Chapter-8---Ancient-DNA-in-the_2019_Ortner-s-Identification-of-Pathological-

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 28

Chapter 8

Ancient DNA in the Study of Ancient


Disease
Anne C. Stone1,2,3 and Andrew T. Ozga2,3
1
School of Human Evolution and Social Change, Tempe, AZ, United States, 2Center for Evolution and Medicine, Tempe, AZ, United States, 3Institute
for Human Origins, Tempe, AZ, United States

INTRODUCTION TO ANCIENT DNA sequencing) was laborious and results often were con-
tested due to difficulty in identifying contamination from
As discussed in this volume, human skeletal remains can modern DNA. In the last 10 years, the development of
contain a multitude of clues about the impact of patho- new extraction and sample preparation methods for
gens and chronic diseases on the overall health of past ancient DNA analysis as well as the application of next-
peoples. However, many pathogens are known to cause generation sequencing (NGS) has facilitated ancient geno-
death quickly, resulting in limited, if any, skeletal mani- mics, including the investigation of ancient pathogen
festations, causing the remains to appear “healthy.” In genomics.
addition, death often may be the result of multiple factors Regardless of methodology, the examination of past
that can include early life insults that contribute to frailty diseases through ancient DNA requires awareness of the
(e.g., DeWitte, 2015, see also Chapter 2) or coinfections. evolutionary context of the pathogen and its host. To
For example, tuberculosis (TB) is the leading cause of accomplish this, ancient DNA researchers need to be con-
death today for individuals with human immunodefi- versant in a number of related fields: population genetics,
ciency virus (HIV) (TB/HIV facts 2015, http://www.who. evolutionary biology, computational biology, human
int/hiv/topics/tb/tbhiv_facts_2015/en/). Such complexities genetics, and pathogen genetics. In order to establish an
can be addressed through the use of ancient DNA analysis appropriate evolutionary context, relevant modern genetic
methods. The study of DNA from archeological remains, data must be included (including from closely related
often referred to as molecular archeology or, more taxa) for comparison with sequences recovered from
recently, paleogenomics, uses techniques from molecular ancient samples. Thus, the initial design of the research
and evolutionary biology to address an array of questions project is crucial so that the data obtained are informative
about the population history and evolution of past peo- from an anthropological or evolutionary medicine per-
ples, domesticates, and pathogens. spective and so that the results are statistically robust.
Although ancient DNA research began in the 1980s Ancient DNA, when successfully obtained, can
(Hagelberg et al., 1989; Higuchi et al., 1984; Pääbo address many questions about the host and the microbial
1985), the focus on ancient disease did not begin until the (including pathogen) environments in which they lived.
early 1990s. This initial research focused primarily on One basic goal in this research is confirming whether a
Mycobacterium tuberculosis and Mycobacterium leprae specific pathogen is present, which is of particular interest
(Arriaza et al., 1995; Rafi et al., 1994; Salo et al., 1994; to the paleopathologist who wants to know whether a par-
Spigelman and Lemma, 1993). In addition to infectious ticular skeletal lesion might be the result of a specific
disease pathogens, some of the early work also examined pathogen. Such analyses could also help indicate the
immune loci, specifically HLA (Ivinson et al., 1992; range of phenotypes caused by the pathogen in question.
Lawlor et al., 1991), and loci related to hemoglobinopa- Importantly, absence of evidence must not be interpreted
thies (Béraud-Colomb et al., 1995; Filon et al., 1995). as evidence of absence, since pathogen DNA may not be
The early era of ancient DNA (i.e., pre-next-generation

Ortner’s Identification of Pathological Conditions in Human Skeletal Remains. DOI: https://doi.org/10.1016/B978-0-12-809738-0.00008-9


© 2019 Elsevier Inc. All rights reserved. 183
184 Ortner’s Identification of Pathological Conditions in Human Skeletal Remains

preserved. In addition, the presence of a particular patho- sequence (Handt et al., 1994; Höss et al., 1996; Pääbo,
gen does not necessarily mean that it was the cause of 1989). In addition, the widespread use of PCR for the
death. From an evolutionary perspective, simply identify- amplification of low template and/or damaged material
ing the presence of a pathogen is not as interesting as (i.e., ancient DNA) revealed the challenges of contamina-
placing the agent of disease into larger epidemiological or tion either during sample excavation, preparation, extrac-
evolutionary contexts. For example (as discussed later in tion, or during the PCR preparation itself, resulting in
this chapter), recent analyses of both Justinian plague and assessments of where during these processes contamina-
Black Death victims have shown that the bacterium tion was likely and recommendations of how to prevent it
Yersinia pestis was indeed the cause of both pandemics (Champlot et al., 2010; Cooper and Poinar, 2000;
(Bos et al., 2011; Schuenemann et al., 2011; Wagner Deguilloux et al., 2011; Leonard et al., 2007; Malmstrom
et al., 2014). This research has laid to rest many debates et al., 2005; Richards et al., 1995).
about the cause of both plague events; however, it has Many of the first-generation ancient DNA studies
also illustrated the power of evolutionary analyses to shed focused on mitochondrial DNA (mtDNA) (Hagelberg and
light on the course of pandemics over time, including Clegg, 1991; Higuchi et al., 1984; Pääbo et al., 1988;
changes in the pathogen itself. In addition, current and Stone and Stoneking, 1993). mtDNA is found in the
future work on the human host, specifically people living mitochondria, the energy-producing organelles typically
before and after these pandemics, may reveal adaptations present in hundreds or even thousands of copies within
that increased resistance to Y. pestis in the affected popu- most cells. Thus, mtDNA is easier to obtain compared
lations over time. Finally, ancient DNA analyses can offer with nuclear DNA, considering it has a much higher copy
information about some of the changes in the human number per eukaryotic cell (estimated at 1100 8800
microbiome over time, including how it has been copies/cell (Bogenhagen and Clayton, 1974)). A few
impacted by changing subsistence patterns and the man- studies targeted pathogen DNA though comparative
ner by which it contributes to health. In this chapter, we pathogen data were often scarce since many pathogens of
will discuss the current methods used to study ancient interest remained uncharacterized either without a com-
DNA along with some of the successes and challenges of plete genome sequence or in terms of their diversity (or
this work. both). As a result, the early ancient pathogen studies
generally focused on identifying the presence of a single
pathogen in a sample (Rafi et al., 1994; Salo et al., 1994;
History/Trajectory of the Field Spigelman and Lemma, 1993) rather than evolutionary
Ancient DNA analyses have been driven by technological analyses. Exceptions to this rule include studies of such
advances in molecular biology as well as theoretical viruses as the influenza virus and HIV, which possess gen-
advances in population genetics and phylogenetics. omes small enough to sequence completely (Taubenberger
During the 1980s, these advances were fostered by the et al., 2005; Worobey et al., 2008). Another major challenge to
invention of the polymerase chain reaction (PCR) (Mullis PCR testing for the presence of a single pathogen is that these
et al., 1986; Saiki et al., 1988). This method became the analyses were, in general, phylogenetically uninformative. In
workhorse of ancient DNA research, as well as much of other words, while many of these studies may have in fact
molecular biology in general, because this technique can found the pathogen in question, it was often not possible to
copy a specific fragment of DNA (or RNA if reverse tran- distinguish the result from contamination or a closely related
scriptase PCR (RT-PCR) is used), essentially doubling the nonpathogenic microbe found in the burial environment. For
number of fragments for every “cycle” of amplification. example, early research on ancient tuberculosis used the
This can be successful even if there are only a few of the IS6110 element or short gene sequences to identify M. tuber-
relevant copies of the desired sequence in the initial culosis complex (MTBC) DNA (e.g., Arriaza et al., 1995; Salo
extraction elution. PCR was followed by one of three pri- et al., 1994; Spigelman and Lemma, 1993). Such sequences do
mary techniques: DNA fragment analysis using restriction not provide sufficient information to distinguish strains, and
enzymes to analyze polymorphic sites, direct Sanger thus, target DNA contaminated by positive controls used in the
sequencing (which provides an “average” or consensus laboratory cannot be clearly excluded.
sequence of the DNA fragment), or cloning, whereby the As a molecular technique, PCR is still common in
PCR products were inserted into a bacterial vector (to genetics laboratories due to its low cost and ease of use.
separate and copy individual molecules of the pool of It is often a quick way to test samples for contamination
DNA products generated by PCR) followed by direct or for the presence of host or pathogen DNA. In particular,
sequencing of some of the clones. The successful amplifi- a method known as quantitative PCR (qPCR), in which
cation of PCR products from ancient DNA is often influ- fluorescent probes adhere to specific DNA sequences,
enced by coextracted inhibitors, the amount of starting can be used to quantify directly the number of DNA mole-
template, DNA damage, and the size of the targeted cules amplified during each cycle of the PCR process
Ancient DNA in the Study of Ancient Disease Chapter | 8 185

(Heid et al., 1996). The resulting amplification curve is an The first ancient DNA studies were aimed primarily at
illustration of the number of DNA molecules present in successfully extracting and then sequencing DNA using
each sample over the course of each PCR cycle. qPCR can traditional Sanger sequencing, most commonly of frag-
be used for a number of purposes: to quantify the number ments of mtDNA (e.g., Hagelberg and Clegg, 1991;
of starting molecules (important for library preparation as Higuchi et al., 1984; Pääbo, 1989). Newer studies that use
discussed in the next section), to detect inhibition due to NGS have generated complete mitochondrial and nuclear
coextracting inhibitors, and to screen samples to assess genome sequences from human samples and full micro-
whether they would be amenable to the newer, more time- bial genomes as part of microbiome analyses (e.g., Green
consuming and expensive NGS methods (Bunce et al., et al., 2010; Poinar et al., 2006; Rasmussen et al., 2010;
2012; Enk et al., 2013; Harkins et al., 2015; King et al., Warinner et al., 2014b). While the methods for ancient
2009; Poinar et al., 2006). However, when testing for the DNA analysis have developed rapidly, due partly to stud-
presence of a pathogen, care must be taken in the design of ies of the ancient Homo lineage including the Neandertal
the qPCR assays to avoid false positives from related envi- and Denisovan genome projects (Green et al., 2010;
ronmental microbes. For example, Harkins et al. (2015) Reich et al., 2010), basic techniques are common in most
showed that the initial design of a qPCR assay using two analyses. These include sample preparation and DNA
primers and a probe that targeted a specific MTBC extraction, which involve decontaminating the outer layer
sequence in the rpoB gene was not, in fact, specific to the of the ancient material before extracting DNA through a
MTBC. This assay also showed a low signal for other variety of methods that include traditional phenol chloro-
mycobacteria outside of the complex. Unfortunately, dam- form and silica-based filtration protocols. These are fol-
aged and/or inhibited ancient TB DNA can also show a lowed either by PCR and Sanger sequencing (common in
low signal. After taking advantage of new rpoB sequences older analyses) or by newer techniques such as DNA
from mycobacteria deposited in NCBI’s sequence database, library preparation, targeted capture (also known as tar-
GenBank, the qPCR primers and probe were redesigned geted enrichment), and NGS, which allows for deep, par-
(resulting in fewer positive samples that were more in line allel sequencing of one or more samples on a single lane/
with qPCR data from other markers as well as data from run of a DNA sequencer. Before briefly discussing each
targeted capture and sequencing). Importantly, this qPCR of these procedures, it is important to review the factors
process is used as a first evaluative step in determining that are thought to affect DNA preservation in a sample.
sample quality and which would then be subjected to the
next-generation methods described below to confirm the
result and allow evolutionary analyses. DNA Preservation
Many factors influence DNA preservation in biological
materials, including the environment, the source material,
Current Methods and the characteristics of the genetic material itself.
Typically, the quality and quantity of DNA isolated from
One of the main challenges of working with ancient DNA ancient material is better from tooth roots than from bone,
compared to modern DNA is that, over time, DNA and better from bone than mummified soft tissue (Alonso
degrades into shorter and shorter fragments, eventually et al., 2001; Kurosaki et al., 1993; O’Rourke et al., 1996;
becoming too small to assemble properly into identifiable Stone et al., 2001). Recent research suggests that the
sequences even with the most advanced bioinformatic petrous portion of the temporal bone and dental calculus
techniques. Early research analyzing ancient DNA used may harbor the best-preserved host DNA (Ozga et al.,
dried tissue (Higuchi et al., 1984; Pääbo, 1985) and bone 2016; Pinhasi et al., 2015). Environments with cooler
(Hagelberg et al., 1989). In recent years, additional mate- mean annual temperatures, neutral or slightly alkaline pH,
rials have been shown to be reservoirs for the preservation and dry conditions are best for DNA preservation,
of ancient genetic material including hair, coprolites although samples found in wet anoxic conditions (with
(paleofeces), and dental calculus (Black et al., 2011; neutral pH) have also yielded DNA (Campos et al., 2012;
Bonnichsen et al., 2001; Poinar et al., 1998). The analysis Hagelberg and Clegg, 1991; Holland et al., 1993; Höss,
of DNA from these ancient materials is feasible due to a 1994; Lawlor et al., 1991; Prüfer et al., 2014). Samples
number of advances in molecular genetics, the first of obtained from permafrost have yielded the oldest DNA
which was PCR (Mullis and Faloona, 1987; Saiki et al., sequences as well as the highest endogenous DNA con-
1988). More recently, new methods of DNA extraction, tent (Höss et al., 1994; Schwarz et al., 2009; Shapiro
library construction, DNA capture, DNA sequencing (spe- et al., 2004). In particular, permafrost has produced the
cifically NGS), and modern contamination quality control oldest ancient genome to date, that of a 750,000-year-old
checks have allowed the field of ancient genomics to Siberian horse (Orlando et al., 2013). DNA preservation
progress substantially. also may depend on the characteristics of the DNA.
186 Ortner’s Identification of Pathological Conditions in Human Skeletal Remains

