lymphocytes in a natural infection. Science 230, 1048–1051. Sevilla, N., Kunz, S., Holz, A., Lewicki, H., Homann, D., Yamada, H., Campbell, K.P., de la Torre, J.C., and Oldstone, M.B.A. (2000). Immunosuppression and resultant viral persistence by specific viral targeting of dendritic cells. J Exp Med 192, 1249–1260. Sevilla, N., McGavern, D.B., Teng, C., Kunz, S., and Oldstone, M.B.A. (2004). Viral targeting of hematopoietic progenitors and inhibition of DC maturation as a dual strategy for immune subversion. J Clin Invest 113, 737–745. Sullivan, B.M., Emonet, S., Welch, M.J., Lee, A.M., Campbell, K.P., de la Torre, J.C., and Oldstone, M.B.A. (2011). Point mutation in the glycoprotein of lymphocytic choriomeningitis virus is necessary for receptor binding, dendritic cell infection, and long-term persistence. Proc Natl Acad Sci U S A 108, 2969–2974. Tatsuo, H., Ono, N., Tanaka, K., and Yanagi, Y. (2000). SLAM (CDw150) is a cellular receptor for measles virus. Nature 406, 893–897. Teijaro, J.R., Ng, C., Lee, A.M., Sullivan, B.M., Sheehan, K.C., Welch, M., Schreiber, R.D., de la Torre, J.C., and Oldstone, M.B.A. (2013). Persistent LCMV infection is controlled by blockade of type I interferon signaling. Science 340, 207–211.

9

Teijaro, J.R., Walsh, K.B., Cahalan, S., Fremgen, D.M., Roberts, E., Scott, F., Martinborough, E., Peach, R., Oldstone, M.B.A., and Rosen, H. (2011). Endothelial cells are central orchestrators of cytokine amplification during influenza virus infection. Cell 146, 980–991. Tishon, A., Lewicki, H., Andaya, A., McGavern, D., Martin, L., and Oldstone, M.B.A. (2006). CD4 T cell control primary measles virus infection of the CNS: regulation is dependent on combined activity with either CD8 T cells or with B cells: CD4, CD8 or B cells alone are ineffective. Virology 347, 234–245. Walsh, K.B., Teijaro, J.R., Wilker, P.R., Jatzek, A., Fremgen, D.M., Das, S.C., Watanabe, T., Hatta, M., Shinya, K., Suresh, M., Kawaoka, Y., Rosen, H., and Oldstone, M.B.A. (2011). Suppression of cytokine storm with a sphingosine analog provides protection against pathogenic influenza virus. Proc Natl Acad Sci U S A 108, 12018–12023. Ward, S.V., George, C.X., Welch, M.J., Liou, L.Y., Hahm, B., Lewicki, H., de la Torre, J.C., Samuel, C.E., and Oldstone, M.B.A. (2011). RNA editing enzyme adenosine deaminase is a restriction factor for controlling measles virus replication that also is required for embryogenesis. Proc Natl Acad Sci U S A 108, 331–336. Wherry, E.J. (2011). T cell exhaustion. Nat Immunol 12, 492– 499. Whitton, J.L., Southern, P.J., and Oldstone, M.B.A. (1988). Analyses of the cytotoxic T lymphocyte responses to glycoprotein and nucleoprotein components of lymphocytic choriomeningitis virus. Virology 162, 321–327. Whitton, J.L., Tishon, A., Lewicki, H., Gebhard, J., Cook, T., Salvato, M., Joly, E., and Oldstone, M.B.A. (1989). Molecular analyses of a five-amino-acid cytotoxic T-lymphocyte (CTL) epitope: an immunodominant region which induces nonreciprocal CTL cross-reactivity. J Virol 63, 4303–4310. Yefenof, E., Klein, G., Jondal, M., and Oldstone, M.B.A. (1976). Surface markers on human B- and T-lymphocytes. IX. Twocolor immunofluorescence studies on the association between EBV receptors and complement receptors on the surface of lymphoid cell lines. Int J Cancer 17, 693–700. Yoshida-Moriguchi, T., Yu, L., Stalnaker, S.H., Davis, S., Kunz, S., Madson, M., Oldstone, M.B.A., Schachter, H., Wells, L., and Campbell, K.P. (2010). O-mannosyl phosphorylation of alpha-dystroglycan is required for laminin binding. Science 327, 88–92. Zuniga, E.I., Liou, L.Y., Mack, L., Mendoza, M., and Oldstone, M.B.A. (2008). Persistent virus infection inhibits type 1 interferon production by plasmacytoid dendritic cells to facilitate opportunistic infections. Cell Host Microbe 4, 374–386. Zuniga, E.I., McGavern, D.B., Pruneda-Paz, J.L., Teng, C., and Oldstone, M.B.A. (2004). Bone marrow plasmacytoid dendritic cells can differentiate into myeloid dendritic cells upon virus infection. Nat Immunol 5, 1227–1234.

Address correspondence to: Michael B.A. Oldstone, MD Viral-Immunobiology Laboratory Department of Immunology & Microbial Science The Scripps Research Institute 10550 North Torrey Pines Road La Jolla, CA 92037 E-mail: [email protected] Received for publication January 6, 2016; received in revised form January 6, 2016; accepted January 6, 2016.

The Anatomy of a Career in Science.

The Anatomy of a Career in Science. - PDF Download Free
128KB Sizes 0 Downloads 11 Views