Posts Tagged ‘bacteriophage’

An examination of the bacteriophages and bacteria of the Namib desert

30 April, 2012

See on Scoop.itVirology News and here

“Bacteria and their viruses (called bacteriophages, or phages), have been found in virtually every ecological niche on Earth. Arid regions, including their most extreme form called deserts, represent the single largest ecosystem type on the Earth’s terrestrial surface. The Namib desert is believed to be the oldest (80 million years) desert. We report here an initial analysis of bacteriophages isolated from the Namib desert using a combination of electron microscopy and genomic approaches. The virus-like particles observed by electron microscopy revealed 20 seemingly different phage-like morphologies and sizes belonging to the Myoviridae and Siphoviridae families of tailed phages. Pulsed-field gel electrophoresis revealed a majority of phage genomes of 55-65 kb in length, with genomes of approximately 200, 300, and 350 kb also observable. Sample sequencing of cloned phage DNA fragments revealed that approximately 50% appeared to be of bacterial origin. Of the remaining DNA sequences, approximately 50% displayed no significant match to any sequence in the databases. The majority of the 16S rDNA sequences amplified from DNA extracted from the sand displayed considerable (94-98%) homology to members of the Firmicutes, and in particular to members of the genus Bacillus, though members of the Bacteroidetes, Planctomycetes, Chloroflexi, and delta-Proteobacteria groups were also observed.”

This serves as a neat, if slightly dated, little introduction to my latest endeavour – and an account of a field trip this last week into the Namib Desert.

I was fortunate enough some time ago to have been invited by Don Cowan, presently of University of Pretoria, to accompany his team to the Gobabeb Research and Training Centre inland of Walvis Bay, in Namibia’s Namib Desert.  They work on extremophiles, and the Namib is a great environment for mining bugs that can withstand high salt and temperatures and severe desiccation – oh, and photosynthesise underground, hiding under semi-tranlucent quartz rocks embedded in the surface soil.  The thinking was that, given my long-time interest in viral diversity and newly-acquired means to do oceanic viromics, I would be interested and even of some help.

And so it has come to pass: I will have my very own hypolith (=rock-colonising blue-green algae) scrapings and the result of diafiltration and concentration of washings of a good few kilos of red dune sand to play with as far as virus genome sequencing and even EM and analytical centrifugation go.

Typical quartz-associated hypolith

We will have fun in the coming months…that, and we will obviously HAVE to go back to Gobabeb, to further investigate whatever it is we find.  A terrible, harsh place, but SOMEONE has to go there…B-)

Nothing beside remains...boundless and bare
The lone and level sands stretch far away
PB Shelley, Ozymandias

A Short History of the Discovery of Viruses – Part 2

7 February, 2012

The following text has now appeared in modified form in an ebook, for sale for US$4.99 on the iBooks Store

The Ultracentrifuge, Eggs and Flu

The ultracentrifuge

A technical development that was to greatly advance the study of viruses was begun in 1923, but only reached fruition by the 1930s: this was the ultracentrifuge, invented and developed first by Theodor (“The”) Svedberg in Sweden as a purely analytical tool, and later by JW Beams and EG Pickels in the USA as an analytical and preparative tool.  The ultracentrifuge revolutionised first, the physical analysis of proteins in solution, and second, the purification of proteins, viruses and cell components, by allowing centrifugation at speeds high enough to allow pelleting of subcellular fractions.

Analytical centrifugation and calculation of molecular weights of particles gave some of the first firm evidence that certain proteins, and virus particles, were large, regular objects.  Indeed, it came to be taken as a given that one of the fundamental properties of a virus particle was its sedimentation coefficient, measured in svedbergs (a unit of 10-13 seconds, shown as S20,W).  This is also how ribosomes of pro- and eukaryotes came to be named: these are known as 70S (prokaryote) and 80S ribosomes, respectively, based on their different sedimentation rates.

The Official Discovery of Influenza Virus

In 1931, Robert Shope in the USA managed to recreate swine influenza by intranasal administration of filtered secretions from infected pigs.  Moreover, he showed that the classic severe disease required co-inoculation with a bacterium – Haemophilus influenza suis – originally thought to be the only agent.  He also pointed out the similarities between the swine disease and the Spanish Flu, where most patients died of secondary infections.  However, he also suggested that the virus survived seasonally in a cycle involving the pig, lungworms, and the earthworm, which is now known to be completely wrong.

This notwithstanding, he found that people who had survived infection during the 1918 pandemic had antibodies protecting them against the swine flu virus, while people born after 1920 did not, which showed that the 1918 human and swine flu viruses were very similar if not identical. This was a very relevant discovery for what happened much later, in the 2009 influenza pandemic, when the same virus apparently came back into the human population from pigs after circulating in them continuously since 1918.

Shope went on in 1932 to discover, with Peyton Rous, what was first called the Shope papillomavirus and later Cottontail rabbit papillomavirus: this causes benign cancers in the form of long hornlike growths on the head and face of the animal. This may explain the sightings in the US Southwest of the near-mythical “jackalope”.

Influenza viruses in pigs

Influenza viruses in pigs

Patrick Laidlaw and William Dunkin, working in the UK at the National Institute for Medical Research (NIMR), had by 1929 successfully characterised the agent of canine distemper – a relative of measles, mumps and distemper morbilliviruses – as a virus, proved it infected dogs and ferrets, and in 1931 got a vaccine into production that protected dogs.  This was made from chemically inactivated filtered tissue extract from infected animals.  Their work built on and completely eclipsed earlier findings, such as those of Henri Carré in France in 1905, who first claimed to have shown it was a filterable agent, and Vittorio Puntoni, who first made a vaccine in Italy from virus-infected brain tissue inactivated with formalin in 1923.

Influenza and Ferrets: the Early Days

Continuing from Laidlaw and Dunkin’s work in the same institute, Christopher Andrewes, Laidlaw and W Smith reported in 1933 that they had isolated a virus from humans infected with influenza from an epidemic then raging.  They had done this by infecting ferrets with filtered extracts from infected humans – after the fortuitous observation that ferrets could apparently catch influenza from infected investigators!  The “ferret model” was very valuable – see here for modern use of ferrets – as strains of influenza virus could be clinically distinguished from one another.

Eggs and Flu and Yellow Fever

Influenza virus and eggs: large-scale culture

Frank Macfarlane Burnet from Australia visited the NIMR in the early 1930s, and learned a number of techniques he used to great effect later on.  Principal among these was the technique of embryonated egg culture of viruses – which he took back to Melbourne, and applied to the infectious laryngotracheitis virus of chickens in 1936.  This is a herpesvirus, first cultivated by JR Beach in the USA in 1932: Burnet used it to demonstrate that it was possible to do “pock assays” on chorioallantoic membranes that were very similar to the plaque assays done for bacteriophages, with which he was also very familiar.  Also in 1936, Burnet started a series of experiments on culturing human influenza virus in eggs: he quickly showed that it was possible to do pock assays for influenza virus, and that

“It can probably be claimed that, excluding the bacteriophages, egg passage influenza virus can be titrated with greater accuracy than any other virus.”

Max Theiler and colleagues in the USA took advantage of the new method of egg culture to adapt the French strain of yellow fever virus (YFV) he had grown in mouse brains to being grown in chick embryos, and showed that he could attenuate the already weakened strain even further – but it remained “neurovirulent”, as it caused encephalitis or brain inflammation in monkeys.  He then adapted the first YFV characterised – the Asibi strain, from Ghana in 1927 – to being grown in minced chicken embryos lacking a spinal cord and brain, and showed in 1937 that after more than 89 passages, the virus was no longer “neurotrophic”, and did not cause encephalitis.   The new 17D strain of YFV was successfully tested in clinical trials in Brazil in 1938 under the auspices of the Rockefeller Foundation, which has supported YFV work since the 1920s.  The strain remains in use today, and is still made in eggs.

Virus purification and the physicochemical era

Given that the nature of viruses had prompted people to think of them as “chemical matter”, researchers had attempted from early days to isolate, purify and characterise the infectious agents.  An early achievement was the purification of a poxvirus in 1922 by FO MacCallum and EH Oppenheimer. 

Much early work was done with bacteriophages and plant viruses, as these were far easier to purify or extract at the concentrations required for analysis, than animal or especially human viruses. 