Specifically, whether the DNA is mitochondrial, nuclear, decreases exponentially (Bar et al., 1988). To understand
or from pathogens with a lipid cell wall (such as TB) may the rate at which DNA degrades over time, Allentoft et al.
impact preservation (Donoghue et al., 2004; Higgins (2012) analyzed 158 radiocarbon-dated bones ranging in
et al., 2015; Nguyen-Hieu et al., 2012; Schuenemann age from B600 to 8000 before present (BP) and found
et al., 2013; Zink et al., 2002). In very cold environments, that the average DNA half-life was 521 years for a 242
theoretical analyses suggest that DNA as old as one mil- base pair (bp) mtDNA sequence. However, this was in a
lion years of age could be recovered (Millar and Lambert, relatively cool environment (burial temperature of
2013). However, the presence or absence of the condi- 13.1 C/55.5 F), and temperature appeared to explain only
tions for ancient DNA preservation noted here do not 38% of the variance in success rate of DNA recovery.
guarantee successful recovery, and poorly preserved bio- Relatively few studies have examined the rate of DNA
logical materials, against all odds, can produce usable degradation over time in different environments (Adler
ancient genetic material. et al., 2011; Colotte et al., 2009; Marota et al., 2002).
Discerning which skeletal tissue is best to sample in a Instead, most have focused on theoretical expectations
specific environmental context is difficult, and results (Smith et al., 2003; Willerslev et al., 2004). Other envi-
vary across studies. In research comparing DNA preserva- ronmental factors affecting DNA recovery include sub-
tion in bone, dentin, and cementum in Neolithic humans stances, such as fulvic acids, naturally found in soil, that
excavated in Germany, Adler et al. (2011) found that can be coextracted with the DNA to inhibit downstream
cementum contained the highest quantities of DNA, and PCR and make DNA analysis difficult, though many pro-
DNA extracted from teeth had a lower number of dam- tocols, such as silica-based purification columns address
aged sequences compared to bone. However, cementum these issues (Poinar et al., 1998; Tuross 1994). These
covers the roots of teeth in a very thin layer; thus, most studies of DNA preservation have focused primarily on
researchers undoubtedly include both cementum and den- host DNA rather than pathogen DNA.
tin in DNA extractions from the tooth root. As an addi- The preservation of pathogen DNA within an individ-
tional contamination control, many researchers remove ual is difficult to predict and is likely affected by patho-
the surface of the tooth root mechanically through abra- gen type and load at the time of death of the host, as well
sion or using diluted bleach (B5% sodium hypochlorite) as many of the environmental features noted above. Also,
prior to DNA extraction; this process likely removes pathogen load can vary substantially across tissues.
much of the cementum and potentially reduces DNA yield Examples of pathogen recovery from very old contexts
(Higgins et al., 2013). Comparisons of tooth types within include Helicobacter pylori from the Iceman (a 5300-
the mouth generally indicate that DNA is best preserved year-old frozen mummy from the Alps), Y. pestis from
in roots of the larger molars (Rubio et al., 2009) and in Eurasian Neolithic and Bronze Age individuals, and,
teeth that have not been compromised by caries or other recently, the presence of the periodontitis-associated “red
pathologies that damage enamel and would foster the complex” (Porphyromonas gingivalis, Tannerella for-
entry of environmental microbes into the interior of the sythia, Treponema denticola) in Neandertals (Andrades
tooth (Higgins and Austin, 2013; Higgins et al., 2011). Valtuena et al., 2017; Maixner et al., 2014, 2016;
Recently, the petrous portion of the temporal bone was Rasmussen et al., 2015; Weyrich et al., 2017). There is
reported to yield more DNA than other skeletal elements, some indication that for chronic diseases that result in
including teeth from archeological sites ranging in age bony lesions, the pathogen may be easiest to obtain from
from 800 to 10,000 years old (Gamba et al., 2014; Pinhasi the bony lesion itself or in nearby bone. However, the
et al., 2015). Newer methods of DNA extraction that lesions themselves, if osteoclastic, may be comprised of
recover small DNA fragments, as well as the use of fragile, less dense bone that results in poor DNA preser-
petrous portions and dental calculus, have also increased vation. Systemic infections, such as Y. pestis, can be
success rates in older contexts and warmer climates detected in tissues throughout the body, though tooth
(Glocke and Meyer, 2017; Meyer et al., 2016; Pinhasi roots and the pulp cavity are typically used, since they
et al., 2015). demonstrate better DNA preservation in general (Bos
Differing microenvironments, even within the same et al., 2011; Devault et al., 2014b). To date, pathogen
burial or excavation site, may cause varying success for DNA has not been reported from petrous temporal sam-
DNA analysis (Hagelberg and Clegg, 1991; Milos et al., ples even from systemic infections (Margaryan et al.,
2007; Stone and Stoneking, 1999). The temperature of the 2018). DNA from pathogens found in the gastrointestinal
surrounding environment and time since death are two system has been recovered from both dental calculus and
variables thought to play major roles in the maintenance coprolites (e.g., Cleeland et al., 2013; Iniguez et al., 2006;
of DNA integrity (Höss et al., 1996; Lindahl, 1993; Rubio Loreille et al., 2001; Warinner et al., 2014b), though
et al., 2009; Smith et al., 2001, 2003). For example, dur- respiratory and systemic pathogens could be expected to
ing the first 20 days postmortem, DNA in human tissues be found in such samples as well. For example, M.
Ancient DNA in the Study of Ancient Disease Chapter | 8 187

tuberculosis can be detected in both modern fecal and cal- or bleach solution. The sample should then be pulverized
culus samples (Eguchi et al., 2003; Kokuto et al., 2015; into dust or small pieces. This process can be carried out
Palakuru et al., 2012), though it has not yet been recov- using a scalpel, a hammer, a bone mill, or a slow-speed
ered from ancient fecal or calculus samples. Properties of drill (a dremel with a sterilized diamond wheel, for exam-
the pathogens themselves may also promote preservation ple). This step increases the surface area of the material,
or make them less likely to be recovered. For example, which improves the recovery of DNA during the extrac-
parasitic eggs built to withstand environmental challenges tion process. Because of the concerns about contamina-
after fecal deposition and mycobacteria with mycolic acid tion and false results, the procedures used to prepare
membranes may preserve better than many other patho- samples and prevent contamination have been the subject
gens (Loreille et al., 2001; Schuenemann et al., 2013), of considerable discussion and analysis in the ancient
while RNA viruses would be less likely to be recovered DNA community (e.g., Cooper and Poinar 2000;
because of the rapid degradation of RNA after death. To Malmstrom et al., 2007; Pilli et al., 2013).
date the only “ancient” RNA analyses have been from the Once the samples have been prepared, the next step is
1918 flu virus as discussed further below. the extraction of DNA from the material, whether that is
tissue, bone, tooth roots, dental calculus, coprolites, soil,
or hair (Adler et al., 2013; Gilbert et al., 2004; Hagelberg
Sample Preparation and DNA Extraction and Clegg, 1991; Ozga et al., 2016; Poinar et al., 1998;
One of the greatest concerns regarding ancient DNA anal- Rasmussen et al., 2011; Slon et al., 2017). The samples
yses is contamination from modern sources and the sur- must first be treated with solutions that allow the DNA to
rounding environment. As such, proper collection, be accessible and remove unwanted contaminants, includ-
storage, and preparation of samples are each important for ing amino acids and proteins. Most techniques for ancient
securing authentic DNA data. Ideally, samples should be DNA extraction use ethylenediaminetetraacetic acid
recovered from the field with the utmost caution so as not (EDTA) and proteinase K to inactivate DNases and digest
to introduce any spurious modern genetic material. If pos- proteins that may hinder proper extraction. The next steps
sible, the sample should be deposited into a sterile collec- of the extraction either take advantage of the fact that
tion container using gloved hands immediately upon most proteins are hydrophobic (and DNA is hydrophilic)
discovery. However, since this is not always possible dur- to successively separate the DNA from the other cell
ing field excavations, all samples must undergo further materials or make use of a binding agent to adhere the
decontamination protocols in the laboratory. All prepara- DNA to a filter and wash away all other elements. This
tion of biological materials should be carried out in a des- first process references phenol chloroform extraction,
ignated ancient DNA laboratory, one that is separate from while the second involves a silica-binding protocol, both
any modern DNA laboratory and thus free of amplified of which are still used regularly within ancient DNA labo-
DNA PCR products that could contaminate the extraction ratories. The phenol chloroform extraction method sepa-
process (Fig. 8.1). This ancient laboratory should have an rates the proteins and hydrophobic lipids from the nucleic
area designated for both sample preparation and extrac- acids and was first used for ancient DNA analyses
tion and have a filtered, enclosed airflow system and (Hagelberg and Clegg, 1991; Higuchi et al., 1984).
ultraviolet (UV) lighting. Sample preparation should be However, this method can also result in coextraction of
performed with gloves, masks, gowns, and sterile materi- inhibitors that can inhibit subsequent reactions; thus, the
als (including scalpels, saws, drills, and containers). use of a silica column which is more efficient at removing
These tools and workbenches should be cleaned regularly inhibitors is a recommended next step. The silica-based
with bleach, ethanol, and molecular grade H2O and UV extraction methods use chaotropic salts (primarily guani-
irradiated before and after each use. Samples should be dinium isothiocyanate or guanidinium hydrochloride) to
stored in a constant temperature environment (preferably catalyze the adsorption of DNA to silica as well as help
a cool dark place with low humidity), as DNA can digest proteins (Dabney et al., 2013; Höss and Pääbo, 1993;
degrade over time under poor storage conditions (Pruvost Rohland and Hofreiter, 2007; Yang et al., 1998). These
et al., 2007; Rubio et al., 2009). The removal of surface methods employ either a silica column or silica-binding
contaminants from samples should be carried out in a des- buffer and have been shown to maximize the recovery of
ignated enclosed area (preferably an out-of-use PCR endogenous DNA (Gamba et al., 2016). Of importance to
hood) in order to prevent any bone dust and debris from particularly degraded samples, the method developed by
entering the extraction area. The surface of the sample Dabney et al. (2013) increases the yield of very short DNA
should be removed by cutting or grinding away the fragments less than 80 bp in size. Extremely short frag-
exposed layers of bone or tooth root. Alternatively, the ments (B25 bp) have been recovered from Middle
surface can be sterilized by irradiating the surface with Pleistocene remains using both enhanced extraction and
UV light or by soaking the material in a hydrochloric acid library preparation methods (Glocke and Meyer, 2017).
188 Ortner’s Identification of Pathological Conditions in Human Skeletal Remains

FIGURE 8.1 A flow chart illustrating the


ancient DNA methods pipeline up to sequenc-
ing. Samples arrive at the laboratory and are
decontaminated using bleach and a UV cross-
linker; the outer layer may also be shaved off
using a Dremel at this point. Protective gear is
donned by the researcher and the sample is pul-
verized. DNA is extracted from the sample
depending on the chosen methodology.
Samples are assessed for quality and go onto
traditional PCR or qPCR probes, or alterna-
tively single/double-stranded library builds.
Samples are then indexed to a specific number
of cycles as designated by qPCR output or the
selected methodology. After another quality
check the samples can be sequenced outright
(referred to as shotgun sequencing) or captured
using homemade/manufactured biotinylated
baits which bind to amplified DNA and then to
a magnet, allowing the nontarget DNA to be
washed away. The target DNA can then
undergo another round of quality control in
order to determine whether more amplification
cycles are necessary before sequencing.

Preprepared extraction kits from biotechnology companies NGS Analyses


can be used, but these typically are not calibrated to the NGS was developed in response to the human genome
low quantities and small fragment sizes of ancient DNA. project in order to increase the throughput of DNA
It is also important to note that these methods will extract sequence analyses (Levy and Myers, 2016; Shendure
all of the DNA in the sample, which may include both et al., 2017; Shendure and Ji, 2008). Poinar et al. (2006)
host and pathogen DNA from the surrounding environ- were the first to apply NGS to ancient DNA research,
ment (Pedersen et al., 2015), along with the genetic mate- using it to obtain 13 million base pairs of DNA, including
rial of any human handlers (Leonard et al., 2007). the complete mtDNA genome from a Siberian woolly
Generally, less than 1% of the DNA present in the extract mammoth approximately 28,000 years old. This analysis
is endogenous to the host in question, and a much smaller used shotgun sequencing, a process where the DNA is
percentage belongs to any specific ancient pathogen. extracted, fragmented into sequences less than 200 300
DNA extraction methods are also available for soil, and base pairs in size (a step not necessary for ancient DNA),
nondestructive methods have been developed for bones, and sequenced using a high-throughput next-generation
teeth, and skin samples (Bolnick et al., 2012; Hofreiter, sequencer. NGS offers a number of advantages over
2012; Rohland et al., 2004; Slon et al., 2017).
Ancient DNA in the Study of Ancient Disease Chapter | 8 189

traditional Sanger sequencing and is available from bio- may result in high clonality, in which a few sequences
technology companies such as Illumina, Pacific dominate an entire sample pool. The primers used in the
Biosciences, Thermo Fisher Scientific (Ion Torrent), and library amplification process contain a series of unique
454 Life Sciences (GS FLX). Specifically, NGS can indices (barcodes) that allow for many samples to be run
sequence millions of DNA molecules in parallel, which in parallel on a single sequencing run (Kircher et al.,
decreases the overall time and cost of obtaining large 2012). The presence of unique barcode identifiers for
amounts of genetic data. For ancient DNA, NGS allows each sample allows for them to be demultiplexed (i.e.,
the investigator to obtain many independent sequence separated) using bioinformatic techniques after sequenc-
reads (i.e., sequence fragments) for a given stretch of ing has been completed. In other words, there is little to
genome without the need for standard PCR and cloning. no crossover between different samples during this pro-
The average number of reads per base pair is known as cess because the unique tags allow users to differentiate
the depth of coverage (so 10 3 depth of coverage means and examine each sample independently. Depending on
that a given base is covered on average by 10, nondupli- the quality of the DNA and the size of the genome in
cate independent sequences). Greater depth of coverage question, it may be necessary to sequence a single sample
results in higher confidence in the true sequence of the at a greater depth of coverage through multiple sequenc-
genome, including better identification of errors caused ing attempts. The creation of the DNA library is particu-
by DNA damage. In turn, this impacts downstream larly useful because it can be amplified and used many
genome reconstructions of host or microbial DNA, allow- times for different purposes (i.e., multiple sequencing
ing for more accurate evolutionary analyses. Another runs or by targeting different parts of the library such as
common term used in the discussion of NGS genome DNA from the host or a specific pathogen).
sequencing is genome coverage or coverage that reflects After amplifying the library, samples are essentially
the percentage of the reference genome that is covered by ready for shotgun sequencing or targeted capture (after
reads at a specific depth. Thus, a coverage of 98% is confirmation of quality and concentration through quality
excellent, while low coverage of only 30% could indicate checks with qPCRs or fragment analyzers). Shotgun
that DNA preservation was low or possibly that the refer- sequencing is completely untargeted, meaning that in
ence genome was not appropriate. Finally, NGS can also addition to host DNA, a number of other sources may be
sequence much smaller DNA fragments compared with detected including DNA from bacteria, fungi, and viruses
standard Sanger sequencing, which is biased toward lon- found in the host or from the burial environment. The
ger fragments to allow primers to anneal and copy main challenge with shotgun sequencing is that if the
properly. endogenous DNA content is very low in any given sam-
Typical workflow for ancient DNA NGS analyses fol- ple, the vast majority of sequencing reads will be from
lows one of two trajectories: the shotgun technique, which environmental microbes. Archeological samples generally
sequences all adapter-ligated DNA within a single sample have less than 1% endogenous DNA (Skoglund et al.,
without bias, and targeted capture, which uses a modern 2012). Exceptions include well-preserved samples, such
genome as “bait” to extract a taxonomically similar as those found in permafrost or caves, where the endoge-
ancient genome (Knapp and Hofreiter, 2010; Marciniak nous DNA content can be higher (e.g., Poinar et al., 2006;
et al., 2015). Both first require the construction of a DNA Rasmussen et al., 2010; Reich et al., 2010). Most analy-
library, and there are several methods for creating librar- ses, particularly of pathogens, use some form of targeted
ies from ancient DNA (Briggs and Heyn, 2012; Gansauge capture to enrich for the DNA of interest prior to sequenc-
et al., 2017; Gansauge and Meyer, 2013; Meyer et al., ing. Usually the pathogen to be targeted is known to be
2012; Rohland et al., 2015; Rohland and Reich, 2012). In present in the sample because of an initial shotgun
general, the DNA is end-repaired and annealed to a set of sequencing run, qPCR results, or through the use of a
adapters (one on each end) that are compatible with the microbial detection array. The latter use a type of targeted
NGS system (such as Illumina or Ion Torrent) that will be enrichment (with baits attached to a glass slide), but they
used for sequencing. Once the adaptors have been target many microbes rather than just one to assess what
adhered, the resulting DNA library is usually checked for is present in ancient samples (Bos et al., 2015; Devault
quality with qPCR in order to determine the extent to et al., 2014b). In order to target a specific pathogen, sev-
which DNA has been successfully recovered from the eral capture methods can be employed (Briggs et al.,
sample. This process also estimates the number of ampli- 2009a,b; Burbano et al., 2010; Carpenter et al., 2013; Fu
fications necessary to prepare the DNA for immediate et al., 2013; Stiller, 2012). At present, in-solution hybrid-
sequencing (shotgun) or targeted capture. This number is ization, using single-stranded DNA or RNA baits to
important because insufficient amplification results in few “fish” for complementary sequences is the most common.
copies of the fragments of interest, resulting in poor Targeted capture can result in much higher coverage of
sequencing results, and too many amplification cycles the desired genome accompanied with lower sequencing
190 Ortner’s Identification of Pathological Conditions in Human Skeletal Remains

