SIGHTINGS



Emerging And
Re-Emerging Viruses
By Alison Jacobson
Department of Microbiology
University of Cape Town
http://www.tulane.edu/~dmsander/WWW/335/Emerging1.html#EMERG
10-8-99
 
 
 
* Introduction * Theoretical Considerations * A Historical Perspective * Examples of Emerging & Potentially Emergent Viruses * Arbovirus Diseases * Aetiology * Epidemiology * Transmission * Immunity * Active Immunity * Passive Immunity * General Clinical Features * Ecological Factors
 
CASES IN POINT: * HANTAVIRUSES: * Hantaan virus * Seoul virus * Four Corners virus * Ebola virus * Influenza virus: A model for emergence * Mutation Frequency * Selective Pressures & Constraints * Conclusion * References
 
 
 
INTRODUCTION
 
Within the last decade there has been an ever increasing awareness of the Darwinian struggle with which the human species is engaged. Microbial and viral predators abound, in no less abundance than before, and still present a constant threat to individual survival as well as to the success of the population at large. Following decades of general complacency in the antibiotic era, a startling turning point has been reached, spurred in large part by the global ravages of the human immunodeficiency virus (HIV) - one of the simplest of viral constructs - which still evades cure or even true understanding. The last two years have been accented by several striking episodes of disease emergence, such as multi-drug resistant tuberculosis, acute coccal infections, the rodent-borne pneumonic hantavirus in the United States, food- and waterborne outbreaks of Salmonella infections, cholera and illnesses caused by the Shigella-like Escherichia coli O157. Against these newer threats is a perpetual backdrop of a multiplicity of infections, which cycle throughout their ecological niches and are encountering opportunities in our modern world to spread with a frightening vigour. Currently, we are experiencing the epidemic potential of HIV, but the next epidemic of human disease may be entirely different. A few examples of general emerging infectious diseases are shown below:
 
Examples of Emerging Infectious Diseases:
 
USA: * E. coli O157:H7 disease * Cryptosporidiosis * Hantavirus pulmonary syndrome * Coccidioidiomycosis * Vancomycin-resistant enterococci and multiply-resistant pneumococci
 
International: * Vibrio cholerae O139 in Asia * Vibrio cholerae O1 in Latin America * Yellow fever virus in Kenya * Rift Valley fever virus in Egypt E coliO157:H7 disease
 
To quote Donald A.Henderson of the U.S. Office of Science and Technology Policy: "The recent emergence of AIDS and Dengue hemorrhagic infections, among others, are serving usefully to disturb our ill-founded complacency about infectious diseases. Such complacency has prevailed in this country (USA) throughout much of my career... It is evident now, as it should have been then, that mutation and change are facts of nature, that the world is increasingly interdependent, and that human health and survival will be challenged, ad infinitum, by new mutant microbes, with unpredictable pathophysiological manifestations...How are we to detect these at an early date so as to be able to devise appropriate preventative and therapeutic modalities? What do we look for? What types of surveillance and reporting systems can one devise?" (Morse 1993).
 
The Centers for Disease Control and Prevention (CDC), in an attempt to ensure that its prevention and control programs keep pace with the numerous and changing health problems that threaten all segments of our diverse society, has identified four priority areas: 1. strengthen core public health functions; 2. develop, maintain, and enrich the capacity to respond to urgent threats to health; 3. develop a nationwide prevention network and program; and 4. promote women's health.
 
Leading the list of urgent threats to health are new and emerging infections. It must be recognized that the health of the developed world's people is inextricably linked to the health of people in other nations, infectious diseases can and do disseminate rapidly around the globe, and global surveillance for emerging infections is vital to public health. While it is vital for us to recognize and promote this awareness, we too have an integral role to play in surveillance and basic research.
 
There seems to be two stages in public health crises. The first is, "I don't believe you." And the second is "Why didn't you tell me?" (Dowdle 1994). Everyone tends to think in very short terms. Although officials need to recognize and respond to the critical `disease of the year', it is also necessary to maintain the infrastructure needed to identify emerging diseases as well as to develop effective ways for combating them. The problem can be made more difficult when resources are set aside for the spotlighted problem without ensuring that the less glamorous infrastructure needs to continue to be met. In this essay I will attempt to address these issues, as well as to highlight appropriate examples of emergent disease, particularly those of viral origin.
 
THEORETICAL CONSIDERATIONS
 
Joshua Lederburg has commented that viruses are humanity's only real competitors for dominion of the planet, serving as both parasites and genetic elements in their hosts (Lederburg 1988). Not only do they have considerable plasticity, enabling them to evolve in new directions, but their genetic and metabolic entanglements with cells uniquely positions them to mediate subtle, cumulative evolutionary changes in their hosts as well. In contrast, they are also able to decimate entire populations. The fact that long term natural selection favours mutualism offers only limited encouragement to our species, with millions of people suffering before an equilibrium can be reached.
 
Coevolution of viruses and host can follow several possible lines estimated by modelling techniques, and pathogens may not always evolve towards lower virulence. For example, a virus strain that kills much faster will not be favoured over a less virulent strain if it has a modest transmission advantage, but it will prevail if it is much more readily transmitted than the less-virulent strain. In the trade-off between transmissibility and virulence, many viruses evolve toward a middle ground, favouring transmissibility but allowing them to retain some virulence. Viruses that are transmitted over a long time, for example HIV, have a selective advantage even when their effective rates of transmission are relatively low (Anderson and May 1987).
 