CG Vinson and AM Petre, working with the infectious agent causing mosaic disease in tobacco – tobacco mosaic virus, or TMV – showed in 1931 that they could precipitate the virus from suspension as if it were an enzyme, and that infectivity of the precipitated preparation was preserved.  Indeed, in their words:

“…it is probable that the virus which we have investigated reacted as a chemical substance”.

Viruses in Crystal

An important set of discoveries started in 1935, when Wendell Stanley in the USA published the first proof that TMV could be crystallised, at the time the most stringent way of purifying molecules.  He also reported that the “protein crystals” were contaminated with small amounts of phosphorus.  An important finding too, using physical techniques including ultracentrifugation and later, electron microscopy, was that the TMV “protein” had a very high molecular weight, and was in fact composed of large, regular particles.  This was a very significant discovery, as it indicated that some viruses at least really were very simple infectious agents indeed.

TMV particle: 95% protein, 5% RNA

However, his conclusion that TMV was composed only of protein was soon challenged, when Norman Pirie and Frederick Bawden working in the UK showed in 1937 that ribonucleic acid (RNA) – which consists of ribose sugar molecules linked by phosphate groups – could be isolated consistently from crystallised TMV as well as from a number of other plant viruses, which accounted for the phosphorus “contamination”.  This resulted in the realisation that TMV and other plant virus particles – now known to be virions – were in fact nucleoproteins, or protein associated with nucleic acid.

Stanley received a share of the Nobel Prize in Chemistry in 1946 for his work on TMV: it is instructive to read his acceptance speech from the time to realise what the state of the science that was becoming virology was at the time.  He wrote:

“Since the original discovery of this infectious, disease-producing agent, known as tobacco mosaic virus, well over three hundred different viruses capable of causing disease in man, animals and plants have been discovered. Among the virus-induced diseases of man are smallpox, yellow fever, dengue fever, poliomyelitis, certain types of encephalitis, measles, mumps, influenza, virus pneumonia and the common cold. Virus diseases of animals include hog cholera, cattle plague, foot-and-mouth disease of cattle, swamp fever of horses, equine encephalitis, rabies, fowl pox, Newcastle disease of chickens, fowl paralysis, and certain benign as well as malignant tumors of rabbits and mice. Plant virus diseases include tobacco mosaic, peach yellows, aster yellows, potato yellow dwarf, alfalfa mosaic, curly top of sugar beets, tomato spotted wilt, tomato bushy stunt, corn mosaic, cucumber mosaic, and sugar cane yellow stripe. Bacteriophages, which are agents capable of causing the lysis of bacteria, are now regarded as viruses”.

Two of the most interesting things about the article, however, are the electron micrographs of virus particles – Stanley had one of the first electron micrsoscopes available at the time –  and the table of sizes of viruses, proteins and cells that had been determined by then by techniques such as ultracentrifugation and filtration: TMV was known to be rodlike, 15 x 280 nm; vaccinia was 210 x 260 nm; poliomyelitis was 25 nm; phages like T2 were known to have a head-and-tail structure.

Seeing is Believing: the Electron Microscope

First Electron Microscope with Resolving Power Higher than that of a Light Microscope. Ernst Ruska, Berlin 1933 Wikipedia CC BY-SA 3.0, https://www.flickr.com/photos/93452909@N00/176059674

First Electron Microscope with Resolving Power Higher than that of a Light Microscope. Ernst Ruska, Berlin 1933
Wikipedia CC BY-SA 3.0, https://www.flickr.com/photos/93452909@N00/176059674

The development of the electron microscope, in Germany in the 1930s, represented a revolution in the investigation of virus structures: while virions of viruses like variola and vaccinia could just about be seen by light microscopy – and had been, as early as 1887 by John Buist and others – most viruses were far too small to be visualised in this way. 

While Ernst Ruska received a Nobel Prize in 1986 for developing the electron microscope, it was his brother Helmut who first imaged virus particles – using beams of electrons deflected off virus particles coated in heavy metal atoms.  From 1938 through the early 1940s, using his “supermicroscope”, he imaged virions of poxviruses, TMV, varicella-zoster herpesvirus, and bacteriophages, and showed that they were all particulate – that is, they consisted of regular and sometimes complex particles, and were often very different from one another.  He even proposed in 1943 a system of viral classification on the basis of their perceived structure.

While electron microscopy was also used medically to some extent thereafter – for example, in differentiating smallpox from chickenpox by imaging particles of variola virus and varicella-zoster virus, respectively, derived from patients’ vesicles – its use was limited by the expense and cumbersome nature of sample preparation. For example, the micrographs in Stanley’s 1946 paper were all done with samples “…prepared with gold by the shadow-casting technique”.

The use of the cumbersome technique of metal shadow-casting, and the highly inconvenient nature of electron microscopy as a routine tool all changed from 1959 onwards, when Sydney Brenner and Robert Horne published “A negative staining method for high resolution electron microscopy of viruses”.  This method involves the use of viruses in liquid samples deposited on carbon-coated metal grids, and then stained with heavy-metal salts such as phosphotungstic acid (PTA) or uranyl acetate.

This simple technique revolutionised the field of electron microscopy, and within just a few years much information was acquired about the architecture of virus particles. Not only were the overall shapes of particles revealed, but also the details of the symmetrical arrangement of their components. Some beautiful examples can be seen here, at the Cold Spring Harbor site.

Depiction of the effects of using a heavy metal salt solution to negatively stain particles on a carbon film. The stain (dark) pools around the particles (light).  Human rotavirus particles, stained from below (left) and by immersion (right).
Images copyright LM Stannard

Depiction of the effects of using a heavy metal salt solution to negatively stain particles on a carbon film. The stain (dark) pools around the particles (light). Human rotavirus particles, stained from below (left) and by immersion (right).
Images copyright LM Stannard

Click here for Part 1: Filters and Discovery

here for Part 3: Phages, Cell Culture and Polio

and here for Part 4: RNA Genomes and Modern Virology

Copyright Edward P Rybicki and Russell Kightley, February 2015, except where otherwise noted.

A Short History of the Discovery of Viruses – Part 1

6 February, 2012

The following text has now appeared in modified form in an ebook, for sale for US$4.99 on the iBooks Store

A Short History of the Discovery of Viruses

While people were aware of diseases of both humans and animals now known to be caused by viruses many hundreds of years ago, the concept of a virus as a distinct entity dates back only to the very late 1800s.  Although the term had been used for many years previously to describe disease agents, the word “virus” comes from a Latin word simply meaning “slimy fluid”.

Porcelain filters and the discovery of viruses

The invention that allowed viruses to be discovered at all was the Chamberland-Pasteur filter.  This was developed in 1884 in Paris by Charles Chamberland, who worked with Louis Pasteur.  It consisted of unglazed porcelain “candles”, with pore sizes of 0.1 – 1 micron (100 – 1000 nm), which could be used to completely remove all bacteria or other cells known at the time from a liquid suspension.  Though this simple invention essentially enabled the establishment of a whole new science – virology – the continued development of the discipline required a string of technical developments, which I will highlight as appropriate.

Pasteur Germ Proof Filter, c. 1890, Pasteur-Chamberland Filter Co., Dayton, Ohio – Museum of Science and Industry (Chicago)

 

As the first in what was to be an interesting succession of events, Adolf Eduard Mayer from Germany, publishing in 1886 on work done in Holland from 1879, showed that the “mosaic disease” of tobacco – or “mozaïkziekte”, as he named it in his paper – could be transmitted to other plants by rubbing a liquid extract, filtered through paper, from an infected plant onto the leaves of a healthy plant.  However, he came to the erroneous conclusion that it must be a bacterial disease.

The first use of porcelain filters to characterize what we now know to be a virus was reported by Dmitri Ivanovski in St Petersburg in Russia, in 1892.  He had used a filter candle on an infectious extract of tobacco plants with mosaic disease, and shown that it remained infectious: however, he concluded the agent was probably a toxin as it appeared to be soluble.

The Dutch scientist Martinus Beijerinck in 1898 reported similar experiments with bacteria-free filtered extracts, but made the conceptual leap and described the agent of mosaic disease of tobacco as a “contagium vivum fluidum”, or contagious living fluid, because he was convinced the infectious agent had a liquid nature. The extract was completely sterile, could be kept for years, but remained infectious.  The term virus was later used to describe such fluids, also called “filterable agents”, which were thought to contain no particles.  The virus causing mosaic disease is now known as Tobacco mosaic virus (TMV).  A paper commemorating Ivanovsky and Beijerinck’s work – “One Hundred Years of Virology” – was published in Journal of Virology 1992 to honour both pioneers.