costs due to the reduction of spurious/unwanted (Tremaroli and Backhed, 2012), vitamin production
sequences. (LeBlanc et al., 2013), education of the immune system
The sequencing process can be carried out in the labo- (Hooper and Macpherson, 2010), and defense against
ratory where the samples were prepared, or shipped to a infection (Brotman, 2011). In combination with ancient
number of sequencing centers around the world. DNA methods, the microbiome methods now allow for an
Operating and maintaining large sequencing machines can investigation into the evolution of host microbiomes
be costly and time-intensive, therefore many researchers across species and over time.
choose to outsource their sequencing to an offsite center, There are two primary methods for examining modern
which then returns the data to the laboratory for analysis. and ancient microbiomes: 16S ribosomal RNA (rRNA)
Depending on the sequencing method used and the sam- targeted amplification, which allows for taxonomic identi-
ples being examined, this can take anywhere from hours fication of bacteria and archaea, and shotgun metage-
to weeks to complete. Once sequencing is complete, the nomics, which is an untargeted amplification method that
data must be filtered to remove the low-quality sequences, allows for sequencing of all DNA present within a sample
the duplicates generated during library amplification, and regardless of its biological source. The first method tar-
any contamination from modern DNA sources. The data gets a hypervariable region within the ubiquitous 16S
are then mapped to reference sequences or de novo rRNA gene in order to distinguish microbial taxa without
assembled (when there is no reference available) to pro- the necessity of bacterial culturing (Woese and Fox,
duce a consensus sequence with identifiable variants 1977). There are nine hypervariable regions of differing
needed for subsequent phylogenetic and population sizes within the 16S rRNA gene, and individual sample
genetic analyses. These bioinformatics analyses typically barcoding allows for multiplexing and pooling of many
require specialized computer programs and scripts as well samples on a single sequencing run (Caporaso et al.,
as high-performance computing resources (Ginolhac 2010). These sequences (referred to as operational taxo-
et al., 2011; Jonsson et al., 2013; Louvel et al., 2016; nomic units, OTUs) are then clustered based on similarity
Peltzer et al., 2016; Schubert et al., 2014). The majority (97% or higher) which correlate to particular bacterial
of NGS analysis pipelines use command line scripting “species.” This method is very common in studies of
and prepackaged programs, though users can create modern human populations, but it is not without its flaws
scripts from the ground up in order to suit their data anal- when used with ancient material, especially considering
ysis needs. Samples may require multiple rounds of most ancient DNA sequences fall below the threshold
extraction, library construction, capture, and sequencing (,200 bp on average) of 16S rRNA variable region
to obtain sufficiently high-quality sequences for subse- detection. For example, the 16S V3 region has been
quent genetic analyses. NGS, along with the methods for shown not to properly reflect microbial taxa within
targeted capture, has transformed ancient DNA research, ancient DNA samples (Ziesemer et al., 2015) when com-
allowing host genomic analyses as well as the investiga- pared to shotgun metagenomic data. Despite these cau-
tion of symbiotic and pathogenic microbes found in hosts. tions, 16S analyses have still been used in a number of
ancient microbiome studies, including the Neandertal oral
cavity (Weyrich et al., 2017), the gut of Otzi, the
Microbiome Analyses Tyrolean iceman (Lugli et al., 2017), and 11th-century
The methods discussed above outline the primary ways pre-Columbian mummies (Santiago-Rodriguez et al.,
that ancient DNA is extracted and analyzed in the labora- 2015, 2016b). Most 16S rRNA data can be analyzed using
tory. However, another type of “targeted” analysis also command line custom scripts or through QIIME
benefiting from the technological advances associated (Quantitative Insights into Microbial Ecology) (Caporaso
with NGS focuses on the “microbiome”; recently, these et al., 2010). QIIME is an open-source bioinformatics
methods have been applied to ancient human gut and oral pipeline that assists in analyzing raw data from 16S rRNA
microbiomes. The term “microbiome” was first coined in reads. Ancient DNA analysis using 16S rRNA sequences
2001 to refer to the pathogenic and commensal microor- and QIIME remains a cost-effective, quick, and easy way
ganisms within and on a particular host (Lederberg and to characterize microbial taxa from a particular environ-
McCray, 2001). In 2007, the National Institutes of Health ment if DNA preservation is good and the biases are
initiated the Human Microbiome Project (HMP) with the considered.
goal of understanding and characterizing the many micro- The second method, which is especially popular for
biota in and on humans from across the globe and asses- degraded samples with average sequence lengths below
sing the influence these microbes have on overall host 300 bp, is known as shotgun metagenomics. Although
health (Group et al., 2009). Human microbiomes are more cost-prohibitive, nontargeted shotgun metagenomics
implicated in a wide range of metabolic, immunological, has several advantages: it goes beyond simple taxonomic
and developmental processes including host metabolism identification, it identifies other nonmicrobial organisms
Ancient DNA in the Study of Ancient Disease Chapter | 8 191

including parasites, fungi, and viruses, and it allows for M. lepromatosis appears to move more rapidly and diag-
studies of gene function. One drawback of this method is nostic skeletal symptoms have not been recognized in this
that there is still debate regarding the best way to analyze form. To date, M. lepromatosis has not been found in
and categorize the data. Whereas 16S rRNA can be ancient individuals, while M. leprae has been detected
binned into OTU groups, shotgun metagenomics is untar- using PCR assays (Haas et al., 2000; Rafi et al., 1994),
geted and therefore cannot give accurate taxonomic abun- SNP assays (Inskip et al., 2015; Watson and Lockwood,
dances. At present, QIIME is not properly tailored to 2009), and full genome sequences (Mendum et al., 2014;
examine shotgun reads, so a variety of methods are Schuenemann et al., 2013). Other than confirming the
employed depending on the biological sources, laborato- diagnosis of leprosy in an individual, questions have
ries, and the bioinformatics expertise of the user. included: When did M. leprae “jump” into humans (and
Additionally, methods for determining bacterial, archaeal, from what)? How are ancient and modern strains in dif-
and eukaryotic taxa can be computationally intensive ferent regions of the world related to each other? And
with shotgun data and the results should be carefully why did leprosy decline in frequency in Europe after the
examined so as not to incorporate environmental or host Medieval period? The last question is the most difficult to
contamination. If a particular microbe is found to be address, given that other factors unrelated to the biology
present in the host through microbiome analyses, tar- of the pathogen may have played the largest role in the
geted capture can be used to increase coverage and depth decline of leprosy in Europe (Boldsen, 2009; Manchester,
as discussed earlier. To authenticate ancient microbiome 1986, 1991; O’Neill, 1993; Roberts, 1986; Steinbock,
analyses, whether generated via 16S, shotgun, or targeted 1976). We do not see differences between the genomes of
capture, the overall damage patterns can be assessed ancient and modern strains that could explain this decline
(through MapDamage, if the target ancient genome has (Benjak et al., 2018; Schuenemann et al., 2013). Ancient
been mapped sufficiently to a reference) (Jonsson et al., 2013) DNA analyses have helped identify the likely time
and modern sequences can be removed through methods period during which M. leprae began affecting humans.
such as SourceTracker (within QIIME) (Caporaso et al., 2010). Specifically, Schuenemann et al. (2013) used the
sequences from the ancient samples to calibrate the
molecular clock. This calibration determined the mutation
APPLICATIONS OF ANCIENT DNA rate per time period, and thus, they could estimate the
time to the most recent common ancestor (TMRCA) of
Ancient DNA of Pathogens That Can Leave
all the M. leprae strains included in the analysis (both
Bony Changes: Leprosy, Tuberculosis, ancient and modern). The results showed that human M.
Brucellosis, Malaria, Syphilis leprae strains have a TMRCA of about 2800 BP and
A major focus of ancient pathogen genomics has been more recent phylogenetic analyses that include additional
infectious agents for diseases identified through the study strains (as well as M. lepromatosis) have shifted that
of diagnostic skeletal changes. Some bony changes, such somewhat to B4000 years BP (Benjak et al., 2018;
as those caused by anemia, have multiple causes includ- Schuenemann et al., 2018).
ing infectious disease. Ancient DNA studies can confirm To date, biogeographical analyses have been limited
the diagnosis or identify potential pathogens as well as to ancient samples from Europe, while modern M. leprae
provide additional data about the spread of pathogens and samples have been obtained from around the world.
their evolutionary history. Examples include the patho- Phylogenetic analyses of the ancient cases show multiple
gens causing leprosy, tuberculosis, malaria, and syphilis. branches of strains in Europe, even within the same ceme-
These examples also reveal some of the challenges for tery, but additional data from multiple time points and
examining ancient DNA from pathogens. geographic regions are needed to discern the origin and
assess how M. leprae subsequently spread. Data from
extant strains point to an origin in Asia or Oceania, while
Leprosy Schuenemann et al. (2018) also suggest Europe as the
Leprosy or Hansen’s disease can be caused by one of two potential geographic origin based on the diversity of
pathogens, M. leprae and Mycobacterium lepromatosis. Of ancient strains. DNA analyses of B150 extant human
these, M. leprae is the pathogen described by Hansen in strains and three nonhuman primate strains revealed that
1874, while M. lepromatosis was recently identified in the most basal lineages are found in the Philippines, New
patients primarily in the Americas (Han et al., 2008; Caledonia, Japan, Korea, and China (Benjak et al., 2018;
Hansen, 1874; Singh et al., 2015). Genome analyses show Honap et al., 2018; Schuenemann et al., 2013). Analyses
that these two mycobacteria diverged approximately 14 of strains from other species indicate exchange with
million years ago (Singh et al., 2015). Some of the clinical humans. For example, armadillos in the United States
and histological signals appear similar (Scollard, 2016); acquired leprosy from humans recently, and the genomic
192 Ortner’s Identification of Pathological Conditions in Human Skeletal Remains

analyses show that these strains are most closely related interspersed repetitive units-variable number of tandem
to branch 3I strains in Europe (Truman et al., 2011). An repeats (Kamerbeek et al., 1997; Supply et al., 2001), but
analysis of lesions in red squirrels on Brownsea Island in their use in evolutionary studies can be problematic
southern England suggests that humans gave M. leprae to because not all alleles are identical by descent and it is
them as well (also branch 3I), while the finding that red difficult to calculate accurate evolutionary rates for repeat
squirrels in several parts of the United Kingdom carry M. elements. These loci have been assessed in a few ancient
lepromatosis (which has not been identified in ancient or DNA studies (e.g., Zink et al., 2003), but one challenge
modern cases in Europe) is quite puzzling (Avanzi et al., for ancient DNA analyses is that it is difficult to discern
2016) and may suggest that rodents are the natural hosts whether the absence of a repeat is due to its actual
of these pathogens. Genome analyses of strains of M. absence in that TB strain or due to preservation issues.
leprae found in nonhuman primates from Africa and the With the introduction of NGS, modern TB genome
Philippines also suggest exchange with humans since they sequences and phylogenies confirmed that animal strains
cluster with human strains from the same geographic within the MTBC were derived and found that they were
region (Honap et al., 2018). Thus, a clear understanding nested with human strains in lineages 5 and 6, common in
of the reservoirs and/or original hosts of M. leprae and M. West Africa (Hershberg et al., 2008). Hershberg et al.
lepromatosis leading to human infection is currently not (2008) also proposed that human tuberculosis arose in
available. Africa and that the early lineages (1, 5, and 6) likely dis-
persed out of Africa with modern humans, followed by
the subsequent spread of “modern” lineages later through
Tuberculosis trade and colonization when human population densities
TB can be caused by any of the members of the MTBC were higher. Ancient DNA analyses of tuberculosis using
but most cases in humans are caused by M. tuberculosis. NGS first focused on strains in Europe (Bouwman et al.,
The origins of TB have long been debated with early 2012; Chan et al., 2013). Bouwman et al. (2012) used
scholars concluding that humans likely acquired TB dur- hybridization capture to target 260 regions in the TB
ing the domestication of cattle since spatial proximity genome, including 218 known SNPs, in DNA extracted
would have facilitated transfer of Mycobacterium bovis from a late-19th-century burial in St. George’s Church
(see Roberts and Buikstra, 2003; Stone et al., 2009). Crypt, Leeds, England. The results showed that hybridiza-
However, the identification of TB lesions in precontact tion capture could be used to obtain genetic data from
Native Americans who were not in contact with cattle ancient tuberculosis strains and that this strain was closely
cast doubt on this hypothesis (Buikstra, 1976; Daniel, related to H37Rv, a strain that was derived from a TB
2000). More recently, molecular evidence showed that M. patient from New York state in 1905 (Kubica et al.,
bovis is derived in comparison with M. tuberculosis 1972). Analyses of additional individuals from St.
(Brosch et al., 2002; Gordon et al., 1999). George’s Crypt as well as five other sites in the United
Ancient DNA analyses of tuberculosis first focused on Kingdom and one in France dating from the 2nd to 19th
confirming its diagnosis in skeletons in the Americas and centuries assessed 11 SNPS (Muller et al., 2014). Of 34
elsewhere by amplifying a fragment of a repetitive ele- samples tested, 6 individuals could be genotyped success-
ment, IS6110, found in all members of the MTBC but fully and compared to extant strains. Though the number
absent from other mycobacteria (e.g., Arriaza et al., 1995; of SNPs examined was limited, the results showed that all
Donoghue et al., 1998; Salo et al., 1994; Taylor et al., of the ancient strains appear to fall into lineage 4 (as
1996; Zink et al., 2001). More recent analyses of modern defined by Hershberg et al., 2008), common today in the
strains have shown that some lineage 1 strains have no Americas and Europe. Strains clustering in lineage 4 were
copies of IS6110 (see Fig. 8.2 for an explanation of the also found in Hungarian burials from the 17th century
TB lineages), and there is one example of IS6110 in a (Chan et al., 2013; Kay et al., 2015). Importantly, Kay
non-MTBC mycobacterial strain (Coros et al., 2008; et al. (2015) also demonstrated that most individuals are
Thierry et al., 1995; van Soolingen et al., 1993). Thus, infected by multiple strains, making analyses more chal-
this repeat element must be used with caution to identify lenging, and they estimate the TMRCA of lineage 4
ancient cases of TB. Additionally, care must be taken in strains to approximately AD 400.
the design of PCR primers or probes (for any pathogen) Questions about the source of ancient tuberculosis in
to be sure that related environmental pathogens are not the Americas and the timing of its origin in humans were
amplified, thus giving a false-positive signal (Harkins addressed by Bos et al. (2014). Using targeted capture to
et al., 2015; Müller et al., 2015). Genetic epidemiological recover the complete genome sequences of M. tuberculo-
analyses of TB from patients today often use analyses of sis from three ancient individuals from the Osmore River
the direct repeat locus (known as spoligotyping) or analy- Valley of Peru, they found that the ancient Peruvian
ses of microsatellite repeats called mycobacterial strains (Fig. 8.2) are most closely related to strains
Ancient DNA in the Study of Ancient Disease Chapter | 8 193

L1 M. tuberculosis L1-L7 FIGURE 8.2 A Bayesian maximum


L5 clade credibility tree of 261 MTBC gen-
omes, with estimated divergence dates in
years BP using a model of population
expansion. The lineages are defined
820–1571 L6
based on analyses of modern strains.
Lineage 1 (L1) strains are typically
1471–2286 found along the rim of the Indian Ocean
L7 1210–2364 Chimpanzee bacillus and in the Philippines. L2 and L3 strains
416–958 are found in East Asia and India/East
M. canettii 5
02
M. microti Africa, respectively. L4 strains are found
2–5
79 M. pinnipedii in Europe and the Americas today. L5
276 2 2510–4576 307–750
5–5 and L6 strains are found in West Africa,
104 1014–1294
1439–2510 and L7 strains were recently identified
MRCA in Ethiopia. Reproduced from Bos et al.
2659–4904 977–1183
2951–5339 Ancient Peruvian humans (2014).