However, although mathematical approaches offer useful insights, they are often inadequate in predicting outcomes because viral emergence and host interactions are so complex, being dependent on both the genetics of the host and external conditions. These predictive attempts, a true test for chaos theorists, have even more variables than say, equations dealing with predictions of meteorological patterns, because such climatic conditions make up just one part of the ecological picture within which biological evolution maneuvers. Furthermore, stock market predictions, another favourite of chaos theorists, can also be seen to form just one piece of the puzzle that is infectious disease, as many of our responses to the latter problem are politically and financially interwoven.
 
A HISTORICAL PERSPECTIVE
 
New patterns of human movement, leading to new contacts across what had once been geographic boundaries, have been seen to give rise to a variety of emergent infections. Examples are the introduction of smallpox into the Americas and of syphilis into Europe. Yellow fever probably emerged in the New World as a result of the African slave trade, which brought Aedes aegypti in water containers of ships. Similarly, the rise of Dengue hemorrhagic fever in Southeast Asia in the late 1940s is attributed to rapid migration to cities with open water storage, which favoured proliferation of the mosquito or other suitable vectors. Of current concern in the USA is the fact that Aedes albopictus, an aggressive and competent dengue virus vector, was brought to Houston in used Asian tires and has established itself in at least 17 American states (Morse and Schluederberg 1990).
 
RECENT EXAMPLES OF EMERGING AND POTENTIALLY EMERGENT VIRUSES
 
Most emergent viruses are zoonotic, with natural animal reservoirs a more frequent source of new viruses than is the spontaneous evolution of a new entity. The most frequent factor in emergence is human behaviour that increases the probability of transfer of viruses from their endogenous animal hosts to man. Rodents and arthropods are most commonly involved in direct transfer, and changes in agricultural practices or urban conditions that promote rodent or vector multiplication favour increased incidence of human disease. Other animals, especially primates, are important reservoirs for transfer by arthropods. Because arthropod transmission plays a very large part in infectious animal disease, specifically potential emergent virus epidemics, I will dedicate the next part of this essay to a discussion of them.
 
ARBOVIRUS DISEASES
 
Approximately 100 of the more than 520 known arthropod-borne viruses (arboviruses) cause human disease. At least 20 of these might fulfill the criteria for emerging viruses, appearing in epidemic form at generally unpredictable intervals (Morse and Schluederberg 1990). These viruses are usually spread by the bites of arthropods, but some can also be transmitted by other means, for example through milk, excreta or aerosols. The arbovirus infections are maintained in nature principally, or to an important extent, through biological transmission between susceptible vertebrate hosts by blood-sucking insects; they multiply to produce viraemia in the vertebrates, multiply in the tissues of the insects and are passed on to new vertebrates by the bites of insects after a period of extrinsic incubation. The names by which these viruses are known are often place names such as West Nile or Rift Valley, or are based on clinical characteristics like yellow fever.
 
AETIOLOGY
 
Most arboviruses are spherical, measuring 17-150 nm or more, a few are rod-shaped, measuring 70 x 200 nm. All are RNA viruses. Many circulate in a natural environment and do not infect man. Some infect man only occasionally or cause only a mild illness; others are of great clinical importance causing large epidemics and many deaths. Specifically, these belong to the Togaviridae, the alphaviruses, flaviviruses, the Bunyaviridae, nairoviruses, phleboviruses and other subgroups.
 
EPIDEMIOLOGY
 
Vertebrate hosts
 
Maintenance, incidental, link and amplifier hosts are categorized according to Stickland Hunter's "Tropical Medicine" (1991) as the following:
 
Maintenance hosts are essential for the continued existence of the virus, usually living in symbiosis with the viruses, without actual disease, but they do develop antibodies. These include birds such as the prairie chicken, pigeon and wood thrush which transmit Eastern and Western equine encephalitis; heron and egrets transmitting Japanese encephalitis and migrating birds which travel over long distances carrying these and other similar viruses; rodents and insectivores such as rats, hedgehogs, lemmings and chipmunks are known to carry louping ill and Colorado tick fever; primates such as monkeys which carry Dengue fever; Leporidae (rabbits and hares) which carry Californian encephalitis; Ungulates (cattle and deer) which are implicated in the transmission of European tick-borne encephalitis; bats which carry Rio Brava virus; and marsupials, reptiles and amphibia such as kangaroos and snakes which also harbour encephalitis-causing viruses.
 
Incidental hosts become infected, but transmission from them does not occur with sufficient regularity for stable maintenance. Man is usually an incidental host, often, but not always, being a dead end in the chain. These hosts may or may not show symptoms. Link hosts bridge a gap between maintenance hosts and man, for example, between small mammals and man by goats (via milk) in tick-borne encephalitis. Amplifier hosts increase the weight of infection, as is the case with pigs which act between wild birds and man in Japanese encephalitis.
 
The populations and characters of the vertebrate hosts and their threshold levels of viraemia are important. Small rodents multiply rapidly and have short lives, thus providing a constant supply of susceptible individuals. In contrast, monkeys and pigs multiply slowly, and once they have recovered from an infection, remain immune for life. African monkeys are relatively resistant to Yellow Fever, but Asian and American monkeys are susceptible, probably because, unlike the African monkeys, they have not been exposed continuously for centuries to the infection. Also, possibly related arboviruses may offer partial immunization.
 