The first animal viruses

The second virus discovered was what is now known as Foot and mouth disease virus (FMDV) of farm and other animals, in 1898 by the German scientists Friedrich Loeffler and Paul Frosch.  Again, their “sterile” filtered liquid proved infectious in calves, providing the first proof of viruses infecting animals – a fact commemorated by an article in 1998 in the Journal of General Virology.  Indeed, some believe that the true discoverers of viruses were these two scientists, as they concluded the infectious agent was a tiny particle, and was not a liquid agent.  The two went further by showing that it was possible to vaccinate cows and sheep against the disease using filtered vesicle extract that had been heated sufficiently to destroy its infectivity: this was possibly the first use of an inactivated virus as a prophylactic vaccine.

In 1898 G Sanarelli, working in Uruguay, described the smallpox virus relative and tumour-causing myxoma virus of rabbits as a virus – but on the basis of sterilisation by centrifugation rather than by filtration.

The first human virus: yellow fever

The first human virus described was the agent which causes yellow fever: this probably originated in Africa, but was spread along with its mosquito vector Aedes aegyptii to the Americas and neighbouring islands by the slave trade.  Indeed, the  declaration of independence from France by Haiti in 1804 was made possible in part by the devastating effect of the disease on the French army sent to quell a slave revolt there. The virus was discovered and reported in 1901 by the US Army physician Walter Reed, after pioneering work in Cuba by Carlos Finlay reported in 1881 hypothesising that mosquitoes transmitted the deadly disease

The agent became the subject of intense study because, in the Spanish-American war in Cuba in the 1890s, about 13 times as many soldiers died of yellow fever as died from wounds. The subsequent eradication of mosquitoes in Panama is probably what allowed the completion of the Panama Canal – stalled because of the death rate among workers from yellow fever and malaria.

Rinderpest and rabies

The paper describing rinderpest as a virus disease

A finding that was later to have great importance in veterinary virology was the discovery by Maurice Nicolle and Adil Mustafa (also known as Adil-Bey), in Turkey in 1902, that rinderpest or cattle plague was caused by a virus.  This had been for several centuries the worst animal disease known worldwide in terms of mortality: for example, an epizootic or animal epidemic in Africa in the 1890s that had started in what is now Ethiopia in 1887 from cattle imported from Asia, had spread throughout the continent by 1897, and killed 80-90% of the cattle and a large proportion of susceptible wild animals in southern Africa.  Many thousands of people died of starvation as a result.  The virus is, incidentally, only the second to have ever been eradicatednearly 100 years after its discovery.

Viruses and Vaccines

Sir Arnold Theiler, a Swiss-born veterinarian working in South Africa, had been appointed as state veterinarian for the Zuid-Afrikaansche Republiek prior to 1899, on the strength of his having produced a smallpox vaccine for miners in the Johannesburg area.  He then developed a crude vaccine against rinderpest by 1897, without knowledge of the nature of the agent: this consisted of blood from an infected animal, injected with serum from one that had recovered – something also shown to work with FMDV by Loeffler and Frosch.  This risky mixture worked well enough, however, to eradicate the disease in the region.  He went on to do the same thing successfully for African horsesickness virus and other disease agents, in an institute (Onderstepoort Veterinary Institute) that still works on the virus.

The description in Annales de l’Institut Pasteur by Remlinger and Riffat-Bay in 1903 of the agent of rabies as a “filterable virus was the culmination of many years of distinguished work in France on the virus, started by Louis Pasteur himself. While Remlinger credited Pasteur with having the notion in 1881 that rabies virus was an ultramicroscopic particle, the fact is that Pasteur and Emile Roux had also, in 1885, effectively made a vaccine against rabies by use of dried infected rabbit spinal cords, without any knowledge of what the agent was.

Title page of the original article in Annales de l’Institut Pasteur Volume 17 of 1903

It is interesting that the same volume of the Annales which reported the rabies agent also has a discussion on whether or not the smallpox agent variola virus and the vaccine against it, vaccinia virus, were differently-adapted variants of the same thing, or were different viruses.

More and more viruses

The viral nature of many disease agents started to be made evident around this time, as more researchers started investigating known diseases.  In 1904, E Baur in Germany described an infectious variegation of Abutilon that could only be transmitted by grafting, that was not associated with visible bacteria.  This is now known to be due to Abutilon mosaic virus, now known to be a single-stranded DNA geminivirus.

Abutilon mosaic

Incidentally, the earliest recorded description of a plant disease was probably in a poem in 752 CE by the Japanese Empress Koken, describing symptoms in eupatorium plants.  It was shown in 2003 that the striking yellow-vein symptoms were caused by a geminivirus infection.

Interestingly, also in 1906, A Zimmermann proposed – in a paper entitled “Die Krauselkrankheit des Maniok” – that the agent of mosaic disease of cassava that had first been described from German East Africa (now Tanzania) in 1894, was a filterable virus.  This was the second geminivirus discovered, although this was only proved in the 1970s.

Cassava affected by a recombinant African cassava mosaic virus in western Kenya, 1997

Cassava affected by a recombinant African cassava mosaic virus in western Kenya, 1997.  Insets, from left: healthy cassava, mild disease, severe disease

In 1906, Adelchi Negri – who had previously discovered the Negri bodies in cells infected with rabies virus – showed that vaccinia virus, the vaccine for the dreaded smallpox caused by variola virus, was filterable.  This was the final step in a long series of discoveries around smallpox, that started with Edward Jenner’s use of what was supposedly cowpox, but may have been horsepox virus to protect people from the disease in 1796.

An Egyptian stele thought to represent a polio victim (1403–1365 BC). Note the characteristic withering of one leg.

An Egyptian stele thought to represent a polio victim (1403–1365 BC). Note the characteristic withering of one leg.

The disease now known as poliomyelitis was first clinically described in England in 1789, as “a debility of the lower extremities”.  However, it had been known since ancient times, and had even been depicted clearly in an Egyptian painting from over 3000 years ago.

An important development in human virology in 1908, therefore, was the finding by Karl Landsteiner and Erwin Popper in Germany that poliomyelitis or infantile paralysis in humans as it was known then, was caused by a virus: they proved this by injecting a cell-free extract of a suspension of spinal cord from a child who had died of the disease, into monkeys, and showing that they developed symptoms of the disease.

Viruses and cancer

In 1908, Oluf Bang and Vilhelm Ellerman in Denmark were the first to associate a virus with leukaemia: they successfully used a cell-free filtrate from chickens with avian leukosis to transmit the disease to healthy chickens.

The first solid tumour-causing virus, or virus associated with cancer, was found by Peyton Rous in the USA in 1911.  He showed that chicken sarcomas, or solid connective tissue tumours, could be transmitted by grafting, but also that a filterable or cell-free agent extracted from a sarcoma was infectious.  The virus was named for him as Rous sarcoma virus, and is now known to be a “retrovirus”,as is chicken leukaemia virus,in the same virus family as HIV.

Eaters of Bacteria: The Phages

"Twort" by Obituary Notices of Fellows of the Royal Society, Vol. 7, No. 20. (Nov., 1951), pp. 504-517.. Licensed under Public Domain via Wikimedia Commons - http://commons.wikimedia.org/wiki/File:Twort.jpg#mediaviewer/File:Twort.jpg

“Twort” by Obituary Notices of Fellows of the Royal Society, Vol. 7, No. 20. (Nov., 1951), pp. 504-517.. Licensed under Public Domain via Wikimedia Commons – http://commons.wikimedia.org/wiki/File:Twort.jpg#mediaviewer/File:Twort.jpg

Two independent investigations led to the important discovery of viruses that infect bacteria. In 1915, Frederick Twort in the UK accidentally found a filterable agent that caused the bacteria he was growing to lyse, or burst open.  Although he showed that it could pass through porcelain filters, and could be transmitted to other colonies of the same bacteria, he was not sure whether or not it was a virus, and referred to it as “the bacteriolytic agent”.  It is interesting that he was actually attempting to grow vaccinia virus in culture, and that it was a contaminating staphylococcus that he noticed was being lysed by his infectious agent.