L4
1779–3422
1254–2343 1653–3113
M. orygis
1271–2429
877–1717

L3 M. caprae

M. bovis

L2

adapted to sea mammals (specifically Southern was the pattern and pace of lineage 4 TB strain introduc-
Hemisphere seals and sea lions which carry MTBC strains tion in the Americas (and elsewhere) during colonization?
that are classified as Mycobacterium pinnipedii). This was What were the evolutionary forces that shaped tuberculo-
unexpected, and it suggests that tuberculosis first arrived sis diversity over time? And where (and from what spe-
in the Americas by jumping from South American seals cies) did M. tuberculosis first jump into humans?
to humans, likely through transmission during butchering
or consumption of undercooked seal meat. Phylogenetic
analyses and mutation rate estimation, calibrated using Brucellosis
the radiocarbon-dated samples, also allowed Bos et al. to Brucellosis is a zoonotic infection transmitted by Brucella
estimate the TMRCA of the MTBC and showed that this through direct contact with animals or their byproducts.
ancestor was more recent (only B3000 6000 years old) This disease can leave skeletal changes in humans that
than previous estimates, which ranged from B35,000 to may be confused with those left by tuberculosis, thus
B3 million years ago (Comas et al., 2013; Gutierrez aDNA offers a particular advantage to paleopathologists
et al., 2005; Hughes et al., 2002). Whether such pinniped- seeking to definitively diagnose such cases. Brucellosis
derived MTBC strains spread to inland parts of South has been identified in a few geographical areas throughout
America as well as North America by human-to-human the globe using ancient DNA techniques. Brucellosis
transmission, or whether different strains spread into DNA was detected in samples from the Middle Bronze
North America via another route, such as from Asia via Age Middle East (3500 BP), Butrint, Albania (800 1000
the Bering Strait during the Inuit-Aleut expansion (intro- BP), and from Medieval Italy (700 BP) (Kafil et al.,
duction through both routes could have occurred), is not 2014; Kay et al., 2014; Mutolo et al., 2012). Additionally,
known. These findings demonstrate the ability of the Kay et al. used shotgun sequencing to recover a 6.5 3
MTBC to infect different mammalian hosts because, after Brucella melitensis genome. They reported that the
the initial “jump” into humans 3000 6000 years ago, a Medieval strain from Sardinia is more closely related to
human strain jumped back into other animals, ultimately Italian strains and suggest an initial zoonotic origin from
including seals, who then eventually passed it back to sheep or goats (Kay et al., 2014). Brucellosis is a com-
humans in the Americas. How often this back and forth monly ignored public health concern found across histori-
has occurred, as well as the selective pressures on the cal texts but usually impacts younger individuals and only
mycobacteria after such jumps, are open questions. leaves skeletal evidence in 20% 85% of cases (Geyik
Additional questions about tuberculosis include: What et al., 2002; Rossetti et al., 2017).
194 Ortner’s Identification of Pathological Conditions in Human Skeletal Remains

Malaria classified as Treponema carateum, only affects the skin


Malaria, caused by several species of Plasmodium, has and does not cause skeletal lesions. The debate about the
likely affected humans (and other primates) for millennia, origin of syphilis centers on three major hypotheses:
though the environmental and settlement changes associ- whether syphilis originated in the Americas and then was
ated with the shift to agriculture likely intensified its brought to Europe by returning soldiers (the Columbian
impact on human populations. This is evidenced by esti- hypothesis), whether it arose in Europe or Africa and
mates of the ages of alleles that confer resistance and esti- spread (the pre-Columbian hypothesis), or whether it is an
mates of when these parasites likely became human- “heirloom” pathogen that spread with humans out of
specific (Hedrick, 2011; Loy et al., 2017; Verrelli et al., Africa and thus was present everywhere prior to European
2002). While malaria itself does not cause bony changes contact (the Unitarian hypothesis which also posits that
in the skeleton, chronic anemia associated with malaria syphilis, yaws, and endemic syphilis are caused by varia-
can leave nonspecific evidence detectable in the bioarch- tion in the same pathogen) (e.g., Baker and Armelagos,
eological record. In addition, some of the alleles that con- 1988; Cook and Powell, 2012; Gogarten et al., 2016;
fer resistance to malaria can also cause anemia (such as Harper et al., 2011).
sickle cell anemia and thalassemia), and these can be Early attempts to obtain ancient DNA from individuals
assessed in ancient skeletons. For example, to examine with treponemal diseases used PCR followed by Sanger
whether a child with skeletal pathologies consistent with sequencing or analysis of restriction fragment length poly-
severe anemia from a 3800-year-old cemetery in Israel morphisms that identified a single SNP characteristic of
had beta-thalassemia, Filon et al. (1995) used PCR to T. pallidum subsp. pallidum. Specifically, Kolman et al.
amplify a fragment of the β-globin gene. Hemoglobin, (1999) used both immunological and PCR analyses to
which transports oxygen in the blood, is comprised of confirm a case of syphilis in an individual from Easter
α-globin and β-globin chains. Individuals with thalasse- Island. However, several subsequent studies were not suc-
mia have a deficiency or complete lack of one of these cessful in the recovery of T. pallidum from ancient bones
two components. In this particular case, Filon et al. found (Barnes and Thomas, 2006; Bouwman and Brown, 2005;
that the child suffered from β-thalassemia major, since it von Hunnius et al., 2007). For example, Barnes and
was homozygous for a null mutation common in the Thomas (2006) noted the challenges in identifying both
Mediterranean that results in no production of the adult M. tuberculosis and T. pallidum given the available meth-
form of β-globin (most people begin producing the adult ods as well as the available comparative data from mod-
form early in childhood). This child also likely survived ern strains. In particular, since relatively few pathogen
to the age of eight because of elevated levels of fetal sequences were in the public databases and little variation
hemoglobin production. Additional studies have searched had been identified, they pointed out the difficulty in dis-
for mutations causing β-thalassemia in the Mediterranean tinguishing between contaminant vs. endogenous patho-
(Hughey et al., 2012; Sallares et al., 2004; Vigano et al., gen sequences. They tried to amplify and clone pathogen
2017). DNA from human remains with known causes of death
Direct testing for malaria in skeletons has been suc- due to syphilis or tuberculosis that were preserved in
cessful in only a few cases (Marciniak et al., 2016; medical collections, but the fragments amplified were
Sallares et al., 2003). Marciniak et al. tested Imperial PCR artifacts or from other bacteria. In addition to testing
period skeletons dating to the 1st 2nd centuries AD from medical and archeological bone samples with known
southern Italy and found that two individuals were syphilis or characteristic markers, von Hunnius et al.
infected with Plasmodium falciparum, which causes (2007) used rabbits at different stages of syphilis infection
severe malaria. The authors were able to capture B51% to test whether different tissues (including bone) showed
of the P. falciparum mtDNA genome in order to identify evidence of the pathogen. Despite the use of five different
the species causing disease, but, unfortunately, they were PCR assays targeting T. pallidum, none of the human
not able to obtain sufficient sequence for informative evo- bone samples yielded positive results and only the bone
lutionary analyses. from a rabbit in the acute state of infection (i.e., soon
after initial infection) tested positive, while those from
latent/chronic stages did not. Thus, by the time most peo-
Syphilis ple show skeletal lesions characteristic of syphilis, the
The causative agent of syphilis (both venereal and con- amount of bacteria in bones is likely very low and thus,
genital) as well as yaws and bejel (also known as endemic difficult to detect. More recently, Montiel et al. (2012)
syphilis) is Treponema pallidum, typically with the differ- posited that infants with congenital syphilis who died
ent diseases being associated with different subspecies of soon after birth would have higher systemic bacterial
T. pallidum (subspecies pallidum, pertenue, and endemi- loads and be the most likely to produce ancient syphilis
cum, respectively). A fourth treponemal disease, pinta, DNA sequences. They tested individuals buried in a crypt
Ancient DNA in the Study of Ancient Disease Chapter | 8 195

dating from the 16th and 17th centuries and, using PCR, population history. Specifically, Rasmussen et al. (2015)
amplified two SNPs specific to T. pallidum subsp. screened shotgun sequences from over 100 Bronze Age
pallidum. individuals from Eurasia, and they found seven indivi-
At present, the ancient genome data necessary to clar- duals, ranging in age from B2800 to 5000 years old and
ify the debate about the origin of syphilis are not yet geographically from Poland to the Altai region of Siberia,
available, and the data from modern pathogenic trepo- with some DNA sequences from Y. pestis. After intensive
nemes are also fairly limited, particularly for T. carateum, shotgun sequencing, they were able to recover the com-
whose genome has not been sequenced (Arora et al., plete genome sequences at a depth of 0.14 29.5 3 . In
2016; Gogarten et al., 2016). Data from modern strains addition, Andrades Valtuena et al. (2017) found similar-
indicate that T. pallidum subspecies pertenue and endemi- aged Y. pestis in other burials from Europe and the
cum cluster with each other and that the TMRCA for T. Caucasus using shotgun sequencing and targeted capture.
pallidum pallidum is estimated to be AD 1611 1859, These results were surprising because of the lack of his-
long after the first historical reports of syphilis (Arora torical or archeological evidence for plague during this
et al., 2016). This late date may reflect the limitations in period. Also, analyses of the Y. pestis genome sequences
geographic strain diversity in the available data, particu- indicated an older TMRCA (5783 years) as well as an
larly in strains from Africa, or it could reflect the success older divergence time (B55,000 years ago) between Y.
of a particular set of strains to the detriment of more viru- pestis and its closest relative Yersinia pseudotuberculosis
lent strains, since there is some indication of more severe (Rasmussen et al., 2015). However, as noted by Wagner
disease in early reports of syphilis. et al. (2014), there are significant challenges in accurately
dating divergence in Y. pestis. Importantly, the genome
data from Rasmussen et al. (2015) and Andrades
Mass Graves and “Invisible” Pathogens: Valtuena et al. (2017) also point to important aspects of
Smallpox, Plague, Cholera, Enteric the evolution of the pathogen. In particular, they show
Dysentery, and Flu that the genetic changes that facilitate infection of and
transmission by fleas were not present in the early strains
Many important diseases causing significant mortality in
of Y. pestis, but these changes are found after B3700 BP.
the past result in either relatively quick recovery or death,
For example, none of the Late Neolithic or Bronze Age
and thus, leave no mark on the skeleton. Most of the
strains have a mutation in the pla gene that is required for
information about these “plagues” comes to us from his-
developing severe bubonic plague, though Andrades
torical records, particularly in years with pandemics,
Valtuena et al. (2017) point out that a less severe form of
while archeological evidence is limited to occasional
bubonic plague may have been possible. Thus, cases of
mass graves and suggestions of depopulation. Recently,
plague in this period were likely either pneumonic or sep-
ancient DNA from mass graves (as well as some individ-
ticemic and acquired either through a zoonotic route (i.e.,
ual graves) is helping to identify the source of specific
rodents) or a less efficient and less severe transmission
historical pandemics, assess the evolution of strains over
via fleas. The introduction of plague into Europe during
the course of a pandemic, and discern why particular
the Late Neolithic may have been associated with the
strains were so virulent.
movement of people, such as Yamnaya pastoralists, from
the Central Asian steppes (Andrades Valtuena et al.,
Yersinia pestis 2017; Rasmussen et al., 2015).
Three major historical plague pandemics have been iden- The ancient DNA data from the first and second pan-
tified: the first pandemic also known as the Justinian pla- demics (i.e., the Justinian plague and Black Death, respec-
que (BAD 541 750), the second pandemic which began tively) show conclusively that both were caused by Y.
with the “Black Death” (BAD 1347 1351) and lasted pestis strains that do not differ significantly from modern
until the early 18th century, and the third pandemic (AD strains in terms of virulence and route of transmission
1855 1954) which generated the strains that persist today (Bos et al., 2011; Harbeck et al., 2013; Schuenemann
(Little, 2008). DNA data have confirmed that Y. pestis et al., 2011; Seifert et al., 2016; Spyrou et al., 2016;
was indeed the cause of all three historical plague pan- Wagner et al., 2014). These two pandemics had a severe
demics and also revealed that it impacted human popula- effect on many populations, likely because of factors such
tions as early as the Late Neolithic and Bronze Ages as a lack of understanding about germ theory and basic
(Andrades Valtuena et al., 2017; Bos et al., 2011; sanitation, the impact of preceding famines on susceptibil-
Rasmussen et al., 2015; Wagner et al., 2014). These earli- ity to infectious disease, coinfection by other pathogens/
est strains of the plague were not identified from mass parasites, and changes in the ecology of vector species
graves but from a general scan of the DNA extracted (e.g., DeWitte, 2015; Sallares, 2007). Analyses of
from individuals for the purpose of understanding human Justinian plague strains show that this strain is distinct
196 Ortner’s Identification of Pathological Conditions in Human Skeletal Remains