Invertebrate hosts
 
Mosquitoes, sandflies and ticks may imbibe virus from a vertebrate in a state of viraemia, after which the virus undergoes an incubation period within the arthropod, known as the extrinsic incubation period. In mosquitoes this period is short: 10 days at 30o C ambient temperature and longer at lower temperatures. Mosquitoes remain infective for life without any apparent ill-effects. In fact, their infectivity appears to increase with time after infection and their effectiveness as transmitters depends upon the frequency with which they bite. It is also possible that arthropods, whose mouth parts are contaminated by virus in the act of feeding, could transmit the virus mechanically if they feed soon afterwards on another animal. For instance, chikungunya virus can be transmitted mechanically by A. aegypti for 8 hours after infection. In general, mosquito-borne viruses may not use ticks as vectors nor can tick-borne viruses reside in mosquitoes.
 
TRANSMISSION
 
Arthropod transmission involves several stages: 1. Ingestion by the arthropods of virus in the blood (usually) or tissue fluids of the vertebrate hosts 2. Penetration of the viruses into the tissue of the arthropods, in the gut wall, or elsewhere after passing through the gut barrier 3. Multiplication of the viruses in the arthropod cells, including those of the salivary glands.
 
Stage 2 and part of stage 3 represent the extrinsic incubation period of the disease (Hunter 1991). The quantity of blood, and therefore the amount of virus ingested, seems to be important as each arthropod species must ingest a minimum quantity of a given virus before multiplication can take place. The same mosquito species can have two different thresholds for two different viruses and if one species has a low threshold, other species may have high thresholds or may be completely resistant. This threshold phenomenon is extremely important in determining the efficiency of a vector and may also vitally affect the course of an epidemic. Viruses reportedly persist in overwintering mosquitoes, while transovarian passage of virus has been seen in some tick species. For mosquitoes the availability of suitable breeding places (and therefore rainfall) is a major factor. An efficient vector may have a wide range of animals on which to feed, but if the arthropod species is abundant, and even if it bites man only infrequently in the presence of other (and preferred) animals, the large numbers enable it to maintain transmission to man. For example, Culex tritaeniorhynchus, which mostly bites birds, Bovidae, dogs and especially porcines, and only to a limited extent man, can maintain transmission of Japanese encephalitis from pigs to man by sheer numbers.
 
Although transmission of arboviruses usually takes place through the bites of arthropods, Lassa virus, for example, may be transmitted through contact with excreta of infective rodents, and others via urine or faeces infecting the nasopharynx, some through aerosol from a patient or others by one bird pecking another.
 
IMMUNITY
 
After a vertebrate has been infected, the arbovirus probably multiplies first in the regional lymph glands where the earliest formation of antibodies also probably takes place. Some do not produce high titres of antibodies in man and some antibodies are short-lived or appear late. In diagnosis, haemagglutination-inhibiting and complement-fixing antibodies are important, but the only protective antibody is of the neutralizing type, which is also the most specific.
 
Arboviruses are grouped according to antigenic characters, but after inoculation of one virus into a fresh animal, not only the homologous antibodies, but also heterologous antibodies reacting with other viruses of the same group tend to appear. Recovery from an infection by a member of one group of arboviruses may provide some degree of resistance to a subsequent infection by another member of the same group. For example, infection with West Nile virus may have modified the Ethiopian epidemic of Yellow Fever in 1962. Again, the effect of prior infection with Zika, Uganda S and other related viruses in the forest belt of Nigeria, leading to a high incidence of related antibodies, is suggested as the explanation of the absence of epidemic Yellow Fever in man in that area. These related infections probably modify the disease rather than prevent infection.
 
ACTIVE IMMUNITY
 
With Yellow Fever, neutralizing antibodies can be found as early as a few days after the beginning of the disease and are found constantly for many years in the sera. The persistence of immunity does not depend on exogenous reinfection. It is probable that a mosquito infected with Yellow Fever is not harmed by it, but continues to excrete the virus throughout life. This means a continuous supply and release of virus, probably from the epithelial cells of the salivary glands. The virus enters man (or other animals) and gains the liver and other epithelia, provoking the early antibodies in the blood, which neutralize circulating viruses. But, as suggested by Hunter (1991), antibodies which can be detected for so many years in man must stem from a continuing stimulus, and the sensitive cells and their progeny probably have a prophase equivalent of the virus incorporated into their genome, with occasional reversion to productive development which provides the stimulus for further antibody formation. A degree of immunity of this kind may possibly be provided when a related virus invades epithelial cells.
 
PASSIVE IMMUNITY
 
Infant rhesus monkeys and human infants born of mothers immune to Yellow Fever have transient protective antibodies in their sera at birth which persist for several months. They are probably placentally transferred, rather than coming from the mother's milk, because antibodies may disappear from infant sera while they are still suckling. Passive immunity induced by injection of homologous immune serum, has been used for protection against tick-borne encephalitis in cases of special risk and similar sera could be used against other infections, particularly after laboratory or hospital accidents.
 