www_sciencedirect_com_science__ob_PdfExcerptURL__imagekey_1-s2_0-S0140673601203833-main_pdf__piikey_S0140673601203833__cdi_271074__orig_article__zone_centerpane__fmt_abst__eid_1-s2_0-S0140673601203833__user_635696_md5_610868932f41b70483e225

The original article published by Twort in The Lancet in 1915

Subsequently, Félix d’Hérelle in Paris published in 1917 that he had discovered a virus that lysed a bacterial agent he was culturing in liquid broth – a Shigella – that caused human dysentery, or diarrhoea.  He named the virus “bacteriophage”, or eater of bacteria, derived from the Greek term “phagein”, meaning to eat.  He showed a number of interesting properties of his shigella-specific bacteriophages, including that they could be adapted to other Shigella species or types by passaging them repeatedly, and that they protected rabbits against infection by lethal doses of bacteria

D’Hérelle’s main interest in his new discovery was in using them as a therapeutic agent for bacterial infections in humans: sadly, this idea did not take off in Europe or the Americas, largely due to the unreliability of the ill-understood phage preparations, although it was extensively exploited in the former USSR. Indeed, he mentored George Eliava who went on to found the Eliava Institute in Tbilisi, Georgia, which became a major centre for the use of bacteriophage cocktails against persistent bacterial infections in humans.  A review on phage therapy from the Institute was recently published to mark the centenary of Twort’s discovery in 1915. An excellent – if slightly childish – animation describing phages and phage therapy can be seen here.

D‘Hérelle’s main interest in his new discovery was in using them as a therapeutic agent for bacterial infections in humans: sadly, this idea did not take off in Europe or the Americas, although it was extensively exploited in the former USSR.

The 1896 paper from Annales de l’Institut Pasteur

Interestingly, and as reported in ViroBlogy previously, what could have been the first discovery of phages was probably described by Ernest Hankin, who had previously proved in India that cholera was caused by bacteria.  In 1896 in Annales de l’Institut Pasteur, he documented that river water downstream of cholera-infested towns on the Jumma river in India contained no viable Cholera vibrio – and that this was a reliable property of the water, and was probably responsible for limiting the spread of cholera

While he did not prove the presence of a “filterable agent”, he was recognised by d’Hérelle and others as having contributed to the discovery of bacteriophages.  In fact, d’Hérelle went to India in 1927, and put cholera phage preparations into wells in villages with cholera patients: apparently the death toll went down from 60% to 8%.

Influenza A viruses in waterbirds – Russell Kightley Media

The virus as human plague: the Spanish Flu

Possibly the worst human plague the world has ever seen swept across the planet between 1918 and 1922: this was known as the Spanish Flu, from where it was first properly reported, and it went on to kill more than 50 million people all over the world.  We now know it to have been H1N1 influenza type A: modern reconstruction of the virus from archived tissue samples and frozen bodies found in permafrost has shown it probably jumped directly into humans from birds.

Most medical authorities at the time thought the disease was caused by bacteria – however, MJ Dujarric de la Rivière, and Charles Nicolle – brother of Maurice – and Charles  Lebailly in France, separately proposed in 1918 that the causative agent was a virus, based on properties of infectious extracts from diseased patients.  Specifically, they found that the infectious agent derived from bronchial expectoration of an infected person was filterable, caused disease in monkeys via nasal administration and human volunteers via subcutaneous injection, and was not present in the blood of an infected monkey.  However, many scientists at the time still doubted that influenza was a viral disease – despite this contemporary comment in the British Medical Journal of 1918.

Conclusions from the Nicolle and Lebailly paper

Translation of this passage (courtesy of Mrs Francoise Williamson):

“Conclusions.

1⁰ The bronchial  expectoration of people suffering from flu, collected during the acute period, is virulent.
2⁰   The monkey (M. cynomolgus)  is sensitive to the virus  by sub-conjunctival and nasal inoculation.
3⁰   The flu agent is a filterable organism.  The inoculation  of the filtrate has indeed reproduced the illness in two of the people injected subcutaneously;  on the other hand when given intravenously it  appears to be ineffective. (two failures out of two tries).
4⁰ It is possible that the influenza virus does not occur in the patient’s blood.  The blood of a monkey with influenza, inoculated subcutaneously, did not infect man;  the negative blood result of subject 2 at D, is however, not convincing, the blood route seeming to be ineffective for  the flu virus transmission.”

Other agents of other diseases were found to be “filterable viruses” in the 1920s, including yellow fever virus by Adrian Stokes in 1927, in Ghana.  Indeed, the US bacteriologist and virologist Thomas Rivers in 1926 counted some sixty-five disease agents that had been identified as viruses.

Virus Assays: Counting the Viruses in the 1920s

The discovery of bacteriophages was a landmark in the history of virology, as it meant that for the first time it was relatively easy to work with viruses: many kinds of bacteria could be grown in solid or liquid culture quite easily, and the life cycle of the viruses could be studied in detail.  In fact, this later led to the birth of molecular biology, as described here

However, the beauty of working with phages was that they could be assayed – or counted in terms of infectious units – so easily, either by the plaque technique or by infections of liquid cultures.  This was not true of viruses of plants or of animals in the absence of similar culture techniques; these could only be assayed in a much more crude method using whole organisms.  One such method was by determining infection endpoints by serial dilution of inoculum, such as the now-famous ID50, or dose infecting 50% of the experimental subjects.

This changed in 1929 for plant viruses, with the demonstration by the plant virus pioneer FO Holmes that local lesions caused by infection of particular types of tobacco by TMV could be used as a means of assaying the infectivity of virus stocks.  This was then extended to other virus/host combinations, and allowed the rapid and quantitative assay of virus stocks – which, as it had done for phages, allowed the study of the properties of plant viruses, and led to their biological isolation and then purification.

TMV-induced local lesions in N. tabacum cv. glutinosa

TMV-induced local lesions in N. tabacum cv. glutinosa

Eggs and animals for virus culture

Chicken eggs for virus growth and assay

Possibly the next most important methodological development in virology after the discovery of phages was the proof that embryonated or fertilized hen’s eggs could be used to culture a variety of important animal and human viruses.  Ernest Goodpasture, working at Vanderbilt University in the USA, showed in 1931 that it was possible to grow fowlpox virus – a relative of smallpox – by inoculating the chorioallantoic membrane of eggs, and incubating them further. 

egg

While tissue culture had in fact been practiced for some time – for example, as early as the 1900s, investigators had grown “vaccine virus” or the smallpox vaccine now called vaccinia virus in minced up chicken embryos suspended in chicken serum – this technique represented a far cheaper and much more “scalable” technique for growing pox- and other suitable viruses.

An important first in a chain of related discoveries was the one by Howard Andervost, at Harvard University in 1929, who showed that human herpes simplex virus could be cultured by injection into the brains of live mice.

This led to the demonstration in 1930 by the South African-born Max Theiler – son of Sir Arnold – also at Harvard, that yellow fever virus could be similarly cultured: this allowed much easier handling of the virus, which until then had to be injected into monkeys in order to multiply it in their livers.  In addition, it allowed the development of attenuated or weakened strains of virus, by him and in parallel by a French laboratory, by serial passage or repeated transmission of the virus between mice. He also incidentally caught yellow fever from one of his mice through a laboratory accident. Culturing in mouse brains also allowed the successful animal testing of vaccine candidates, and of protective antisera, for which Theiler was awarded the Nobel Prize in 1951.  

Until 2008, this was the first and only recognition of virus vaccine work by the Nobel Foundation.

A consequence of this work was the landmark in medical virology that was the development of human vaccines against yellow fever virus, by Wilbur Sawyer in the USA in 1931: this followed on Theiler’s mouse work in using brain-cultured virus plus human immune serum from recovered patients to immunize humans – very similar to Theiler Senior’s strategy with rinderpest, more than thirty years later.

here for Part 2: The Ultracentrifuge, Eggs and Flu

here for Part 3: Phages, Cell Culture and Polio

and here for Part 4: RNA Genomes and Modern Virology

Copyright Edward P Rybicki and Russell Kightley, February 2012, except where otherwise noted.

Virology Africa 2011: viruses at the V&A Waterfront 2

19 December, 2011

We thank Russell Kightley for permission to use the images

Marshall Bloom (Rocky Mountain Laboratories, NIAID) opened the plenary session on Thursday the 1st of December, with a talk on probing the pathogen-vector-host interface of tickborne flaviruses.   Although thoroughly infected with a rhinovirus, he held our attention most ably while reminding us that while many flaviviruses are tick borne, the hard and soft body ticks that vector them are very phylogenetically different – as different as they are from spiders – meaning that if similar flaviruses replicated in them, these viruses may have much wider host range than we know.