from the strain causing the Black Death, and it does not from two undated medical human tissue samples from a
appear to have any living descendants (Feldman et al., museum in Czech Republic. Through amino acid racemi-
2016; Wagner et al., 2014). The dating of the emergence zation, they estimated that these viruses were 60 120
of these strains is difficult because the substitution rate years old. They then used these estimated dates to cali-
varies, apparently speeding up during pandemics and then brate the molecular clock, obtaining an older time of AD
slowing during sylvatic cycles. This may be due to 1350 for the common ancestor. The accuracy of racemiza-
changes in selection patterns as the bacteria switch hosts, tion for estimating dates that then were used for calibra-
differences in the amount of replication during pandemic tion, however, has been questioned (Porter et al., 2017).
and endemic phases or other factors (Cui et al., 2013; In addition to difficulties in dating historical samples for
Wagner et al., 2014). In addition, a high-coverage calibration, the pox viruses have many insertion deletion
Justinian plague genome reconstructed by Feldman et al. polymorphisms that make alignment complicated, and
(2016) enabled a clear assessment of plasmids, substitu- these can also impact the estimates of divergence times.
tions, and structural changes, some of which include loci By carefully considering the insertion deletion differ-
that have been implicated in virulence. ences among strains and using a better reference genome,
Unlike the first pandemic strains, the second pandemic Smithson et al. (2017) created new genome alignments of
strains are at the root of a branch that includes the strains modern as well as the Lithuanian mummy variola virus
causing the third pandemic which persist today (Bos genomes. They then used these for new analyses that sug-
et al., 2011, 2012; Spyrou et al., 2016; Wagner et al., gest that the common ancestor of the existing variola
2014). SNP analyses of multiple individuals affected by virus genomes dates to AD 1470 1563 and may diverge
strains during the second pandemic show that these strains from the camelpox/taterapox clade around 1250 2000
were identical or very similar to one another, pointing to years earlier. Interestingly, these studies point to the role
its rapid spread during the Black Death (AD 1346 1353) of global trade in disseminating strains and also suggest
and subsequent persistence in Europe until the late 18th that variolation and vaccination had a major impact on
century (Bos et al., 2011, 2016; Haensch et al., 2010; strain diversity. While the molecular data currently point
Seifert et al., 2016). While there has been debate about to a recent origin, they are limited by the fact that these
whether the Y. pestis causing the second pandemic was may not fully represent the diversity of past strains. This
introduced multiple times or once and subsequently per- could account for the discrepancy between the molecular
sisted, these data suggest that the latter hypothesis is dates to a recent common ancestor and archeological and
likely correct. In addition, the data suggest that the second historical evidence pointing to older cases.
pandemic strains ultimately spread back into central/east
Asia and gave rise to the strains that started the third pan-
demic (Spyrou et al., 2016; Wagner et al., 2014). Food and Waterborne Outbreaks
While there is great interest in the major epidemics that
have affected humans, most deaths in the past were likely
Smallpox due to more mundane causes such as diarrhea and sepsis.
To date, the variola virus, which causes smallpox, is the In particular, water and foodborne bacteria and viruses
only human pathogen to be eradicated as a result of vacci- were major causes of mortality and morbidity prior to
nation efforts (Fenner et al., 1988). Although it is closely modern water treatment and hygienic food preservation/
related to the viruses causing taterapox (found in African preparation practices. Many of these cases were likely
gerbils) and camelpox (found in camels), where and when limited to one or a few families in a community.
the variola virus emerged as a human pathogen is However, severe outbreaks caused by pathogens such as
unknown (Esposito et al., 2006; Gubser et al., 2004; Li Vibrio cholerae, Salmonella typhi, and Shigella dysenter-
et al., 2007). Because smallpox does leave a mark on the iae, particularly in urban areas, can result in mass fatali-
skin (i.e., pox), the ancient DNA research has focused on ties. Cholera is well-known for its impact on cities in the
mummies with discernable evidence of disease. 19th century and for more recent outbreaks after natural
Specifically, analyses of European and Siberian cases, disasters or civil conflict such as in Haiti and Yemen
along with archived modern strains, show that the genetic (Hendriksen et al., 2011; Johnson, 2006; Qadri et al.,
diversity of strains is fairly low and point to a fairly 2017). Cholera is commonly transmitted via infected
recent origin (Biagini et al., 2012; Duggan et al., 2016; water or food, and it can kill people extraordinarily
Pajer et al., 2017). For example, in an evolutionary analy- quickly. Devault et al. (2014a) obtained DNA from a pre-
sis that also incorporated variola virus genome data from served intestinal sample from a victim of the second pan-
a Lithuanian mummy from the 17th century, Duggan demic in Philadelphia and recovered the genome of V.
et al. (2016) estimate the TMRCA of these strains to AD cholera. Their analyses showed that this strain had all of
1588 1645. Pajer et al. (2017) obtained genome data the major regions associated with virulence in classical
Ancient DNA in the Study of Ancient Disease Chapter | 8 197

strains. In addition, despite some structural differences, Alaskan victim of the pandemic who was buried in per-
the core genome was relatively conserved, pointing to mafrost and ultimately the sequences of the eight genes
selective constraints. Finally, they estimated the origin of comprising the coding portion of the genome were
the classical strains to between 1797 and 1813, in agree- obtained (Basler et al., 2001; Reid et al., 1999, 2000,
ment with historical accounts of the first cholera pan- 2004; Taubenberger et al., 1997, 2005). This in turn
demic in 1817. Analysis of cholera genomes is enabled the reconstruction of the virus so that it could be
challenging because of the high rate of recombination and characterized (Tumpey et al., 2005). These analyses
rapid mutation rate. As a result, estimating the origin of showed that the virus was very closely related to avian flu
all pathogenic V. cholera is difficult, and Devault et al. virus but with changes at key residues such that it could
suggest that it likely dates to the first epidemiological infect mammals. Interestingly, why the 1918 flu virus was
transition. so virulent is not completely understood. The hemaggluti-
Examples of waterborne and foodborne pathogens nin and neuraminidase genes, which are important for
from the archeological record are currently limited, but virus entry and exit from cells, do not show changes that
recently Vagene et al. (2018) identified Salmonella enteri- indicate increased virulence. It may be the combination of
ca subspecies enterica serovar Paratyphi C in individuals changes in the different genes that resulted in such a
from a contact period Mixtec epidemic burial ground at severe impact.
Teposcolula-Yucundaa in the state of Oaxaca, Mexico. More recently, Xiao et al. (2013) extracted RNA from
They extracted DNA from the pulp cavity of teeth from a formalin-fixed paraffin-embedded tissue sample dating
29 individuals (5 precontact and 24 postcontact). The to 1918 from a male who died at Camp Upton and used
postcontact burials were typically multiple burials, and this to construct a cDNA library. A cDNA library is com-
they dated to the cocoliztli epidemic in AD 1545 1550. prised of complementary DNA synthesized from RNA
To discern the cause of the epidemic, Vagene et al. templates; thus the library should be complementary to
employed a screening approach using shotgun sequencing RNA viruses and messenger RNA from the host and bac-
and the program MALT to identify possible pathogens. teria in the extract. This was then sequenced using NGS
They found 10 individuals with sequences from S. enteri- technology. They recovered the complete 1918 flu virus
ca. Following targeted capture, they recovered five gen- genome (at 3000 3 coverage) as well as gene expression
omes that clustered with S. enterica subspecies enterica sequences from bacteria, including Streptococcus pneu-
serovar Paratyphi C genomes in phylogenetic analyses. moniae, and from the host. S. pneumoniae causes pneu-
Of the five ancient genomes, two had higher coverage monia, which is commonly associated with influenza. The
and depth, allowing robust comparisons with modern analyses of sequences associated with host gene expres-
strains. The Paratyphi C strain of S. enterica causes sion showed an excess linked to inflammatory and cell
enteric fever and was likely introduced by an asymptom- death responses (Xiao et al., 2013). Analyses of this 1918
atic carrier from Europe. Such analyses can help shed flu genome found seven nonsynonymous changes com-
light on the epidemics that affected newly contacted pared with the 1918 flu genome sequenced previously
populations, particularly in the Americas. from Alaska. These analyses show some of the challenges
in understanding exactly why a pathogen is so virulent,
even in a well-studied pathogen.
Influenza
The influenza virus is another pathogen that can be the
cause of a pandemic. Flu pandemics occur roughly every “Invisible” Pathogens (to the
40 years, at times when there is an antigenic shift in the Paleopathological Record)
virus. The first clearly identified flu pandemic occurred in Most pathogenic infections leave no visible marks on
1580, though earlier cases and possible pandemics exist human skeletal material, even under ideal preservation
(Potter, 2001). Ancient DNA techniques have been used conditions, and many infections leave nondiagnostic skel-
to sequence genomes of the flu virus from the 1918 etal lesions or only leave diagnostic lesions in some frac-
(“Spanish” flu) pandemic, which was particularly virulent. tion of cases. As ancient DNA methods improve and the
The genome was stitched together over time using reverse number of samples tested increases, these hidden patho-
transcription PCR, since the flu virus is an RNA virus, gens are sometimes revealed. For example, sepsis, which
followed by Sanger sequencing. It was actually the first is a body response that occurs as result of an infection,
genome sequence from an “ancient” pathogen. The initial can be caused by a number of factors including the pres-
sequencing of fragments of several genes was performed ence of bacteria in the blood. Although soft tissue is
on a preserved sample from a US serviceman who died unlikely to preserve in the fossil record outside of anoxic
during the epidemic (Taubenberger et al., 1997). permafrost conditions, abscesses can sometimes calcify and
Subsequent analyses focused on lung tissue from an maintain within the skeletal record. Devault et al. (2017)
198 Ortner’s Identification of Pathological Conditions in Human Skeletal Remains

sequenced genetic material recovered from two miner- sequence looks very similar (rather than ancestral) to
alized “nodules” on the ribs of a Late Byzantine era modern HBV strains found in the Mediterranean basin, so
female from Troy, Anatolia (present day Turkey, dated authentication relies on DNA damage patterns and the
to 790 860 BP). These nodules tested positive for lack of HBV in controls (Patterson Ross et al., 2018). The
Gardnerella vaginalis, which is found in modern-day medieval HBV genome from Petersberg, Germany, also
pregnancy infections, and Staphylococcus saprophyti- clustered with strains found in the Mediterranean basin
cus, which is commonly present in urinary tract infec- (group D), while the Korean HBV sequence clusters with
tions, suggesting this individual suffered from maternal modern group C strains (Kahila Bar-Gal et al., 2012;
sepsis (Devault et al., 2017). Krause-Kyora et al., 2018). However, the sequences from
Another example of a pathogen that does not leave Neolithic individuals show that HBV has been in Europe
any skeletal evidence is H. pylori, a bacterium which is at least 7000 years, and these strains do not have close
found in the stomach of a majority of the world’s popula- modern relatives (falling between modern human and
tion (Atherton, 2006). However, it has been detected in nonhuman primate HBV strains) (Krause-Kyora et al.,
mummified soft tissue. Specifically, H. pylori genome 2018).
sequences have been obtained from two individuals who
were recovered from receding glaciers: Kwäday Dän
Ts’ı̀nchi (Long Ago Man Found) dating to AD Parasites and Commensals (Lice, Worms,
1670 1850, who was recovered in British Columbia and the Microbiome)
(Swanston et al., 2011) and Ötzi dating to the Cooper Initial ancient DNA analyses of parasites focused primar-
Age (B5300 BP) who was recovered in the Italian Alps ily on worms found in coprolites and on ectoparasites
(Maixner et al., 2016). In addition, H. pylori has been found on mummies or preserved clothing. More recently,
detected and characterized at the vacA gene in well- the gut and oral microbiomes have also been of interest,
preserved mummified individuals from 17th-century providing insights into dietary differences across human
Korea (Shin et al., 2018). Analyses of worldwide strains populations and time periods as well as identifying com-
of H. pylori show that it is geographically structured, mon oral or gut commensals and pathogens. This section
likely jumped into humans in Africa roughly will specifically examine the parasites on us (ectopara-
85,000 120,000 BP, and dispersed with humans out of sites), within us (using dental calculus), and excreted
Africa (Falush et al., 2003; Moodley et al., 2012). from us (coprolites or colon contents). For decades, the
Interestingly, both Kwäday Dän Ts’ı̀nchi and Ötzi have standard method of detection for ancient parasites within
H. pylori genomes that inform us about contact and or derived from host organisms was microscopy. Some
admixture between populations. Kwäday Dän Ts’ı̀nchi fossils date back millions of years including marine lice
carried H. pylori that was a recombinant of strains from (Urda rostrata) dated to 168 million years ago (Nagler
the Americas and Europe, while Ötzi’s H. pylori shows et al., 2017) and even a tongue worm (Invavita piratica)
that the admixture between central Asian and northeast found within microscopic crustaceans which dated back
African strains that produced the strains found in Europe to 425 million years ago (Siveter et al., 2015). However,
today occurred much more recently than previously many organisms cannot be identified by physical features
thought (Maixner et al., 2016; Swanston et al., 2011). alone and many parasite eggs were later found to be mis-
Finally, another example of a pathogen that does not classified (Cleeland et al., 2013).
leave skeletal evidence is hepatitis B virus (HBV). This
virus was recovered from a well-preserved child mummy
from Naples, Italy, dating to AD 1509 1629, a Korean Lice
mummy dating to AD 1612 1752, and from Neolithic Evolutionary analyses indicate that many parasites,
and medieval individuals from archeological sites in including lice, have been present throughout human his-
Germany (Kahila Bar-Gal et al., 2012; Krause-Kyora tory. For example, Pediculus humanus has fed on human
et al., 2018; Patterson Ross et al., 2018). The genome blood since at least 5 7 million years ago, diverging
sequence from the Italian mummy, as well as previously from chimpanzee lice (Pthirus pubis) (Reed et al., 2004).
published HBV data, indicate that the sequence changes More recently, the human louse diverged into two groups:
found in the data do not have a predictable rate; in other the head louse (P. humanus capitis) and body louse (P.
words, the mutations are not clock-like so molecular humanus humanus). Early molecular studies showed three
clock methods cannot be used to estimate divergence distinct clades of body lice on humans diverging 0.7 to 1
times (Patterson Ross et al., 2018). Because the child died million years ago (Reed et al., 2004). Additional studies,
in the 16th century, the virus clearly diversified before including samples derived from Pre-Columbian mummies
then. The lack of temporal signal also makes authenticat- (Raoult et al., 2008), demonstrated the presence of three
ing the ancient data more challenging. The ancient phylotypes distinguished by location on the human body
Ancient DNA in the Study of Ancient Disease Chapter | 8 199