GENERAL CLINICAL FEATURES
 
Most arbovirus infections are inapparent, that is they produce no symptoms or often only mild ones (fever and occasional rash). For example, in an epidemic of Japanese encephalitis it was estimated that for each case of apparent disease there were 500-1000 inapparent infections. If clinical manifestations arise after infection they do so after an intrinsic incubation period lasting from a few days to a week or more. Some arboviruses damage the endothelial lining of the capillaries increasing permeability which allows the virus to pass the blood brain barrier causing meningoencephalitis. Others damage the parenchymatous organs by direct damage to the cells in which they are situated, while with others damage is caused by the immune system of the host from the formation of antigen- antibody complexes and disordered complement formation which damage the renal tubules and alter the coagulation and fibrinolytic systems of the body causing haemorrhage (viral hemorrhagic fevers). There is a general pattern of biphasic illness, the first phase associated with viraemia ending when antibodies appear in the blood and the second phase when the virus is located in organs, such as the liver or brain.
 
The onset of clinical manifestations is usually abrupt, generally occurring after the onset of viraemia. Fever is usual and is sometimes the only sign. In many cases the clinical manifestations last only while the virus is disseminated, but in other cases there is remission, short or long. If long, the disease is biphasic. After this, fever returns with signs indicating localization of the virus in certain organs. If the period of viraemia has been symptomless and the virus becomes localized in the central nervous system, encephalitis appears. In hemorrhagic cases there is a special risk of shock which can rapidly become irreversible unless promptly treated (Hunter 1991).
 
ECOLOGICAL FACTORS
 
Microorganisms and viruses are adapted to extremely diverse econiches. One of the most complex sets of adaptive characteristics concern arthropod transmission of viruses. The arthropod-borne viruses are spectacular examples of emergence and re-emergence resulting from innocent environmental manipulation or natural environmental change. Deforestation, amateur irrigation and the introduction of new species (usually livestock) gives rise to many virus disease threats of humans and animals. Important aspects of ecological change and their relation to arbovirus life cycles are: 1. Population movements and the intrusion of humans and domestic animals into new arthropod habitats, particularly tropical forests 2. Deforestation, with development of new forest-farmland margins and exposure of farmers and domestic animals to new arthropods 3. Irrigation, especially primitive irrigation systems, which are oblivious to arthropod control 4. Uncontrolled urbanization, with vector populations breeding in accumulations of water (tin cans, old tires etc.) and sewage 5. Increased long distance air travel, with potential for transport of arthropod vectors 6. Increased long-distance livestock transportations, with potential for carriage of viruses and arthropods (especially ticks) 7. New routing of long-distance bird migration brought about by new man-made water impoundments (Murphy 1994).
 
To illustrate the effect ecological change can have on the emergence of a new disease and the course of it afterwards one can look to dengue, one of the most rapidly expanding diseases in tropical parts of the world, with millions of cases occurring each year. For example, Puerto Rico had five dengue epidemics in the first 75 years of this century, but has had six epidemics in the past 11 years, at an estimated cost of over $150 million. Simultaneously, Brazil, Nicaragua and Cuba have had their first major dengue epidemic in over 50 years, involving multiple virus types. At the lethal end of the dengue spectrum is dengue haemorrhagic fever, first occurring in the Americas in 1981. Since then, 11 countries have reported cases, and since 1990 over 3000 cases have been reported annually. Figure 3 illustrates the extent of dengue occurrence globally.
 
The primary reason that dengue is emerging and re-emerging is vector control. National priority lists are political in nature and tend to emphasize daily problems, not episodic ones. Expensive mosquito control tends to fall off the bottom of the list. Meanwhile, as older cheaper chemicals lose effectiveness or are banned, new and expensive chemicals replace them. Before 1970, A. aegypti, the vector of dengue and Yellow Fever, was targeted for regional or even global eradication through the use of DDT (dichlorodiphenyltrichloroethane) (Murphy 1993). Obviously, this solution is no longer applicable, but nothing has effectively supplanted it.
 
 
CASES IN POINT
 
HANTAAN VIRUS
 
The hantavirus (mentioned above) is also the focus of much international attention. During the Korean War of 1950-1952, thousands of United Nations troops developed a mysterious disease marked by fever, headache, hemorrhage and acute renal failure; the mortality rate was 5-10%. Despite much research, the agent of this disease remained unknown for 28 years, when a new virus, named Hantaan virus, was isolated in Korea from field mice. Recently, related viruses have been found in many parts of the world in association with different rodents and as the cause of human diseases with a variety of little-known local names. Epidemic haemorrhagic fever, one of the most important diseases in China, causes more than 100000 cases per year. Transmission to humans is primarily by inhalation of aerosolized excreta. In May 1993 a cluster of deaths in the southwestern United States set in motion a multiagency local, state and federal investigation that led to the discovery of a highly pathogenic hantavirus and to the definition of a new clinical syndrome (Peters 1994).
 
SEOUL VIRUS
 
Another virus of current interest in the USA, Seoul virus, was identified about 10 years ago in Korea as a Hantaan-like virus whose natural host is the urban rat. Serologic surveys detect it worldwide, including seroprevalence rates of 12% in urban rats in Philadelphia and about 64% in Baltimore rats (Le Duc 1986). Although acute hemorrhagic fever was not identified in inner-city Baltimore, 1.3% of 1148 local residents were antibody-positive and the possibility of viral association with chronic renal disease is under study.
 