He pointed out that while about 95% of the virus life cycle takes place in a tick, transmission to a vertebrate means suddenly adapting to a very different host.  Infection in ticks is persistent, as befits their vector role – but vertebrate infection generally is not.  It was interesting, as a sometime plant virologist, to hear that they look for dsRNA as a marker for replication, and do Ab staining for it: the technique was invented with plant viruses, and very few other virologists seem to appreciate that dsRNA can be quite easily isolated and detected.

They compared Vero and tick cells for virus replication, and saw significant differences: while tick cells could go out to 60+ days and look fine, Vero cells were severely affected at much shorter times post infection.  There was also 100-fold less virus in tick cells, and prominent tubular structures in old infected tick cells.  He noted that ticks evade host defences quite efficiently: eg they suppress host clotting during feeding, and there is huge gene activation in the tick during feeding.  In another study to envy, they are doing array work on ticks to see what is regulated and how.

 Linda Dixon (Institute for Animal Health, Pirbright, UK) recounted her lab’s work on African swine fever (ASFV), a poxvirus-like large DNA virus.  The virus is endemic to much of Africa, and keeps escaping – and there is no effective  vaccine to prevent spread, so regulation is by slaughter.  There are 3 types of isolate, with the most highly pathogenic causing up to 10% fatality and a haemorrhagic syndrome.  She described how in 2007 the virus had spread from Africa to Georgia, then in 2009 to southern Russia and all way to the far north, in wild boar.

There are more than 50 proteins in the dsDNA-containing virion; two infectious forms similar to the poxviruses with multilayer membranes and capsid layers can form, and neutralising Ab play no part in protection as a result.  They studied the interaction of viruses with cells and the immune system, and compared the genomes of pathogenic and non-pathogenic strains, in order to understand how to develop an effective vaccine.

The biggest differences were large deletions in non-virulent isolates, including genes coding for  proteins responsible for binding to RBC, and various immune evasion multicopy genes.  They planned to target regions to delete to make an attenuated virus for vaccine.  They had found non-essential genes involved in immune evasion, and ones that lower virulence, and had been systematically cutting them out.  She noted that pigs can be protected if they survive natural infection and if vaccinated with TC-attenuated virus, and can be protected by passive transfer of Abs from immune pigs – which indicated that an effective live vaccine was very possible.

Subunit vaccines were being investigated, and they had found partial protection with baculovirus-expressed proteins.  They were doing genome-wide screens for protective Ag, and were pooling Ags expressed from predicted ORFs in immunization trials – up to 47 Ags without reduction in specific  T cell responses.

Discovery One

My former labmate Dion du Plessis (Onderstepoort Veterinary Institute, OVI) made a welcome return to Cape Town, with a talk entitled “2011: A Phage Odyssey”.  He explained the title by noting the distinct resemblance of P1 coliphage to the Discovery One spacecraft dreamed up by Arthur C Clarke and Stanley Kubrick – and then went on to exuberantly and idiosyncratically recount a brief history of bacteriophages and their use in biotechnology since their discovery.  A revelation from his talk was that the first discovery of phages was probably described by a gentleman named Hankin, in 1896 in Annales de l’Institut Pasteur: he

The 1896 paper from Annales de l'Institut Pasteur

showed that river water downstream of cholera-infested towns on the Jumma river in India contained no viable Cholera vibrio – and that this was a reliable property of the water.  We were also introduced to the concept of turtles as undertakers in the Ganges….

He took us through the achievements of the Phage Group of Max Delbruck and others – where science was apparently fun, but also resulted in the establishment of modern molecular biology – through to the use of phages as exquisitely sensitive indicators immunochemistry studies in the 1960s.

All too soon we got to the modern uses of phages, with 3 types of gene library – random peptide, fragmented gene, and antibody V regions – being used to make recombinant phage tail proteins to be used for “panning” and enrichment purposes, in order to select either specific antibodies or antigens.  Dion manages a research programme at OVI aimed at developing a new generation of veterinary vaccines – and has for some years now been making significant progress in generating reagents from a chicken IgY single-chain Fv phage display library.

Carolyn Williamson (IIDMM, UCT) gave us an update on CTL epitopes associated with control of HIV-1 subtype C infections.  She said that it was now known that genome-wide association studies (GWAS) gives you certain HLAs which are associated with low viral load, and others with high – meaning that to some extent at least, control of infection was down to genetic luck.  She noted that they and others had shown that CTL escape was quick: this generally happened in less than 5 weeks in acute phase infections.

They had looked for evidence of a fitness cost of CTL escape – and shown that it exists.  She noted that this meant that even if one has “bad” HLA genes, if one was infected with a virus with fitness cost mutations from another, that one could still control infection.

It had been shown that “controllers” mainly have viruses with attenuating mutations, or have escapes in the p24 region – and it was a possible vaccine strategy to include these mutated epitopes in vaccines to help people with infections control their infections.

An interesting topic she broached was that of dual infections – there was the possibility of modelling if infection with two different viruses results in increased Ab neutralisation breadth, and if one would get different results if infections were staggered, possibly with increased nAb evolution if isolates were divergent.  She noted it was possible to track recombination events with dual virus infections too.

It was interesting that, as far as Ab responses went, there were independent responses to 2 variants and one could get a boost in Ab titres to the superinfecting virus, but not a boost to Abs reacting with the originally-infecting virus

Carolyn was of the opinion that HIV vaccines needed to include CTL epitopes where escape is associated with fitness cost.  She also reiterated that superinfection indicated that one can boost novel responses, which I take to mean that therapeutic applications are possible.

Ulrich Desselberger (University of Cambridge) is a long-time expert on rotaviruses and the vaccines against them, and it was a pleasure to finally hear him speak – and that he was mentoring young people in South Africa.  He said that more than a third of children admitted to hospital worldwide were because of rotavirus infections, meaning that the viruses were still a major cause of death and morbidity – and they were ubiquitous.

He reviewed the molecular biology and replication cycle of rotaviruses in order to illustrate where they could be targeted for prevention of infection or therapy, and noted that drugs that interfere with lipid droplet homeostasis interfere with rotavirus replication because 2 viral proteins associated proteins of lipid droplets.

He stated that there were lots of recent whole-genome sequences – we already there were many types, based on the 2 virion surface proteins; we  now know that other genes are also highly variable.  As far as correlates of immunity were concerned, VP7 & 4 were responsible for eliciting neutralising Ab.  Additionally, protective efficacy of VP6 due to elicitation of non-neutralising Ab had been shown in mice – but not in piglets, and not convincingly in humans.  Abs to VP2, and NSP2 and 4 were also partially protective in humans.  It was interesting that protection was not always correlated with high titre nAb responses.

He noted that in clinical disease primary infections partially protected against subsequent infections which are normally milder; subsequently no disease was seen even when infection occurred.  Cross-protection occurred at least partially after initial infection, and this got better after more exposure.  There was evidence one could get intracellular neutralisation by transcytosed Ab, and especially to VP6.  Ab in the gut lumen was a good indication of protection.

As far as the live modern vaccines were concerned, Merck’s Rotateq elicited type-specific nAb, with 9% of recipients shedding live virus.  GSK’s Rotarix gets elicits cross-reactive nAb and one gets 50% of recipients shedding virus.

While the vaccines seemed safe, he noted that where vaccines had been introduced, efficacy ranged from 90% in the USA and Europe, down to as low as 48% in Bangladesh, Malawi and SA, due to type mismatch, and that efficacy was correlated inversely with disease incidence and child mortality generally.  He mentioned that there had been much VLP work, but that none of the candidates was near licensure.

Johan Burger (Stellenbosch University) spoke on one of the more important non-human virus problems in our immediate environment – specifically, those affecting wine grape production in our local area.  He opened by stating that SA now produced 3.7% of the world’s wine, making grapes a nationally and especially locally important crop.  Leafroll disease was a major worldwide problem – as well as being the reason for the wonderful autumn reddening seen in grapevines, it also significantly limited production in affected vineyards.  His laboratory has done a lot of work in both characterising viruses in grapevine, and trying to engineer resistance to them.  Lately they were also investigating the use of engineered miRNAs as a response to and means of controlling, virus infection.