instead of geography. Improved genetic methods resulted 2010; Obregon-Tito et al., 2015; Schnorr et al., 2014;
in the renaming of these “types” as “clades” and teased Yatsunenko et al., 2012) as well as through ancient DNA
out the relationship between ancient lice retrieved from analyses of past people. Presently, the absence of pro-
Roman Period combs (1st century AD to 6th century BC) cessed foods and antibiotics in the diet of indigenous
found at sites in Israel (Amanzougaghene et al., 2016). communities is thought to reflect the preindustrial/metro-
They confirmed that clade B lice existed in the Middle politan human microbiome. A number of studies have
East and thus questioned the hypothesis that clade B had examined the differences between traditional and industri-
an American origin since these combs dated prior to AD alized communities in both oral and gut bacterial taxo-
1492. Additional samples show a third Clade C that origi- nomies. One such example is the examination of the
nated in Africa and Asia and can even be traced along genus Treponema, which has been detected in extant
paths of human migration (Boutellis et al., 2014; Drali hunter-gatherers and 1000-year-old Mexican coprolites,
et al., 2015). Interestingly, pubic lice (P. pubis) belong to but is missing from healthy urban populations (Schnorr
a different clade that is more closely related to lice found et al., 2014; Tito et al., 2012). The precise reason behind
in gorillas (Reed et al., 2007). its absence is still not fully understood. It is currently
thought that the microbiota within the mammalian gut
Parasites in Feces coevolved with their respective hosts and have involve-
ment in both metabolism and immune response (Groussin
Fossilized fecal material, better known as coprolites, have
et al., 2017), but the manner by which microbes first
been found at many archeological sites across the globe
come to inhabit the human gut is still up for debate.
(Appelt et al., 2016) and date back as far as the Paleozoic
Additionally, through comparisons of the human micro-
era (270 million years ago) (Dentzien-Dias et al., 2013).
biome to those of other primates, Moeller and colleagues
Fecal samples have also been analyzed after removal
have been able to estimate divergence times of bacterial
from the intestinal tracts of mummies. Initial studies of
genomes during hominid evolution (Moeller et al., 2013,
fecal material involved macroscopic/microscopic analysis
2014, 2016). Studies suggest that although there are taxo-
and identification of species through phenotypic charac-
nomic overlaps in gut microbiota based on host geogra-
teristics. Prior to gut microbiome investigations, feces
phy, many strains have been vertically transmitted for an
were examined mainly in order to explore parasite distri-
unknown amount of time and evolved with the host itself.
bution across populations (Araujo et al., 2015; Leles
One of the earliest ancient microbiome studies exam-
et al., 2014). Coprolites from Rio Zape, Mexico (1400
ined a 1300 BP coprolite from Durango, Mexico (Tito
BP), underwent additional microscopic and 18S rRNA
et al., 2008). The resultant taxonomy showed that its
examination to detect Ascaris parasites (Cleeland et al.,
microbiome resembled modern feces both taxonomically
2013). Ascaris and other worms have been found in many
and functionally. This shotgun analysis was complemen-
other fecal samples, including those from pre-Columbian
ted later by 16S rRNA V3 analyses of other archeological
South America, Spanish mummies, Medieval burials from
coprolites from Hinds Cave (8000 BP, USA), Caserones
Belgium, and late-17th-century Koreans (Iniguez et al.,
(1600 BP, Chile), and Rio Zape (Tito et al., 2012). The
2006; Jaeger et al., 2016; Leles et al., 2008; Loreille
coprolites were shown to preserve DNA sufficiently well
et al., 2001; Oh et al., 2015; Shin et al., 2009). Many of
to allow Tito and colleagues to make comparisons with
these studies moved beyond microscopy to other high-
modern rural and cosmopolitan fecal microbiota commu-
throughput methods which allow for the genotyping of
nities. They found that the Hinds Cave coprolite micro-
ancient human gut parasites (Cote et al., 2016). New
biome did not resemble modern feces, while that from
methods may even allow for DNA from these parasites to
Caserones was similar to organic compost matter, indicat-
be recovered from soil in future studies, as was the case
ing that it did not preserve the original microbiome char-
for ancient human DNA recovered from environmental
acteristics. Subsequent studies of paleofeces discovered in
samples in Siberia (Slon et al., 2017).
Puerto Rico (AD 5 1170) showed that microbial and fun-
gal sequences can be used to identify unique cultural
The Gut Microbiome affiliations (Huecoid and Saladoid) as well as preserve
Fecal samples are also increasingly valuable for under- parasites from raw fish that were considered a dietary sta-
standing the ancient microbiome. In particular, how the ple for both communities (Cano et al., 2014).
microbiome has changed over time and across space is The growing field of ancient viromics has also exam-
important for understanding microbial, parasitic, and viral ined viruses in coprolites. Viruses or phages (viruses that
pathogenicity and the trajectory of human evolution infect bacteria) are an important component of the larger
(Schnorr et al., 2016). This topic has been addressed microbiome picture, particularly because they may regu-
through examination of modern remote indigenous human late many bacteria. They are thought to outnumber bacteria
communities and nonhuman primates (Contreras et al., by at least 10 or 100 to 1 (Chibani-Chennoufi et al., 2004).
200 Ortner’s Identification of Pathological Conditions in Human Skeletal Remains

In the first study of an ancient virome, a 14th-century investigated metabolites (Velsko et al., 2017). Velsko
coprolite from Belgium was found to include systemic et al. (2017) found that certain biological molecules
pathogens, intestinal parasites, and a number of bacterio- (monounsaturated and long-chain fatty acids) are more
phages (Appelt et al., 2014a,b). Such analyses can also prone to preservation compared to others (carbohydrates,
be performed in other animals. For example, frozen dipeptides, free amino acids, and free nucleotides).
Caribou coprolites dating from 700 to 3230 BP tested Recent reviews of research using dental calculus highlight
positive for a number of plant- and insect-infecting the variety of research questions that have been addressed
viruses (Ng et al., 2014). Another study confirmed the and potential directions for future studies (Huynh et al.,
presence of ancient bacteriophages in the intestinal tract 2016b; Warinner, 2016; Warinner et al., 2015; Weyrich
of 11th-century pre-Columbian Andean mummies, show- et al., 2015).
ing that they are preserved during the mummification
process (Santiago-Rodriguez et al., 2016a).
FUTURE PROSPECTS FOR ANCIENT
The Oral Microbiome PATHOGEN RESEARCH
The oral cavity is home to a diverse ecosystem of micro- Ancient DNA research has resulted in many surprises
scopic organisms, and this is often reflected in the dental over the past 30 years, including findings that Denisovans
calculus. The use of dental calculus as a source of DNA admixed with some groups of modern humans, that pla-
from ancient or degraded contexts is relatively new, but gue affected people as early as the Late Neolithic and
both host DNA and DNA from microbes found in the Bronze Ages, and that seals brought tuberculosis to South
mouth can be recovered (Adler et al., 2013; Black et al., America. Predicting what will be discovered next is diffi-
2011; Huynh et al., 2016a; Ozga et al., 2016; Preus et al., cult, but a number of major challenges, as well as stimu-
2011; Santiago-Rodriguez et al., 2017; Warinner et al., lating questions, remain. Some of the big questions
2014b; Weyrich et al., 2017). Dental calculus (sometimes include how did major demographic and subsistence
called tartar) is a hardened dental plaque that contains changes (such as during the Neolithic transition) change
minerals, bacterial cells, and food particles (Warinner human pathogens and the microbiome? Did agriculture
et al., 2015). It is commonly found today in people who and pastoralism foster zoonotic jumps (and did the distri-
do not have regular dental care, and it is fairly ubiquitous butions of pathogens in domesticated animals change)?
across the archeological record (Warinner et al., 2015) Additional questions relate to the impact of contact
even being found in Miocene apes (Hershkovitz et al., between populations: What new pathogens did modern
1997). DNA sequences extracted from European dental humans leaving Africa encounter? How did immune
calculus have shed light on the effects of the shift from alleles that introgressed from archaic humans into modern
hunter-gatherers to farming on the microbiome (Adler humans assist in adaptation to these new environments?
et al., 2013), changes in commensal and pathogen diver- What was the pace and extent of the “Columbian
sity including the absence of antibiotic resistance genes in Exchange”? What is the origin of syphilis? What is the
medieval Germany (Warinner et al., 2014b), and the role of major trade routes (such as the Silk Road) in
assessment of methanogen diversity and decreased spreading pathogens? Finally, there are questions related
Methanobrevibacter oralis in 14th 19th-century France to the biology of pathogens or their hosts: What are the
compared to modern populations (Huynh et al., 2016a). circumstances and adaptations that allow a zoonosis to
Dental calculus from pre-Columbian Puerto Rico also become a successful human pathogen? How did selective
showed distinct differences compared to modern calculus, pressures on the human immune system shift through
which may be reflective of shifts away from the horticul- time? How is the disease course influenced by coinfec-
turalist lifestyle and diet found in the past (Santiago- tions, and are certain pathogens more likely to cooccur?
Rodriguez et al., 2017). What characterizes pathogens thought to have coexisted
In addition, dental calculus has provided considerable with humans for millennia (i.e., “heirloom” pathogens)
information about diet from DNA sequence data and from versus pathogens thought to have jumped into humans
phytoliths, starches, and proteins embedded in the calcu- recently (i.e., “souvenir” pathogens) (Armelagos et al.,
lus (Cristiani et al., 2016; Hardy et al., 2009; Henry and 2005; Sprent, 1962) or is this just the same process with
Pipemo, 2008; Power et al., 2015; Radini et al., 2016; the difference being time? Why do some pathogens cause
Santiago-Rodriguez et al., 2017; Warinner et al., 2014a; skeletal lesions in some hosts but not in others?
Wesolowski et al., 2010). Such studies are important since To answer these questions and to better understand
dietary changes over time have had major implications health and disease in the past, insights from genetic data
for host health. Interestingly, a recent study of calculus (both ancient and modern) will need to be combined with
from modern and historic (200 BP) samples has also other data from paleopathology, osteology, and
Ancient DNA in the Study of Ancient Disease Chapter | 8 201

bioarcheology as well as archeology, history, and micro- microbiome) has taken and investigate how it changes
biology. For example, such data could be incorporated over geographic space and time. Reconstruction of
into host pathogen models of evolution to predict the ancient pathogens (in vivo or in silico) can be used to
consequences of introducing a pathogen into a system in assess why some strains of a virus or bacteria might be
which it does not currently exist (McCallum et al., 2001). more virulent than others. Laboratory and analytic meth-
While we cannot extract the reproductive ratio and many ods have made great strides since ancient DNA research
of the other parameters often used in epidemiological began in the 1980s, allowing for glimpses into aspects of
analyses directly from the archeological record, we can past population history long thought inaccessible.
adapt models to investigate how population movements, Methodological improvements now allow for the success-
novel technologies, and anthropogenic activities (such as ful recovery of DNA from many environments that are
human encroachment upon wildlife habitats) may have generally not conducive to DNA preservation and from
enhanced the risk of pathogen transmission among much older contexts. New materials, such as coprolites
humans. In tandem, we can examine how changes in the and calculus, provide a means of analyzing ancient micro-
pathogen might have facilitated transmission and spread. biome profiles as well as host DNA. Finally, there is also
The latter is possible through functional studies in modern the possibility of incorporating ancient protein and epige-
pathogens and through the use of genetic engineering to netic data into ancient disease research. In particular, pro-
recreate a pathogen in vitro under highly controlled condi- teins can be useful for indicating stress levels, immune
tions as was the case with the 1918 influenza virus. Not function, signs of starvation or obesity, and other physio-
surprisingly, the recreation of the 1918 flu virus was not logical information in the host (Jones et al., 2016;
without controversy (von Bubnoff, 2005). Sawafuji et al., 2017), while epigenetic data can provide
Significant challenges also remain for ancient patho- information about gene expression in the target tissue,
gen DNA research. Searching for pathogen DNA is very and these can also be related to disease. Such data can be
much like searching for a needle in a haystack, though important for understanding chronic noncommunicable
improvements in laboratory methods and bioinformatic disease progression and host susceptibility to disease in
techniques have made this search much easier. Despite general. In sum, ancient DNA research opens up new pos-
this, the search is relatively expensive since many sam- sibilities for assessing health and disease in the past and
ples must be deeply screened in order for pathogen DNA will likely offer many new surprises and insights in the
detection, and additional resources are needed to recover years to come.
a sufficient amount for evolutionary analyses. At present,
samples from many environments are not amenable to
ancient DNA analyses, and in many cases, they may REFERENCES
never be. This leads to a series of methodological ques- Adler, C.J., Haak, W., Donlon, D., Cooper, A., Consortium, T.G., 2011.
tions related to these challenges: Which tissues (and dur- Survival and recovery of DNA from ancient teeth and bones. J.
ing which stages of infection) are most likely to yield Archaeol. Sci. 38, 956 964.
pathogen DNA? Will methodological improvements allow Adler, C.J., Dobney, K., Weyrich, L.S., Kaidonis, J., Walker, A.W.,
the accurate characterization of RNA viruses? How do we Haak, W., et al., 2013. Sequencing ancient calcified dental plaque
shows changes in oral microbiota with dietary shifts of the Neolithic
identify pathogens that are extinct and distinguish them
and Industrial revolutions. Nat. Genet. 45 (4), 450 455. 455e451.
from benign environmental bacteria? What is the charac-
Allentoft, M.E., Collins, M., Harker, D., Haile, J., Oskam, C.L., Hale,
ter and distribution of environmental microbes around the M.L., et al., 2012. The half-life of DNA in bone: measuring decay
world (and how does this impact preservation in the kinetics in 158 dated fossils. Proc. Biol. Sci. 279 (1748),
archeological record)? Even if most of these challenges 4724 4733.
are surmounted, ancient DNA data do not always produce Alonso, A., Andelinovic, S., Martin, P., Sutlovic, D., Erceg, I., Huffine,
answers, as in the case of HBV. In addition, our under- E., et al., 2001. DNA typing from skeletal remains: evaluation of
standing of the microbial world is poor, with limited multiplex and megaplex STR systems on DNA isolated from bone
representation in DNA databases. Thus, there is a critical and teeth samples. Croat. Med. J. 42 (3), 260 266.
need to categorize microbes from many environments. Amanzougaghene, N., Mumcuoglu, K.Y., Fenollar, F., Alfi, S.,
Analyses of ancient pathogens (paleopathogenomics?) Yesilyurt, G., Raoult, D., et al., 2016. High Ancient genetic diversity
of human lice, Pediculus humanus, from Israel reveals new insights
are a new and increasingly productive area of research
into the origin of Clade B lice. PLoS One 11 (10), e0164659.
that provide insights into the evolutionary histories of the
Andrades Valtuena, A., Mittnik, A., Key, F.M., Haak, W., Allmae, R.,
many microbes that humans harbor. This can include Belinskij, A., et al., 2017. The stone age plague and its persistence
identifying the cause of known epidemics or pandemics in Eurasia. Curr. Biol. 27 (23), 3683 3691. e3688.
and refining case studies of specific individuals. More Appelt, S., Armougom, F., Le Bailly, M., Robert, C., Drancourt, M.,
importantly, it enables us to see the evolutionary steps 2014a. Polyphasic analysis of a middle ages coprolite microbiota,
that a pathogen or an ecosystem (if we think of the Belgium. PLoS One 9 (2), e88376.
202 Ortner’s Identification of Pathological Conditions in Human Skeletal Remains