"FOUR CORNERS VIRUS"
 
The disease hantavirus pulmonary syndrome (HPS), is characterized by an initial fever followed by the abrupt onset of acute pulmonary edema and shock. After recognition of the initial cases by observant clinicians in the Southwest, investigations were swiftly mounted by local university and public health workers but, in spite of efficient and competent studies, failed to find the cause. By the time the CDC became involved, a number of possible causative agents had been ruled out, leading most of the investigators to believe they were dealing with a new entity. This observation led to a broadly based approach to the field epidemiology and the laboratory study of the disease. Samples from the field investigations were distributed among many different laboratories of the National Center for Infectious Disease (NCID) for analysis by the most sensitive classic and modern molecular biological tests for a wide range spectrum of infectious agents.
 
Somewhat surprisingly, successful results were obtained after only a few days of straightforward serologic tests for hantaviruses. The hemoconcentration, thrombocytopenia and shock observed in some of the patients had raised speculation about the involvement of these viral agents; however they had been previously known as associated with renal syndromes only. The serologic results came from established techniques such as indirect fluorescent-antibody assays and enzyme-linked immunosorbent assays. The next steps utilized reverse transcription and PCR amplification of RNA in postmortem tissue samples (60% of confirmed cases to date have been fatal), using consensus primers based on known hantavirus RNA sequences. These yielded products with sequences typical of hantavirus but clearly different from any known member of the genus. This provided additional evidence for the hantavirus etiology and linked the new hantavirus closely to the human disease by its presence in the tissues of people dying of the infection. Using the genomic sequences from human tissues, investigators were subsequently able to implicate the deer mouse as the principle reservoir of the virus.
 
Hantaviruses have traditionally been difficult to propagate, and this one was no exception. Thus a full-length cDNA clone of the small RNA segment of the virus was synthesized. This technique provided a diagnostic reagent of increased sensitivity that could be made widely available. Eventually, full length RNA sequences were developed for the medium segment and a partial sequence was determined for the large segment, permitting the definitive determination that the new virus, isolated weeks later and registered as Muerto Canyon virus, was not a reassortant of any known hantavirus.
 
Immunohistochemical identification of hantavirus antigens and in situ hybridization with genomic sequences also confirmed the hantavirus etiology of the syndrome. The extensive presence of antigen in pulmonary capillaries provided an explanation for the pathophysiology and target organ specificity differing from that of other known disease-causing hantaviruses. This method, when applied to paraffin- imbedded tissues, has also served as a retrospective diagnostic tool, firmly identifying fatal cases from 10 to 15 years ago.
 
The rapid recognition of the hantavirus etiology of this disease was important in that it alleviated heightened fear among the general American population, and saved lives by focusing public health recommendations on the avoidance of contact with potentially infected rodents. Different hantaviruses have been isolated in Louisiana, Florida and also Brazil, indicating the uncommon, yet widespread nature of this disease. Recently (Diglisic 1994), isolation of a hantavirus from Mus musculus captured in Yugoslavia was reported.
 
As stated by C.J. Peters, chief of the Special Pathogens Branch of the Division of Viral and Rickettsial Diseases at NCID, the crucial role of modern techniques in virology was possible only in a context of past hantavirus research, and as part of efforts of a multidisciplinary team of clinicians, epidemiologists, field ecologists and classic microbiologists. The need for basic research is highlighted by the applied practical success which resulted from it, as was the case in identifying a new strain of hantavirus. Future research will need to investigate the molecular mechanisms for induction of pulmonary edema and an appropriate blocking therapy. The evolutionary relationships of the hantaviruses and their rodent host specificity must be understood to predict the future course of transmission, and finally the basis for the different tropisms of the viruses must be examined at a molecular level.
 
EBOLA VIRUS
 
Ebola virus, a member of the <Filoviruses.htmlFiloviridae, burst from obscurity with spectacular outbreaks of severe, haemorrhagic fever. It was first associated with an outbreak of 318 cases and a case-fatality rate of 90% in Zaire and caused 150 deaths among 250 cases in Sudan. Smaller outbreaks continue to appear periodically, particularly in East, Central and southern Africa. In 1989, a haemorrhagic disease was recognized among cynomolgus macaques imported into the United States from the Philippines. Strains of Ebola virus were isolated from these monkeys. Serologic studies in the Philippines and elsewhere in Southeast Asia indicated that Ebola virus is a prevalent cause of infection among macaques (Manson 1989).
 
These threadlike polymorphic viruses are highly variable in length apparently owing to concatemerization. However, the average length of an infectious virion appears to be 920 nm. The virions are 80 nm in diameter with a helical nucleocapsid, a membrane made of 10 nm projections, and host cell membrane. They contain a unique single-stranded molecule of noninfectious (negative sense ) RNA. The virus is composed of 7 polypeptides, a nucleoprotein, a glycoprotein, a polymerase and 4 other undesignated proteins. Proteins are produced from polyadenylated monocistronic mRNA species transcribed from virus RNA. The replication in and destruction of the host cell is rapid and produces a large number of viruses budding from the cell membrane.
 