His group has for a couple of years been involved in “metaviromic” or high-throughput sequencing studies of grapevines, with some significant success in revealing unsuspected infections.  In this connection, he and Don Cowan pointed out that they had lots of data that they ignore – but which we should keep and study, as a resource for other studies not yet thought of.

As far as Johan’s work went, novel viruses kept popping up, including grapevine virus E (GVE), which hitherto had only been found in Japan.  They were presently looking at Shiraz disease, which was unique to SA, and was still not understood.  This was infectious, typified by a lack of lignification which led to rubbery vines, and kills plants in 5 years.  It also limits the production of the eponymous grapes – a crime when SA shirazes seem to be doing so well!

Veterinary Virology and Vaccines parallel session.

I again dodged the clinical / HIV session because of my personal biases, and was again treated to a smorgasbord of delight: everyone spoke well, and to time, and I was really gratified to see so many keen, smart young folk coming through in South African virology.  It was also very interesting to see highly topical subjects like Rift Valley fever and rare bunyavirus outbreaks being thoroughly covered, so I will concentrate on these.

P Jansen van Veeren (NICD, Johaanesburg) was again a speaker, this time representing his absent boss, Janusz Paweska.  He gave an account of the 2010 Rift Valley fever outbreak in SA, and epidemiological findings in humans – something of keen interest to me.  He said there had been some forecasting success for outbreaks in East Africa; however, there were long gaps between outbreaks, which were generally linked to abnormal rainfall and movement of mosquito and animal hosts.  RVFV isolates differed in pathogenicity but were structurally and serologically indistinguishable – because virulence was due to the NSs protein, and not a virion component.  He recounted how artificial flooding of a dambo in Kenya resulted in a population boom in the floodwater Aedes mosquitoes responsible for inititating an outbreak, and then of the Culex which maintained the epidemic.  He said there was a strong correlation between viral load and disease severity.

In terms of South African epidemiology, there had been smaller outbreaks from 2008 round the Kruger National Park (NE SA), then in the Northern Cape and KZN in 2009.  People had been infected from autopsy of animals, and handling butchered animal parts.  The 2010 outbreak started in the central Free State after an unusually wet period, and had then spread to all provinces except Limpopo and KZN.  In-house serological methods at the NICD were validated in-house too: these were HAI screening and IgM and IgG ELISAs and a virus neutralisation test.  They had got 1600+ samples of human serum, and confirmed 242 cases of disease and 26 deaths for 2010.

He noted that with winter rains there was a continuous outbreak in the Western Cape, and in 2011 the epidemic had started again in the Eastern and Western Cape Provinces, but has since tailed off.  Some 82% of human cases were people who occupationally handled dead animals, although there was some possibility of transmission by mosquitoes.

In human cases there was viraemia from 2-7 days, with IgM present transiently from 3 days at low level.  They had sequenced partial GP2 after PCR from 47 isolates, and showed some recombination occurring.  The 2010 isolates were very closely related to each other, and to a 2004 Namibian isolate.  There had been no isolation from mosquitoes yet.

Two talks on FMDV followed: Belinda Blignaut (OVI and Univ Pretoria) spoke on indirect assessment of vaccine matching by serology, and Rahana Dwarka (OVI) on a FMDV outbreak in KZN Province in 2011.  Belinda’s report detailed how 6 of 7 serotypes of FMDV occur in SA, with SAT-1 and -2 and O the most common – and that vaccines needed to be matched to emerging strains.  This was done by indirect vaccine matching tests such as serological r-value, determined by the ratio of the reciprocal serum titre to the heterologous virus against that to the homologous virus.  They had put 4 different viruses into cattle and got sera to test a range of 26 newly isolated viruses.  While they had not got sequence from the test panel viruses, indications were that topotype 3 viruses are antigenically more disparate and that a vaccine consisting of topotype 1 or 2 antigens may not be effective in the control of FMD.

In introducing Rahana’s talk, the chair (Livio Heath, OVI) mentioned that there had been 5 different major animal pathogens causing outbreaks in SA over the last 3 years – and that they had to produce reagents and validate tests for ASFV, classical swine fever (CSF) and FMDV, etc, with each outbreak.  Rahana described how they had neutralisation assays and blocking and competition ELISA for FMDV, as well as a big database of isolates from buffalo in KZN – so they were well-placed to type viruses found in cattle in the region.

C van Eeden (Univ Pretoria) had an intriguing account of their investigation of the occurrence of an orthobunyavirus causing neurological symptoms in horses and wildlife.  Horses seem to be particularly vulnerable to many of the viruses involved in such disease, and so are a useful sentinel species.  Shuni virus was first isolated from Culicoides midges and sheep and a child in Nigeria in the 1960s.  SA workers subsequently found it in some livestock and Culex mosquitoes and in horses.  The virus was shown to be a neurologic disease agent in horses and wildlife – then disappeared for some 30 years, much like Ebola.  There is apparently a new research unit at UP with a BSL3 lab, so they are well equipped to do tests with the virus.

Ms van Eeden noted that the incidence of encephalitic disease in humans and animal in SA is underreported, and the causes are mainly unknown – a revelation to me!  Horses are susceptible to many of the agents, and are useful sentinels – workers have identified flavi- and alphaviruses in some outbreaks, but many are not IDed.  They had done cell culture and EM on samples from an ataxic horse: they got a bunyavirus-like virus by EM, and did bunya-specific PCR, and got Shuni virus back.  Sequence relationships showed no linkage to type of animal or date, in subsequent samplings from horses, crocodiles,  a rhino and a warthog, and from blood, brain and spinal cord.  All positive wildlife were sampled in Limpopo Province; horses only from most other provinces.

She noted that latest cases were neurological, whereas previously these were mainly febrile.  The virus accounted for 10% all neurological cases, with a 50% fatality rate.  She noted further that vets often work without masks or gloves, and so had no protection from exposure in such cases….  There was no idea on what the vector was, but they would like to test mosquitoes, etc.  Ulrich Desselberger suggested  rodents may be a reservoir, but they don’t know if this is true.

Stephanie van Niekerk (Univ Pretoria) investigated alphaviruses as neurological disease agents in African wildlife.  The most common alphaviruses in SA are Sindbis and Middelburg viruses.  Old World alphaviruses are usually not too bad, and cause arthritic and febrile symptoms, while New World cause severe neurological diseases.  Sindbis was been found in SA outbreaks in 1974.  However, Stephanie noted that a severe neurological type had appeared since 2008 in horses.  Accordingly, they looked at unexplained cases in wildlife in the period 2009-2011: brain and spinal cord samples were investigated for all cases.  They found alphavirus in a number of rhinos, buffalo, warthog, crocodiles and jackal – and all except for one rhino were Middelburg virus.  They want to isolate viruses in cell culture, and increase the size of regions used for cDNA PCR.  Stephanie said the opinion was that the values of the animal involved justifies the development of vaccines.

 

Virology Africa 2011: viruses at the V&A Waterfront 1

12 December, 2011

We thank Russell Kightley for permission to use the images

Anna-Lise Williamson and I again hosted the Virology Africa Conference (only the second since 2005!), at the University of Cape Town‘s Graduate School of Business in the Victoria & Alfred Waterfront in Cape Town.  While this was a local meeting, with just 147 attendees, we had a very international flavour in the plenaries: of 18 invited talks, 9 were by foreign guests.  Plenaries spanned the full spectrum of virology, ranging from discovery virology to human papillomaviruses to HIV vaccines to tick-borne viruses to bacteriophages found in soil to phages used as display vectors, and to viromes of whole vineyards.  There were a further 52 contributed talks and 41 posters, covering topics from human and animal clinical studies, to engineering plants for resistance to viruses.