Appelt, S., Fancello, L., Le Bailly, M., Raoult, D., Drancourt, M., dates and gene sequences form individual hairs. J. Archaeol. Sci. 28
Desnues, C., 2014b. Viruses in a 14th-century coprolite. Appl. (7), 775 785.
Environ. Microbiol. 80 (9), 2648 2655. Bos, K.I., Schuenemann, V.J., Golding, G.B., Burbano, H.A.,
Appelt, S., Drancourt, M., Le Bailly, M., 2016. Human coprolites as a Waglechner, N., Coombes, B.K., et al., 2011. A draft genome of
source for paleomicrobiology. Microbiol. Spectr. 4 (4), 59 74. Yersinia pestis from victims of the Black Death. Nature 478 (7370),
Araujo, A., Reinhard, K., Ferreira, L.F., 2015. Palaeoparasitology 506 510.
human parasites in Ancient material. Adv. Parasitol. 90, 349 387. Bos, K.I., Stevens, P., Nieselt, K., Poinar, H.N., Dewitte, S.N., Krause,
Armelagos, G.J., Brown, P.J., Turner, B., 2005. Evolutionary, historical J., 2012. Yersinia pestis: new evidence for an old infection. PLoS
and political economic perspectives on health and disease. Soc. Sci. One 7 (11), e49803.
Med. 61 (4), 755 765. Bos, K.I., Harkins, K.M., Herbig, A., Coscolla, M., Weber, N., Comas, I.,
Arora, N., Schuenemann, V.J., Jager, G., Peltzer, A., Seitz, A., Herbig, et al., 2014. Pre-Columbian mycobacterial genomes reveal seals as a
A., et al., 2016. Origin of modern syphilis and emergence of a pan- source of New World human tuberculosis. Nature 514, 494 497.
demic Treponema pallidum cluster. Nat. Microbiol. 2, 16245. Bos, K.I., Jager, G., Schuenemann, V.J., Vagene, A.J., Spyrou, M.A.,
Arriaza, B.T., Salo, W., Aufderheide, A.C., Holcomb, T.A., 1995. Pre- Herbig, A., et al., 2015. Parallel detection of ancient pathogens via
Columbian tuberculosis in northern Chile: molecular and skeletal array-based DNA capture. Philos. Trans. R. Soc. Lond. B Biol. Sci.
evidence. Am. J. Phys. Anthropol. 98, 37 45. 370 (1660), 20130375.
Atherton, J.C., 2006. The pathogenesis of Helicobacter pylori-induced Bos, K.I., Herbig, A., Sahl, J., Waglechner, N., Fourment, M., Forrest, S.
gastro-duodenal diseases. Annu. Rev. Pathol. 1, 63 96. A., et al., 2016. Eighteenth century Yersinia pestis genomes reveal
Avanzi, C., Del-Pozo, J., Benjak, A., Stevenson, K., Simpson, V.R., the long-term persistence of an historical plague focus. Elife 5,
Busso, P., et al., 2016. Red squirrels in the British Isles are infected e12994.
with leprosy bacilli. Science 354 (6313), 744 747. Boutellis, A., Abi-Rached, L., Raoult, D., 2014. The origin and distribu-
Baker, B.J., Armelagos, G.J., 1988. The origin and antiquite of syphilis. tion of human lice in the world. Infect. Genet. Evol. 23, 209 217.
Curr. Anthropol. 29, 703 737. Bouwman, A.S., Brown, T.A., 2005. The limits of biomolecular palaeo-
Bar, W., Kratzer, A., Machler, M., Schmid, W., 1988. Postmortem stabil- pathology: Ancient DNA cannot be used to study venereal syphilis.
ity of DNA. Forensic Sci. Int. 39 (1), 59 70. J. Archaeol. Sci. 32, 703 713.
Barnes, I., Thomas, M.G., 2006. Evaluating bacterial pathogen DNA Bouwman, A.S., Kennedy, S.L., Muller, R., Stephens, R.H., Holst, M.,
preservation in museum osteological collections. Proc. Biol. Sci. 273 Caffell, A.C., et al., 2012. Genotype of a historic strain of
(1587), 645 653. Mycobacterium tuberculosis. Proc. Natl. Acad. Sci. U.S.A. 109 (45),
Basler, C.F., Reid, A.H., Dybing, J.K., Janczewski, T.A., Fanning, T.G., 18511 18516.
Zheng, H., et al., 2001. Sequence of the 1918 pandemic influenza Briggs, A.W., Heyn, P., 2012. Preparation of next-generation sequencing
virus nonstructural gene (NS) segment and characterization of libraries from damaged DNA. Methods Mol. Biol. 840, 143 154.
recombinant viruses bearing the 1918 NS genes. Proc. Natl. Acad. Briggs, A.W., Good, J.M., Green, R.E., Krause, J., Maricic, T., Stenzel,
Sci. U.S.A. 98 (5), 2746 2751. U., et al., 2009a. Targeted retrieval and analysis of five Neandertal
Benjak, A., Avanzi, C., Singh, P., Loiseau, C., Girma, S., Busso, P., mtDNA genomes. Science 325 (5938), 318 321.
et al., 2018. Phylogenomics and antimicrobial resistance of the lep- Briggs, A.W., Good, J.M., Green, R.E., Krause, J., Maricic, T., Stenzel,
rosy bacillus Mycobacterium leprae. Nat. Commun. 9 (1), 352. U., et al., 2009b. Primer extension capture: targeted sequence
Béraud-Colomb, E., Roubin, R., Martin, J., Maroc, N., Gardeisen, A., retrieval from heavily degraded DNA sources. J. Vis. Exp. (31),
Trabuchet, G., et al., 1995. Human β-globin gene polymorphisms 1573.
characterized in DNA extracted from ancient bones 12,000 years Brosch, R., Gordon, S.V., Marmiesse, M., Brodin, P., Buchrieser, C.,
old. Am. J. Hum. Genet. 57, 1267 1274. Eiglmeier, K., et al., 2002. A new evolutionary scenario for the
Biagini, P., Theves, C., Balaresque, P., Geraut, A., Cannet, C., Keyser, Mycobacterium tuberculosis complex. Proc. Natl. Acad. Sci. U.S.A.
C., et al., 2012. Variola virus in a 300-year-old Siberian mummy. N. 99, 3684 3689.
Engl. J. Med. 367 (21), 2057 2059. Brotman, R.M., 2011. Vaginal microbiome and sexually transmitted
Black, J., Kerr, S., Henebry-Delfon, L., Lorenz, J.G., 2011. Dental calcu- infections: an epidemiologic perspective. J. Clin. Invest. 121 (12),
lus as an alternate source of mitochondrial DNA for analysis of skel- 4610 4617.
etal remains. SCA Proc. 25, 1 7. Buikstra, J.E., 1976. Prehistoric Tuberculosis in the Americas. Northwestern
Bogenhagen, D., Clayton, D.A., 1974. The number of mitochondrial University Archaeological Program, Evanston, IL, 182 pp.
deoxyribonucleic acid genomes in mouse L and human HeLa cells. Bunce, M., Oskam, C.L., Allentoft, M.E., 2012. Quantitative real-time
Quantitative isolation of mitochondrial deoxyribonucleic acid. J. PCR in aDNA research. In: Shapiro, B., Hofreiter, M. (Eds.),
Biol. Chem. 249 (24), 7991 7995. Ancient DNA: Methods and Protocols. Humana Press, California,
Boldsen, J.L., 2009. Leprosy in Medieval Denmark--osteological and pp. 121 132.
epidemiological analyses. Anthropol. Anz. 67 (4), 407 425. Burbano, H.A., Hodges, E., Green, R.E., Briggs, A.W., Krause, J.,
Bolnick, D.A., Bonine, H.M., Mata-Miguez, J., Kemp, B.M., Snow, M. Meyer, M., et al., 2010. Targeted investigation of the Neandertal
H., Leblanc, S.A., 2012. Nondestructive sampling of human skeletal genome by array-based sequence capture. Science 328 (5979),
remains yields ancient nuclear and mitochondrial DNA. Am. J. 723 725.
Phys. Anthropol. 147 (2), 293 300. Campos, P.F., Craig, O.E., Turner-Walker, G., Peacock, E., Willerslev,
Bonnichsen, R., Hodges, L., Ream, W., Field, K.G., Kirner, D.L., Selsor, E., Gilbert, M.T., 2012. DNA in ancient bone where is it located
K., et al., 2001. Methods for the study of ancient hair: radiocarbon and how should we extract it? Ann. Anat. 194 (1), 7 16.
Ancient DNA in the Study of Ancient Disease Chapter | 8 203

Cano, R.J., Rivera-Perez, J., Toranzos, G.A., Santiago-Rodriguez, T.M., Dabney, J., Knapp, M., Glocke, I., Gansauge, M.T., Weihmann, A.,
Narganes-Storde, Y.M., Chanlatte-Baik, L., et al., 2014. Nickel, B., et al., 2013. Complete mitochondrial genome sequence of
Paleomicrobiology: revealing fecal microbiomes of ancient indige- a Middle Pleistocene cave bear reconstructed from ultrashort DNA
nous cultures. PLoS One 9 (9), e106833. fragments. Proc. Natl. Acad. Sci. U.S.A. 110 (39), 15758 15763.
Caporaso, J.G., Kuczynski, J., Stombaugh, J., Bittinger, K., Bushman, F. Daniel, T.M., 2000. The origins and precolonial epidemiology of tuber-
D., Costello, E.K., et al., 2010. QIIME allows analysis of high- culosis in the Americas: can we figure them out? Int. J. Tuberc.
throughput community sequencing data. Nat. Methods 7 (5), Lung Dis. 4 (5), 395 400.
335 336. DeWitte, S.N., 2015. Setting the stage for medieval plague: pre-black
Carpenter, M.L., Buenrostro, J.D., Valdiosera, C., Schroeder, H., death trends in survival and mortality. Am. J. Phys. Anthropol. 158
Allentoft, M.E., Sikora, M., et al., 2013. Pulling out the 1%: whole- (3), 441 451.
genome capture for the targeted enrichment of ancient DNA Deguilloux, M.F., Ricaud, S., Leahy, R., Pemonge, M.H., 2011. Analysis
sequencing libraries. Am. J. Hum. Genet. 93 (5), 852 864. of ancient human DNA and primer contamination: one step back-
Champlot, S., Berthelot, C., Pruvost, M., Bennett, E.A., Grange, T., ward one step forward. Forensic Sci. Int. 210 (1 3), 102 109.
Geigl, E.M., 2010. An efficient multistrategy DNA decontamination Dentzien-Dias, P.C., Poinar Jr., G., de Figueiredo, A.E., Pacheco, A.C.,
procedure of PCR reagents for hypersensitive PCR applications. Horn, B.L., Schultz, C.L., 2013. Tapeworm eggs in a 270 million-
PLoS One 5 (9), e13042. Available from: https://doi.org/10.1371/ year-old shark coprolite. PLoS One 8 (1), e55007.
journal.pone.0013042. Devault, A.M., Golding, G.B., Waglechner, N., Enk, J.M., Kuch, M.,
Chan, J.Z., Sergeant, M.J., Lee, O.Y., Minnikin, D.E., Besra, G.S., Pap, Tien, J.H., et al., 2014a. Second-pandemic strain of Vibrio cholerae
I., et al., 2013. Metagenomic analysis of tuberculosis in a mummy. from the Philadelphia cholera outbreak of 1849. N. Engl. J. Med.
N. Engl. J. Med. 369 (3), 289 290. 370 (4), 334 340.
Chibani-Chennoufi, S., Bruttin, A., Dillmann, M.L., Brussow, H., 2004. Devault, A.M., McLoughlin, K., Jaing, C., Gardner, S., Porter, T.M.,
Phage-host interaction: an ecological perspective. J. Bacteriol. 186 Enk, J.M., et al., 2014b. Ancient pathogen DNA in archaeological
(12), 3677 3686. samples detected with a Microbial Detection Array. Sci. Rep. 4,
Cleeland, L.M., Reichard, M.V., Tito, R.Y., Reinhard, K.J., Lewis Jr., C. 4245.
M., 2013. Clarifying prehistoric parasitism from a complementary Devault, A.M., Mortimer, T.D., Kitchen, A., Kiesewetter, H., Enk, J.M.,
morphological and molecular approach. J. Archaeol. Sci. 40 (7), Golding, G.B., et al., 2017. A molecular portrait of maternal sepsis
3060 3066. from Byzantine Troy. Elife 6, e20983.
Colotte, M., Couallier, V., Tuffet, S., Bonnet, J., 2009. Simultaneous Donoghue, H.D., Spigelman, M., Zias, J., Gernaey-Child, A.M.,
assessment of average fragment size and amount in minute samples Minnikin, D.E., 1998. Mycobacterium tuberculosis complex DNA in
of degraded DNA. Anal. Biochem. 388, 345 347. calcified pleura from remains 1400 years old. Lett. Appl. Microbiol.
Comas, I., Coscolla, M., Luo, T., Borrell, S., Holt, K.E., Kato-Maeda, 27 (5), 265 269.
M., et al., 2013. Out-of-Africa migration and Neolithic coexpansion Donoghue, H.D., Spigelman, M., Greenblatt, C.L., Lev-Maor, G., Bar-
of Mycobacterium tuberculosis with modern humans. Nat. Genet. 45 Gal, G.K., Matheson, C., et al., 2004. Tuberculosis: from prehistory
(10), 1176 1182. to Robert Koch, as revealed by ancient DNA. Lancet Infect. Dis. 4
Contreras, M., Costello, E.K., Hidalgo, G., Magris, M., Knight, R., (9), 584 592.
Dominguez-Bello, M.G., 2010. The bacterial microbiota in the oral Drali, R., Mumcuoglu, K.Y., Yesilyurt, G., Raoult, D., 2015. Studies of
mucosa of rural Amerindians. Microbiology 156 (Pt 11), 3282 3287. Ancient lice reveal unsuspected past migrations of vectors. Am. J.
Cook, D.C., Powell, M.L., 2012. Treponematosis: past, present, and Trop. Med. Hyg. 93 (3), 623 625.
future. In: Grauer, A.L. (Ed.), A Companion to Paleopathology. Duggan, A.T., Perdomo, M.F., Piombino-Mascali, D., Marciniak, S., Poinar,
Blackwell Publishing Ltd, pp. 472 491. D., Emery, M.V., et al., 2016. 17(th) Century Variola virus reveals the
Cooper, A., Poinar, H.N., 2000. Ancient DNA: do it right or not at all. recent history of smallpox. Curr. Biol. 26 (24), 3407 3412.
Science 289, 1139. Eguchi, J., Ishihara, K., Watanabe, A., Fukumoto, Y., Okuda, K., 2003.
Coros, A., DeConno, E., Derbyshire, K.M., 2008. IS6110, a PCR method is essential for detecting Mycobacterium tuberculosis
Mycobacterium tuberculosis complex-specific insertion sequence, is in oral cavity samples. Oral Microbiol. Immunol. 18 (3), 156 159.
also present in the genome of Mycobacterium smegmatis, suggestive Enk, J., Rouillard, J.M., Poinar, H., 2013. Quantitative PCR as a predic-
of lateral gene transfer among mycobacterial species. J. Bacteriol. tor of aligned ancient DNA read counts following targeted enrich-
190 (9), 3408 3410. ment. BioTechniques 55 (6), 300 309.
Cote, N.M., Daligault, J., Pruvost, M., Bennett, E.A., Gorge, O., Esposito, J.J., Sammons, S.A., Frace, A.M., Osborne, J.D., Olsen-
Guimaraes, S., et al., 2016. A new high-throughput approach to Rasmussen, M., Zhang, M., et al., 2006. Genome sequence diversity
genotype Ancient human gastrointestinal parasites. PLoS One 11 and clues to the evolution of variola (smallpox) virus. Science 313
(1), e0146230. (5788), 807 812.
Cristiani, E., Radini, A., Edinborough, M., Boric, D., 2016. Dental calcu- Falush, D., Wirth, T., Linz, B., Pritchard, J.K., Stephens, M., Kidd, M.,
lus reveals mesolithic foragers in the Balkans consumed domesti- et al., 2003. Traces of human migrations in Helicobacter pylori
cated plant foods. Proc. Natl. Acad. Sci. U.S.A. 113 (37), populations. Science 299 (5612), 1582 1585.
10298 10303. Feldman, M., Harbeck, M., Keller, M., Spyrou, M.A., Rott, A.,
Cui, Y., Yu, C., Yan, Y., Li, D., Li, Y., Jombart, T., et al., 2013. Trautmann, B., et al., 2016. A high-coverage Yersinia pestis genome
Historical variations in mutation rate in an epidemic pathogen, from a sixth-century justinianic plague victim. Mol. Biol. Evol. 33
Yersinia pestis. Proc. Natl. Acad. Sci. U.S.A. 110 (2), 577 582. (11), 2911 2923.
204 Ortner’s Identification of Pathological Conditions in Human Skeletal Remains