Epidemics have resulted from person to person transmission, nosocomial spread or laboratory infections. The mode of primary infection and the natural ecology of these viruses are unknown. Association with bats has been implicated directly in at least 2 episodes when individuals entered the same bat-filled cave in Eastern Kenya. Ebola infections in Sudan in 1976 and 1979 occurred in workers of a cotton factory containing thousands of bats in the roof. However, in all instances, study of antibody in bats failed to detect evidence of infection, and no virus was isolated form bat tissue.
 
The index case in 1976 was never identified, but this large outbreak resulted in 280 deaths of 318 infections. The outbreak was primarily the result of person to person spread and transmission by contaminated needles in outpatient and inpatient departments of a hospital and subsequent person to person spread in surrounding villages. In serosurveys in Zaire, antibody prevalence to Ebola virus has been 3 to 7%. The incubation period for needle- transmitted Ebola virus is 5 to 7 days and that for person to person transmitted disease is 6 to 12 days.
 
The virus spreads through the blood and is replicated in many organs. The histopathologic change is focal necrosis in these organs, including the liver, lymphatic organs, kidneys, ovaries and testes. The central lesions appear to be those affecting the vascular endothelium and the platelets. The resulting manifestations are bleeding, especially in the mucosa, abdomen, pericardium and vagina. Capillary leakage appears to lead to loss of intravascular volume, bleeding, shock and the acute respiratory disorder seen in fatal cases. Patients die of intractable shock. Those with severe illness often have sustained high fevers and are delirious, combative and difficult to control.
 
EBOLA SEROLOGY
 
The serologic method used in the discovery of Ebola was the direct immunofluorescent assay. The test is performed on a monolayer of infected and uninfected cells fixed on a microscopic slide. IgG- or IgM-specific immunoglobulin assays are performed. These tests may then be confirmed by using western blot or radioimmunoprecipitation. Virus isolation is also a highly useful diagnostic method, and is performed on suitably preserved serum, blood or tissue specimens stored at -70oC or freshly collected.
 
TREATMENT OF EBOLA
 
No specific antiviral therapy presently exists against Ebola virus, nor does interferon have any effect. Past recommendations for isolation of the patient in a plastic isolator have given way to the more moderate recommendation of strict barrier isolation with body fluid precautions. This presents no excess risk to the hospital personnel and allows substantially better patient care, as shown in Table 2. The major factor in nosocomial transmission is the combination of the unawareness of the possibility of the disease by a worker who is also inattentive to the requirements of effective barrier nursing. after diagnosis, the risk of nosocomial transmission is small.
 
PREVENTION AND CONTROL OF EBOLA
 
The basic method of prevention and control is the interruption of person to person spread of the virus. However, in rural areas, this may be difficult because families are often reluctant to admit members to the hospital because of limited resources and the culturally unacceptable separation of sick or dying patients from the care of their family. Experience with human disease and primate infection suggests that a vaccine inducing a strong cell- mediated response will be necessary for virus clearance and adequate protection. Neutralizing antibodies are not observed in convalescent patients nor do they occur in primates inoculated with killed vaccine. A vaccine expressing the glycoprotein in vaccinia is being prepared for laboratory evaluation.
 
INFLUENZA VIRUS: A MODEL FOR VIRAL EMERGENCE
 
Influenza, both human and avian, would be very high on the list of disease with epidemic potential. Up to 20% of the population has been seen to become ill during a single epidemic, with 50000 deaths per year in the United States (Murphy 1994). In the 1918 pandemic more than 500000 people throughout the world died.
 
The probability of interspecies transfer can be increased not only by increased contact between humans and an animal reservoir, but also by increased opportunity for viral genetic reassortment or recombination within animal or insect hosts. Because influenza virus has an eight-segmented genome, it has considerable freedom for such a genetic reassortment. While small epidemics may arise from mutation (antigenic drift), all known human pandemic strains have been the result of reassortment, mostly involving the hemagglutinin (H) gene. Kida et al.(1988) and also Scholtissek and Naylor (1988) state that influenza virus maintained in shore and migrating birds infect ducks raised on farms and reassort in pigs, from which new strains emerge to infect humans.
 
Virulent strains of influenza virus can also arise from a single mutation, even if pandemic strains have not generally arisen this way. For example, in 1983 a single mutation in a relatively avirulent strain gave rise to an H5N2 strain that caused a fatal epidemic in chickens in Pennsylvania. The point mutation in the H gene changed threonine to lysine, exposing a previously glycosylated site. Similarly, and remarkably, if pigs are infected experimentally with an avirulent mutant, the swine virulent parental phenotype emerges within a few days, indicating rapid evolution and emergence in vivo of the virulent form (Kilbourne et al. 1988).
 
For viruses with non-segmented genomes, recombination provides another genetic avenue for emergent diseases. For instance, viral genetic sequence analysis revealed that Western equine encephalomyelitis virus, an alphavirus, arose from a recombination event that seems to have involved a Sindbis-like virus and Eastern equine encephalomyelitis virus, probably occurring some 100-200 years ago (Hahn et al. 1988). Genetic recombination also seems to have occurred between the envelope protein genes of human T lymphotropic virus (HTLV)-I and HTLV-II (Doolittle et al. 1989).
 