A special 1-day workshop on “Human Papillomaviruses – Vaccines and Cervical Cancer Screening” preceded the main event: this was sponsored by Merck Sharp & Dohme, Roche and Aspen Pharmacare, and had around 90 attendees.  Anna-Lise Williamson (NHLS & IIDMM, UCT) opened the workshop with a talk entitled “INTRODUCTION TO HPV IN SOUTH AFRICA – SCREENING FOR CERVICAL CANCER AND VACCINES”, and set  the stage for Jennifer Moodley (Community Health Dept, UCT) to cover health system issues around the prevention of cervical cancer in SA, and the newly-minted Dr Zizipho Mbulawa (Medical Virology, UCT) to speak on the the impact of HIV infection on the natural history of HPV.  This last issue is especially interesting, given that HIV-infected women may have multiple (>10) HPV types and progress faster to cervical malignancies, and HPV infection is a risk factor for acquisition of HIV.  The Roche-sponsored guest, Peter JF Snijders (VU University Medical Center, Amsterdam), gave an excellent description of novel cervical screening options using primary HPV testing, to be followed by two accounts of cytological screening in public and private healthcare systems in SA, by Irene le Roux (National Health Laboratory Service) and Judy Whittaker (Pathcare), respectively.  Ulf Gyllensten (University of Uppsala, Sweden) described the Swedish experience with self-sampling and repeat screening for the prevention of cervical cancer, especially in groups that are not reached by standard screening modalities.  Hennie Botha and Haynes van der Merwe (both University of Stellenbosch) closed out the session with talks on the effect of the HIV pandemic on cervical cancer screening, and a project aimed at piloting adolescent female vaccination against HPV infection in Cape Town.

The next part of the Workshop overlapped with the Conference opening, with a Keynote address by Margaret Stanley (Cambridge University) on how HPV evades host defences (sponsored by MSD), and another by Hugues Bogaert (HB Consult, Gent, Belgium)) on comparisons of the cross-protection by the two HPV vaccines currently registered worldwide (sponsored by Aspen Pharmacare). Margaret Stanley’s talk was a masterclass on HPV immunology: the concept that such a seemingly simple virus (only 8 kb of dsDNA) could interact with cells in such a complex way, was a surprise for all not acquainted with the viruses.  Bogaert’s talk was interesting in view of the fact that the GSK offering, which has only only two HPV types, raises far higher titre antibody responses than the MSD vaccine with four HPV types, AND seems to elicit better cross-protective antibodies: this should help inform choice of product from the individual point of view.  However, the fact that MSD seems able to respond better to national healthcare system tenders in terms of price per dose is also a major factor in the adoption stakes.

The Conference proper started with a final address by Barry Schoub, long-time but now retired Director of the National Institute of Virology / National Institute of Communicable Diseases in Johannesburg, and also long-time CEO of the Poliomyelitis Research Foundation (PRF): this is possibly the premier funding agency for anything to do with viruses in South Africa, and a major sponsor of the Conference.  He spoke on the history of the PRF, and how it had managed to shepherd an initial endowment of around 1 million pounds in the 1950s, to over ZAR100 million today – AND to dispense many millions in research project and bursary funding in South Africa over several decades.

The first session segued into a welcoming cocktail reception and registration at the Two Oceans Aquarium in the V&A Waterfront: this HAS to be one of the only social events for an academic conference where the biggest sharks are the ones in the tank, and not in the guest list!  I think people were suitably blown away – as always, in the aquarium – and the tone was set for the rest of the meeting.  The wine and food were good, too.

The first morning session of the conference featured virus hunting and HIV vaccines, as well as plant-made vaccines and more HPV.  W Ian Lipkin (Columbia University, USA) opened with “Microbe Hunting” – which lived up to its title very adequately, with discussion of a plethora of infectious agents.  As well as of the methods newly used to discover them, which include high-throughput sequencing, protein arrays, very smart new variants on PCR….  I could see people drooling in the audience; the shop window was tempting enough to make one jump ship to work with him without a second thought.  He said that probably 99% of vertebrate viruses remain to be discovered, and that advances in DNA sequencing technology were a major determinant in the rapidly-increasing pace of discovery.  He made the point that while the emphasis in the lab had shifted from wet lab people to bioinformatics, he thought it would move back again as techniques get easier and more automated – meaning (to me) that there is no substitute for people who understand the actual biological problems.  It was interesting that, while telling us of his work on the recently-released blockbuster “Contagion” – where “the virus is the star!” – he showed a slide with a computer in the background running a recombination detection package called RDP, which was designed in South Africa.  It can also be seen in the trailer, apparently.  Darren Martin will not be looking for royalties or screen credits, however.

Don Cowan (University of the Western Cape) continued the discovery theme, albeit with bacteriophages as the target rather than vertebrate viruses.  It is worth emphasising that phages probably represent the biggest source of genetic diversity on this planet – and given how even the most extreme of microbes have several kinds of viruses, as Don pointed out, it is possible that this extends to neighbouring planets too [my speculation – Ed].  He occupies an interesting niche – much like the microbes he hunts – in that he specialises in both hot and cold terrestrial desert environments, which are drastically understudied in comparison to marine habitats.  He made the interesting point that metagenome sequencing studies such as his own generate data that is in danger of being discarded without reuse, given that folk tend to take what they are interested out of it and neglect the rest.

Anna-Lise Williamson (NHLS, IIDMM, UCT) then described the now-defunct SA AIDS Vaccine Initiative vaccine development project at UCT.  It is rather sobering to revisit a project that used to employ some 45 people, and had everything from Salmonella, BCG, MVA, DNA and insect cell and plant-made subunit HIV vaccines in the pipeline – and now employs just 5, to service the two vaccines that made it into into clinical trial.  The BCG-based vaccines continued to be funded by the NIH, however, and the SA National Research Foundation funds novel vaccine approaches.  Despite all the funding woes, the first clinical trial is complete with moderate immunogenicity and no significant side effects, and two more are planned: these are an extension of the first – HVTN073/SAAVI102 – with a Novartis-made subtype C gp140 subunit boost, and the other is HVTN086/SAAVI103, which compprises different commbinations of DNA, MVA and gp140 vaccines.

It was clear from the talk that if South Africa wants to support local vaccine development, the government needs to support appropriate management structures to enable this – and above all, to provide funding.  However, all is not lost, as much of the remaining expertise in several of the laboratories that were involved in the HIV vaccine programme can now involve themselves in animal vaccine projects.

Plant-made HPV16 VLPs

Ed Rybicki made it an organisational one-two with an after-tea plenary on why production of viral vaccines in plants is a viable rapid-response option for emerging or re-emerging diseases or bioterror threats.  The talk briefly covered the more than 20 year history of plant-made vaccines, highlighting important technological advances and proofs of concept and efficacy, and concentrated on the use of transient expression for the rapid, high-level expression of subunit vaccines.  Important breakthoughs that were highlighted included the development of the Icon Genetics TMV-based vectors, Medicago Inc and Fraunhofer USA’s recent successes with H5N1 and H1N1 HA protein production in plants – and the Rybicki group’s successes with expression of HPV L1-based and E7 vaccine candidates.  The talk emphasised how the technology was inherently more easily scalable, and quicker to respond to demand, than conventional approaches to vaccine manufacture – and how it could profitably be applied to “orphan vaccines” such as for Lassa fever.

Ulf Gyllensten had another innings in the main conference, with a report on a study of a possible linkage of gene to disease in HPV infections – which could explain why some people clear infections, and why some have persistent infections.  They used the Swedish cancer registry (a comprehensive record since the end of the 1950s) to calculate familial relative risk of cancer of the cervix (CC): relative risk was  2x for a full sister, the same for a mother-daughter pair and the risk for a half sister was 50% higher while risk was not linked to non-biological siblings or parents, meaning the link was not environmental.  A preliminary study found HLA alleles associated with CC, and increased carriage of genes was linked to increased  viral load.   A subsequent genome wide association study using an Omni Express Bead Chip detecting700K+ SNPs yielded one area of major interest, on Chr 6 – this is a HLA locus.  They got 3 independent signals in the HLA region and can now potentially link HPV type and host genotype for a prediction of disease outcome.  Again, the kinds of technology available could only be wished for here; so too the registry and survey options.

Molecular and General Virology contributed talks parallel session

I attended this because of my continued fascination with veterinary and plant viruses – and because Anna-Lise was covering the Clinical and Molecular session – and was not disappointed: talks were of a very high standard, and the postgraduate students especially all gave very good accounts of themselves.

Melanie Platz (Univ Koblenz-Landau, Germany) kicked off with a description of a fascinating interface between mathematics and virology for early warning, spatial awareness and other applications.  She gave an example using a visual representation of risk using GIS for Chikungunya virus, based on South African humidity and temperature data going back nearly 100 years: this had a 3D plot model, into which one could plug data to get predictions of mosquito likelihood.  They could generate risk maps from the data, to both inform public and policy / planning.  They had a GUI for mobile devices for public information, including estimates of risk and what to do about it, including routes of escape.

Cover Illustration: J Virol, October 2011, volume 85, issue 20

This was followed by one of my co-supervised PhD students, Aderito Monjane – who recently got the cover of Journal of Virology with his paper on modelling maize streak virus (MSV) movement and evolution, so I will not detail more here.  However, even as a co-supervisor I was blown away by the fact that he was able to show animations of MSV spread – at  30 km/yr, across the whole of sub-Saharan Africa.

Christine Rey of Wits University provided another state-of-the-art geminivirus talk, with an account of the use of siRNAs and derivatives for silencing cassava-infecting geminiviruses.  They were using genomic miRNA precursors as templates to make artificial miRNAs containing viral sequences, meaning they got no interference with nuclear processing and there was less chance of recombination with other viruses, a high target specificity, and the transgenes would not be direct targets of virus-coded suppressors.  They could also use multiple miRNAs to avoid mutational escape.  The concept was successful in tobacco, and they had got transformation going well for cassava, so hopes were high for success there.

Dionne Shepherd (UCT) spoke on our laboratory’s 15+ year work on engineered resistance in maize to MSV.  She pointed out that the virus threatens the livelihood of 200 million+ subsistence farmers in Africa, and is thought to be the biggest disease concern in maize – which is still the biggest edible crop in Africa.  Most of the work has been described elsewhere with another journal cover; however, new siRNA-based constructs still under investigation were even more effective than the previous dominant negative mutant-based protection: the latter gave 50-fold reduction in virus replication, but silencing allowed > 200-fold suppression of replication.

2-colour surface rendition of HcRNAV

Arvind Varsani – a former UCT vaccinology PhD who is now a structural biology and virology lecturer at Univ Christchurch (NZ) – described what is probably the first 3D structure of a virus to come out of Africa.  This was of a 30 nm isometric ssRNA virus – Heterocapsa circularisquama RNA virus (HcRNAV) – infecting a dinoflagellate, which is one of the most noxious red tide bloom agents and is a major factor in killing farmed oysters.  The virus apparently controls the diatom populations.  There are two distinct strains of virus, and specificity of infection is due to the entry process, as biolistic bombardment obviates the block.  The single capsid protein probably has the classic jelly-roll β-barrel fold, but they observe a new packing arrangement that is only distantly related to the other ssRNA (+) virus capsids known.  They will go on to look at structural differences between strains that change cell entry properties.

FF Maree from the Onderstepoort Veterinary Institute and the Univ Pretoria spoke on structural design of FMDV to improve vaccine strains: they wished to engineer viruses by inserting the cell culture adapted HSPG-binding signature sequence and to mutate capsid residues to increase the heat stability of SAT-2 subtype virus vaccines.  If they put the signature sequence in a SAT1 virus, they found it could infect CHO cells – which do not express any of 4 integrins that FMDV binds to, but are far better for large-scale production of the virus than the BHK cells used till now.  It was also possible to increase hydrophobic interactions in the capsid by modeling: eg a VP2 Ser to Tyr replacement gave a considerably better thermal inactivation profile to the virus.

Daria Rutkowska (Univ Pretoria) detailed how African horsesickness orbivirus (AHSV) VP7 protein had significant potential as a scaffold that could act as a vaccine carrier.  The native protein formed as trimers assembled in a VP3+VP7 “core” particle; however, the VP7 when expressed alone could form soluble trimmers – and the “top” domain hydrophilic loop can tolerate large inserts.  The group had very promising FMDV P1 peptide responses from engineered VP7 constructs, including protection of experimental animals.

P Jansen van Veeren of the National Institute of Communicable Disease in Johannesburg finished off the session, with a description of the cellular pathology caused by Rift Valley fever bunyavirus (RVFV) in mice in acute infections.  The virus seems to have been of particular international interest recently as a potential bioterror agent; however, global warming is also responsible for its mosquito vector spreading outside of its natural base in Africa to the Arabian peninsula, and there are fears of the virus getting into Europe soon.  While there are vaccines against the virus, including a live attenuated version, none are licenced for human use.  It was interesting to hear that the viral NP appears to be the main immunogen, as there are massive amounts of NP produced in infection, and huge responses to it in infected animals – and NP immunisation protects mice.  There is a good Ab response but it is not neutralising, while NP is released independently of other proteins from infected cells.  The liver is the major target of virus infection, with a bias to apoptosis of hepatocytes and severe inflammatory responses.  Viral load is linked to these effects and is much lower in vaccinees.  Immunisation reduces liver replication markedly; that in the spleen less so.  A screen of cytokines and other gene responses showed a big down-regulation of many genes in non-vaccinated mice to do with cytokines, and down-regulation of B and T cells and NK cells.  He thinks recombinant vaccine candidates should have both the surface glycoproteins and the NP in order to be effective – and that there is a major need for proper reagents for big animal studies.

Viruses as nanomachines! Or: what you can believe from YouTube

6 December, 2010

I have for some years now been teaching my undergrad students that virus particles are nanomachines: that is, they are highly sophisticated nanoscale (read: ultramicroscopic) devices whose function is to specifically deliver genetic material into an environment where it can be expressed and replicated, so as to make more virus particles.

Nanoscale von Neumann machines, then – and if you want to see what a macroscale vN machine could do, just watch “2010: Odyssey Two“.

Ah, but what’s a von Neumann machine, you ask?  Well, I note Wikipedia has the following:

  • Self-replicating machines, a class of machines that can replicate themselves
  • Universal Constructors, self-replicating cellular automata
  • Von Neumann probes, hypothetical space probes capable of self-replication
  • Nanorobots capable of self-replication

I especially like the last two – because, as I showed in a previous blog post, I like the idea of virus particles or virions as “inner space craft”.  That this neatly marries my recreational and professional reading is no coincidence – because they cross-pollinate one another, in that I get ideas about the nature of viruses from SF, and my virology training informs scenarios I would like to write about.  Someday.  Soon, possibly.  Really.  Instead of writing about parallel universes contactable via the internet….

However, there is more to viruses and nanotechnology than phages with contractile tails, whether or not they have been around for billions of years: mimiviruses too have both nanoscale DNA loading and rapid-delivery systems, as previously discussed here.

Although I have a passing fondness for possibly my most successful animation – made from actual EMs, done by Linda Stannard.

T4 phage infecting a cell

So it was with some pleasure I saw recently on YouTube a video labelled “Viruses are nanotechnology (how a virus works)“.  I was a little less pleased when a voice confidently announced that “…a virus isn’t alive, people – it’s non-metabolising…”, as if that was the sole and necessary criterion for life.  I am at one with another Polish-named person – one Bernard Korzeniewski – in thinking  that life is (from MicrobiologyBytes)

The phenomenon associated with the replication of self-coding informational systems” © E.P. Rybicki, 1996. Incidentally, I find another person with a Polish name has said something very similar, in 2001 – which means it must be true. Bernard Korzeniewski describes life as: “A network of inferior negative feedbacks subordinated to a superior positive feedback.”

See, no mention of metabolism – or even of cells!  But what got the hairs on the back of my neck standing up, however, was some of the rest of it – delivered in a smooth, folksy manner, with stunning video footage.  Absolute cr@p, most of it: viruses are too complicated to have evolved, so they have to be alien nanotech???

Obviously some weird kind of conspiracy theory cross technobabble – but very seductive, to the uninformed.  Some of the comments are also just out of this world – literally!

Fortunately, there are some real science videos out there too – some of which I have also used in lecturing, if only to illustrate just how cool structural biology can be when used to study viruses.  Prime among these is one of T4 virus (Enterobacteria phage T4) infecting E coli; another magical one  from the same source is a depiction of the molecular motor which winds DNA into T4 heads.  A longer video has Michael Rossman, whose lab did the structural work behind the videos, explaining how the phenomenon could be useful in understanding viruses like herpesviruses in humans, which also appear to have molecular motors for DNA delivery – and, of course, how we can mess with them.

Self-assembly of viruses is also a good topic for video – and the full-length  Seyet T4 video is stunning in this regard.  So too is this one, showing a PhiX174 microvirus particle assembling.  One of my favourites, though, is the simplest: this is the depiction of how simple shapes can be induced to self-assemble into a virus-like particle – just by shaking.

I suppose, like everything, you get what you pay for with YouTube: which is nothing, most of the time.

But every now and then, a gem – which is what makes it fun to look.  I’m off to hunt down a Rolling Stones video virus replication videos!