MUTATION FREQUENCY
 
 
The mutation rate of any genome is inversely proportional to its size. However, RNA viruses usually have higher mutation rates than do DNA viruses of the same genome size. This is generally ascribed to the lack of error-correcting mechanisms in RNA synthesis. High mutation rates have been reported in influenza genes. The changes occurred in the non-structural protein (NS) gene of influenza A virus during a single cycle of replication in tissue culture. A mutation rate of approximately 10-5 changes per nucleotide site per replication cycle was observed (Parvin et al. 1986). Similar tissue culture experiments revealed mutation rates of about 10-fold lower for poliovirus and 10-fold higher for Rous sarcoma virus. Also, the evolution rate of influenza virus has been considered, this is in contrast to the mutation rate as it takes place when the viruses are passaged into humans. The evolution rate of the influenza A virus NS gene is 1.95 x 10-3 changes/site/year, several orders of magnitude greater than that of eukaryotic genes.
 
Doolittle et al. (1989) looked at the evolutionary rates of change of ten genes from retroviruses. Overall, the reverse transcriptase showed the slowest rate of change and the outer portion of the envelope protein the most rapid, evolving three times faster. The core portion of the gag protein changed about 1.6 times as fast as the transcriptase, the proteinase 1.8 times as fast, and the 140 amino acids at the amino terminal of gag 2.5 times as fast. The viral proteinase is pepsin-like, and that from HIV is as similar to that of visna virus as human pepsin is to its fungal homologue. The proteinases of HIV and HTLV-I differ from one another even more than human pepsin does from the fungal proteinase. In other words, the retroviruses are changing extraordinarily rapidly. Most species of RNA viruses actually consist of a population of genomes showing considerable variation around a master sequence (Domingo et al. 1988). The population concept is important, as in experimental systems, defective members of the genomic population can play a significant role in viral expression.
 
SELECTIVE PRESSURES AND CONSTRAINTS
 
It is of interest to determine, what, if any, limits are placed on virus variation. Despite high mutation rates and opportunities for genetic reassortment, many factors act to minimize emergence of new influenza A epidemics (Morse and Schluederberg 1988). even though avian and human influenza viruses are widespread (in humans an estimated 100 million infections yearly), pandemic influenza viruses emerge infrequently (every 10-40 years). Powerful constraints appear to exist since pandemic human influenza strains vary in their H gene, whereas the neuraminidase and most other genes are conserved.
 
These constraints on viral evolution are not surprising when one considers the selective pressures imposed by the host at each stage of the virus life cycle. Tissue tropism determinants, include site of entry, viral attachment proteins, host cell receptors, tissue- specific genetic elements (for example promoters), host cell enzymes (like proteinase), host transcription factors, and host resistance factors such as age, nutrition and immunity. Host factors contribute significantly: sequences such as hormonally responsive promoter elements and transcriptional regulatory factors can link viral expression to cell state.
 
The interaction of virus and host is thus complex but highly ordered, and can be altered by changing a variety of conditions. Unlike bacterial virulence, which is largely mediated by bacterial toxins and virulence factors, viral virulence often depends on host factors, such as cellular enzymes that cleave key viral molecules. Because virulence is multigenic, defects in almost any viral gene may attenuate a virus. For example, some reassortments of avian influenza viruses are less virulent in primates than are either parental strain, indicating that virulence is multigenic (Treanor and Murphy 1990).
 
Viral and host populations can exist in equilibrium until changes in environmental conditions shift the equilibrium and favour rapid evolution (Steinhauer and Holland 1987). It seems reasonable to expect that new viruses will emerge occasionally, but the stochastic and multifactorial nature of viral evolution makes it difficult to predict such events. According to Doolittle, retrovirus evolution is sporadic, with retroviruses evolving at different rates in different situations. For instance, the human endogenous retroviral element is shared with chimpanzees, indicating no change in over 8 million years, whereas strains of HIV have diverged in mere decades. Endogenous retroviruses carried in the germline evolve slowly compared with infective retroviruses. Generation of new viral pathogens is rare, and often possible only because of high mutation rates that permit many neutral mutations to accumulate before selective pressure forces a change. The seeming unpredictability of these events ensure that recognition of new pathogens must await their emergence.
 
CONCLUSION
 
The proposed American fiscal budget for 1995 allows allocations for the CDC which remain basically the same as those for past years and the $11.5 billion budget for the National Institutes of Health includes only a modest increase for non-AIDS infectious and immunological diseases research (Cassell 1994). In view of the magnitude of the problem, this budget is unacceptable. Currently, infectious diseases remain the leading cause of death worldwide. In the United States infectious diseases directly account for 3 and indirectly account for 5 of the 10 leading causes of death, AIDS is the ninth leading cause. Infectious diseases account for 25% of all visits to physicians in the United States. In total, the annual cost of AIDS and other infectious diseases reached $120 billion in 1992, about 15% of the nation's total health-care expenditure. The expanding pool of immunodeficient patients due to the AIDS epidemic, cancer treatment, transplant recipients, and hemodialysis has caused an explosion of opportunistic infections due to a number of fungal, parasitic, viral and bacterial agents.
 
According to the Gail H. Cassel, president of the American Society of Microbiology, the public health system is not prepared to meet the challenges of new and re-emerging infections. Perhaps the most obvious defect is inadequate disease surveillance and reporting. In America, only one-quarter of the states have a professional position dedicated to surveillance of food-borne and waterborne diseases. In 1992, only $55000 was spent on federal, state and local levels tracking drug-resistant bacterial and viral infections. In addition, the public health laboratories are eroding. Overall, CDC's budget for infectious diseases unrelated to AIDS has declined approximately 20% in the last decade. This is the case in the developed world of the United States, and we in developing South Africa are certainly no better off in terms of disease surveillance and concomitant protection. It should be clear that a mixture of basic and applied research related to infectious disease is needed. Coupled with this, better diagnostic techniques, prevention strategies and risk factor analysis is needed. Finally, enhanced communication among health care professionals and the public is integral in coming to terms and dealing with this issue. The American National Institute of Allergy and Infectious Diseases (NIAID) plans to develop a research and training infrastructure to elucidate the mechanisms of molecular evolution and drug resistance and to learn more about actual disease transmission through molecular and environmental studies and to continue their emphasis on vaccine development. For example, NIAID-funded research has already led to the creation of a new Haemophilus influenzae type B vaccine which is expected to save nearly $400 million in health-care costs each year. Similarly, the NIH spent less than $27 million dollars to find the connection between Helicobacter pylori and chronic peptic ulcers, yet using antibacterial therapy for the disease will save $760 million dollars in health care costs annually.
 
Given the diverse nature of threats from infectious diseases, it is not adequate merely to face each crisis as it emerges, as this may provide a strategy which proves to be too little and too late. Instead, a more holistic approach is required. This must include a global perspective as well as the need to address the issue of infectious disease within the context of shared environmental responsibility. Improved health care derived from socioeconomic betterment is crucial, as are long term policies involving systems thinking as opposed to the limiting nature of long term over-specialization.
 
REFERENCES
 
Cassel G.H. 1994. New and Emerging Infections in the Face of a Funding Crisis. ASM News 60, 5: 251-4
 
Culliton B.J. 1990. Emerging Viruses, Emerging Threat. Science 247: 279- 280
 
Diglisic G., Xiao S-Y., Gligic A., Obradovic M., Stojanovic R., Velimirovic D., Lukac V., Rossi C.A. and J.W. DeLuc. 1994. Isolation of a Puumala-like Virus from Mus musculus Captured in Yugoslavia and Its Association with Severe Hemorrhagic Fever with Renal Syndrome. J. Inf. Dis. 169: 204-7
 
Domingo E. and J.J. Holland. 1988. High error rates, population equilibrium and evolution of RNA replication systems. In: RNA genetics. Vol. 3. (eds. Domingo E., Holland J.J. and P. Ahlquist). Boca Raton, FL: CRC Press.
 
Doolittle R.F., Feng D.F., Johnson M.S. and M.A. McClure. 1989. Origins and evolutionary relationships of retroviruses. Q. Rev. Biol. 64: 1-30
 
Dowdle W. 1994. Dowdle Reflects on CDC, Changing Demands of Disease Surveillance. ASM News. 60, 5:237-8
 
Hahn C.S., Lustig S., Strauss E.G. and J.H. Strauss. 1988. Western equine encephalitis virus is a recombinant virus. Proc. Natl. Acad. Sci. USA. 85: 5997-6001
 
Hughes J.M. and J. R. La Montagne. 1994. The Challenges Posed by Emerging Infectious Diseases. ASM News 60, 5: 248-50
 
Hunter S. 1991. Tropical Medicine. 7th edn. W.B. Saunders Company
 
Kida H., Shortridge K.F. and R.G. Webster. 1988. Origin of the hemagglutinin gene of H3N2 influenza viruses from pigs in China. Virology. 162: 160-166
 
Kilbourne E.D., Easterday B.C. and S. McGregor. 1988. Evolution to predominance of swine influenza virus hemagglutinin mutants of predictable phenotype during single infections of the natural host. Proc. Natl. Acad. Sci. USA. 85: 8098-8101
 
Lederburg J. 1994. Emerging Infections: Private Concerns and Public Responses. ASM News. 60, 5: 233
 
Manson-Bahr D.E.C. and D.R. Bell. 1989. Manson's Tropical Diseases. 19th edn. Bailliere Tindall.
 
May R.M. and R.M. Anderson. 1987. Transmission dynamics of HIV infection. Nature. 326: 137-142
 
Morse S.S. (ed.). 1993. Emerging Viruses. Oxford University Press.
 
Morse S.S. and A. Schluederberg. 1990. Emerging Viruses: The Evolution of Viruses and Viral Diseases. J. Inf. Dis. 162: 1-7
 
Murphy F.A. 1994. Infectious Diseases. Adv.Vir. Res. 43: 2-52
 
Parvin J.D., Moscona A., Pan W.T., Leider J.M. and P. Palese. 1986. Measurement of the mutation rates of animal viruses: influenza A virus and poliovirus type 1. J. Virol. 62: 3084-3091
 
Peters C.J. 1994. Molecular Techniques Identify a New Strain of Hantavirus. ASM News. 60, 5: 242-3
 
Steinhauer D. and J.J. Holland. 1987. Rapid evoltuion of RNA viruses. Annu Rev. Microbiol. 41: 409-433
 
Treanor J. and B. Murphy. Genes involved in the restriction of replication of avian influenza A viruses in primates. In: Applied virology research: virus variation and epidemiology. Vol. 2. New York: Plenum Press (eds. Kurstak E., Marusyk R.G., Murphy F.A. and M.H.V. van Regenmortel.





SIGHTINGS HOMEPAGE