Fenner, F., Henderson, D.A., Arita, I., Jezek, Z., Ladnyi, I.D., Gutierrez, M.C., Brisse, S., Brosch, R., Fabre, M., Omais, B., marmiesse,
Organization, W.H., 1988. Smallpox and Its Eradication. World M., et al., 2005. Ancient origin and gene mosaicism of the progeni-
Health Organization, Geneva. tor of Mycobacterium tuberculosis. PLoS Pathog. 1 (1), e5.
Filon, D., Faerman, M., Smith, P., Oppenheim, A., 1995. Sequence anal- Haas, C.J., Zink, A., Palfi, G., Szeimies, U., Nerlich, A.G., 2000. Detection
ysis reveals a β-thalassaemia mutation in the DNA of skeletal of leprosy in ancient human skeletal remains by molecular identifica-
remains from the archaeological site of Akhziv, Israel. Nat. Genet. tion of Mycobacterium leprae. Am. J. Clin. Pathol. 114 (3), 428 436.
9, 365 368. Haensch, S., Bianucci, R., Signoli, M., Rajerison, M., Schultz, M.,
Fu, Q., Meyer, M., Gao, X., Stenzel, U., Burbano, H.A., Kelso, J., et al., Kacki, S., et al., 2010. Distinct clones of Yersinia pestis caused the
2013. DNA analysis of an early modern human from Tianyuan black death. PLoS Pathog. 6 (10), e1001134.
Cave, China. Proc. Natl. Acad. Sci. U.S.A. 110 (6), 2223 2227. Hagelberg, E., Clegg, J.B., 1991. Isolation and characterization of DNA
Gamba, C., Jones, E.R., Teasdale, M.D., McLaughlin, R.L., Gonzalez- from archaeological bone. Proc. Biol. Sci. B 244, 45 50.
Fortes, G., Mattiangeli, V., et al., 2014. Genome flux and stasis in a Hagelberg, E., Sykes, B., Hedges, R., 1989. Ancient bone DNA ampli-
five millennium transect of European prehistory. Nat. Commun. 5, fied. Nature 342, 485.
5257. Han, X.Y., Seo, Y.H., Sizer, K.C., Schoberle, T., May, G.S., Spencer, J.
Gamba, C., Hanghoj, K., Gaunitz, C., Alfarhan, A.H., Alquraishi, S.A., S., et al., 2008. A new Mycobacterium species causing diffuse lepro-
Al-Rasheid, K.A., et al., 2016. Comparing the performance of three matous leprosy. Am. J. Clin. Pathol. 130 (6), 856 864.
ancient DNA extraction methods for high-throughput sequencing. Handt, O., Höss, M., Krings, M., Pääbo, S., 1994. Ancient DNA: meth-
Mol. Ecol. Resour. 16 (2), 459 469. odological challenges. Experientia 50, 524 529.
Gansauge, M.T., Meyer, M., 2013. Single-stranded DNA library prepara- Hansen, G.A., 1874. Undersøgelser angående spedalskhedens årsager.
tion for the sequencing of ancient or damaged DNA. Nat. Protoc. 8 Norsk Mag. Laegevidenskaben 3, 38.
(4), 737 748. Harbeck, M., Seifert, L., Hansch, S., Wagner, D.M., Birdsell, D., Parise,
Gansauge, M.T., Gerber, T., Glocke, I., Korlevic, P., Lippik, L., Nagel, K.L., et al., 2013. Yersinia pestis DNA from skeletal remains from
S., et al., 2017. Single-stranded DNA library preparation from highly the 6(th) century AD reveals insights into Justinianic Plague. PLoS
degraded DNA using T4 DNA ligase. Nucleic Acids Res. 45 (10), Pathog. 9 (5), e1003349.
e79. Hardy, K., Blakeney, T., Copeland, L., Kirkham, J., Wrangham, R.,
Geyik, M.F., Gur, A., Nas, K., Cevik, R., Sarac, J., Dikici, B., et al., Collins, M., 2009. Starch granules, dental calculus and new perspec-
2002. Musculoskeletal involvement of brucellosis in different age tives on ancient diet. J. Archaeol. Sci. 36 (2), 248 255.
groups: a study of 195 cases. Swiss Med. Wkly 132 (7 8), Harkins, K.M., Buikstra, J.E., Campbell, T., Bos, K.I., Johnson, E.D.,
98 105. Krause, J., et al., 2015. Screening ancient tuberculosis with qPCR:
Gilbert, M.T., Wilson, A.S., Bunce, M., Hansen, A.J., Willerslev, E., challenges and opportunities. Philos. Trans. R. Soc. Lond. B Biol.
Shapiro, B., et al., 2004. Ancient mitochondrial DNA from hair. Sci. 370 (1660), 20130622.
Curr. Biol. 14 (12), R463 R464. Harper, K.N., Zuckerman, M.K., Harper, M.L., Kingston, J.D.,
Ginolhac, A., Rasmussen, M., Gilbert, M.T., Willerslev, E., Orlando, L., Armelagos, G.J., 2011. The origin and antiquity of syphilis revisited:
2011. mapDamage: testing for damage patterns in ancient DNA an appraisal of Old World pre-Columbian evidence for treponemal
sequences. Bioinformatics 27 (15), 2153 2155. infection. Am. J. Phys. Anthropol. 146 (Suppl. 53), 99 133.
Glocke, I., Meyer, M., 2017. Extending the spectrum of DNA sequences Hedrick, P.W., 2011. Population genetics of malaria resistance in
retrieved from ancient bones and teeth. Genome Res. 27 (7), humans. Heredity (Edinb.) 107 (4), 283 304.
1230 1237. Heid, C.A., Stevens, J., Livak, K.J., Williams, P.M., 1996. Real time
Gogarten, J.F., Dux, A., Schuenemann, V.J., Nowak, K., Boesch, C., quantitative PCR. Genome Res. 6 (10), 986 994.
Wittig, R.M., et al., 2016. Tools for opening new chapters in the Hendriksen, R.S., Price, L.B., Schupp, J.M., Gillece, J.D., Kaas, R.S.,
book of Treponema pallidum evolutionary history. Clin. Microbiol. Engelthaler, D.M., et al., 2011. Population genetics of Vibrio choler-
Infect. 22 (11), 916 921. ae from Nepal in 2010: evidence on the origin of the Haitian out-
Gordon, S.V., Brosch, R., Billault, A., Garnier, T., Eiglmeier, K., Cole, break. MBio 2 (4), e00157-00111.
S.T., 1999. Identification of variable regions in the genomes of Henry, A.G., Pipemo, D.R., 2008. Using plant microfossils from dental
tubercle bacilli using bacterial artificial chromosome arrays. Mol. calculus to recover human diet: a case study from Tell al-Raqa’i,
Microbiol. 32 (3), 643 655. Syria. J. Archaeol. Sci. 35 (7), 1943 1950.
Green, R.E., Krause, J., Briggs, A.W., Maricic, T., Stenzel, U., Kircher, Hershberg, R., Lipatov, M., Small, P.M., Sheffer, H., Niemann, S.,
M., et al., 2010. A draft sequence of the Neandertal genome. Homolka, S., et al., 2008. High functional diversity in
Science 328 (5979), 710 722. Mycobacterium tuberculosis driven by genetic drift and human
Group, N.H.W., Peterson, J., Garges, S., Giovanni, M., McInnes, P., demography. PLoS Biol. 6 (12), e311.
Wang, L., et al., 2009. The NIH Human Microbiome Project. Hershkovitz, I., Kelly, J., Latimer, B., Rothschild, B.M., Simpson, S.,
Genome Res. 19 (12), 2317 2323. Polak, J., et al., 1997. Oral bacteria in miocene Sivapithecus. J.
Groussin, M., Mazel, F., Sanders, J.G., Smillie, C.S., Lavergne, S., Hum. Evol. 33 (4), 507 512.
Thuiller, W., et al., 2017. Unraveling the processes shaping mamma- Higgins, D., Austin, J.J., 2013. Teeth as a source of DNA for forensic iden-
lian gut microbiomes over evolutionary time. Nat. Commun. 8, tification of human remains: a review. Sci. Justice 53 (4), 433 441.
14319. Higgins, D., Kaidonis, J., Austin, J., Townsend, G., James, H., Hughes,
Gubser, C., Hue, S., Kellam, P., Smith, G.L., 2004. Poxvirus genomes: a T., 2011. Dentine and cementum as sources of nuclear DNA for use
phylogenetic analysis. J. Gen. Virol. 85 (Pt 1), 105 117. in human identification. Aust. J. Forensic Sci. 43 (4), 287 295.
Ancient DNA in the Study of Ancient Disease Chapter | 8 205

Higgins, D., Kaidonis, J., Townsend, G., Hughes, T., Austin, J.J., 2013. Jaeger, L.H., Gijon-Botella, H., Del Carmen Del Arco-Aguilar, M.,
Targeted sampling of cementum for recovery of nuclear DNA from Martin-Oval, M., Rodriguez-Maffiotte, C., Del Arco-Aguilar, M.,
human teeth and the impact of common decontamination measures. et al., 2016. Evidence of helminth infection in Guanche Mummies:
Investig. Genet. 4 (1), 18. integrating paleoparasitological and paleogenetic investigations. J.
Higgins, D., Rohrlach, A.B., Kaidonis, J., Townsend, G., Austin, J.J., Parasitol. 102 (2), 222 228.
2015. Differential nuclear and mitochondrial DNA preservation in Johnson, S., 2006. The Ghost Map: The Story of London’s Most
post-mortem teeth with implications for forensic and ancient DNA Terrifying Epidemic and How it Changed Science, Cities and the
studies. PLoS One 10 (5), e0126935. Modern World. Riverhead Books, London.
Higuchi, R., Bowman, B., Freiberger, M., Ryder, O.A., Wilson, A.C., Jones, J., Mirzaei, M., Ravishankar, P., Xavier, D., Lim do, S., Shin, D.
1984. DNA sequences from the quagga, an extinct member of the H., et al., 2016. Identification of proteins from 4200-year-old skin
horse family. Nature 312, 282 284. and muscle tissue biopsies from ancient Egyptian mummies of the
Hofreiter, M., 2012. Nondestructive DNA extraction from museum spe- first intermediate period shows evidence of acute inflammation and
cimens. Methods Mol. Biol. 840, 93 100. severe immune response. Philos. Trans. A Math. Phys. Eng. Sci. 374
Holland, M.M., Fisher, D.L., Mitchell, L.G., Rodriquez, W.C., Canik, J. (2079), 20150373. Available from: http://dx.doi.org/10.1098/
J., Merril, C.R., et al., 1993. Mitochondrial DNA sequence analysis rsta.2015.037.
of human skeletal remains: identification of remains from the Jonsson, H., Ginolhac, A., Schubert, M., Johnson, P.L., Orlando, L.,
Vietnam War. J. Forensic Sci. 38 (3), 542 553. 2013. mapDamage2.0: fast approximate Bayesian estimates of
Honap, T.P., Pfister, L.A., Housman, G., Mills, S., Tarara, R.P., Suzuki, ancient DNA damage parameters. Bioinformatics 29 (13),
K., et al., 2018. Mycobacterium leprae genomes from naturally 1682 1684.
infected nonhuman primates. PLoS Negl. Trop. Dis. 12 (1), Kafil, H.S., Hosseini, S.B., Sohrabi, M., Asgharzadeh, M., 2014.
e0006190. Brucellosis: presence of zoonosis infection 3 500 years ago in North
Hooper, L.V., Macpherson, A.J., 2010. Immune adaptations that main- of Iran. Asian Pacific J. Trop. Dis. 4, S684 S686.
tain homeostasis with the intestinal microbiota. Nat. Rev. Immunol. Kahila Bar-Gal, G., Kim, M.J., Klein, A., Shin, D.H., Oh, C.S., Kim, J.
10 (3), 159 169. W., et al., 2012. Tracing hepatitis B virus to the 16th century in a
Höss, M., 1994. Ancient DNA. Horm. Res. 43, 118 120. Korean mummy. Hepatology 56 (5), 1671 1680.
Höss, M., Pääbo, S., 1993. DNA extraction from Pleistocene bones by a Kamerbeek, J., Schouls, L., Kolk, A., van Agterveld, M., van Soolingen,
silica-based purification method. Nucleic Acids Res. 21 (16), D., Kuijper, S., et al., 1997. Simultaneous detection and strain differ-
3913 3914. entiation of Mycobacterium tuberculosis for diagnosis and epidemi-
Höss, M., Pääbo, S., Vereshchagin, N.K., 1994. Mammoth DNA ology. J. Clin. Microbiol. 35 (4), 907 914.
sequences. Nature 370, 333. Kay, G.L., Sergeant, M.J., Giuffra, V., Bandiera, P., Milanese, M.,
Höss, M., Jaruga, P., Zastawny, T.H., Dizdaroglu, M., Pääbo, S., 1996. Bramanti, B., et al., 2014. Recovery of a medieval Brucella meliten-
DNA damage and DNA sequence retrieval from ancient tissues. sis genome using shotgun metagenomics. MBio 5 (4), e01337-
Nucleic Acids Res. 24 (7), 1304 1307. 01314.
Hughes, A.L., Friedman, R., Murray, M., 2002. Genomewide pattern of Kay, G.L., Sergeant, M.J., Zhou, Z., Chan, J.Z., Millard, A., Quick, J.,
synonymous nucleotide substitution in two complete genomes of et al., 2015. Eighteenth-century genomes show that mixed infections
Mycobacterium tuberculosis. Emerg. Infect. Dis. 8 (11), were common at time of peak tuberculosis in Europe. Nat.
1342 1346. Commun. 6, 6717.
Hughey, J.R., Du, M., Li, Q., Michalodimitrakis, M., King, C., Debruyne, R., Kuch, M., Schwarz, C., Poinar, H., 2009. A
Stamatoyannopoulos, G., 2012. A search for beta thalassemia muta- quantitative approach to detect and overcome PCR inhibition in
tions in 4000 year old ancient DNAs of Minoan Cretans. Blood ancient DNA extracts. BioTechniques 47 (5), 941 949.
Cells Mol. Dis. 48 (1), 7 10. Kircher, M., Sawyer, S., Meyer, M., 2012. Double indexing overcomes
Huynh, H.T., Nkamga, V.D., Signoli, M., Tzortzis, S., Pinguet, R., inaccuracies in multiplex sequencing on the Illumina platform.
Audoly, G., et al., 2016a. Restricted diversity of dental calculus Nucleic Acids Res. 40 (1), e3.
methanogens over five centuries, France. Sci. Rep. 6, 25775. Knapp, M., Hofreiter, M., 2010. Next generation sequencing of Ancient
Huynh, H.T., Verneau, J., Levasseur, A., Drancourt, M., Aboudharam, DNA: requirements, strategies and perspectives. Genes 1 (2),
G., 2016b. Bacteria and archaea paleomicrobiology of the dental cal- 227 243.
culus: a review. Mol. Oral Microbiol. 31 (3), 234 242. Kokuto, H., Sasaki, Y., Yoshimatsu, S., Mizuno, K., Yi, L., Mitarai, S.,
Iniguez, A.M., Reinhard, K., Carvalho Goncalves, M.L., Ferreira, L.F., 2015. Detection of Mycobacterium tuberculosis (MTB) in fecal spe-
Araujo, A., Paulo Vicente, A.C., 2006. SL1 RNA gene recovery cimens from adults diagnosed with pulmonary tuberculosis using the
from Enterobius vermicularis ancient DNA in pre-Columbian human Xpert MTB/Rifampicin Test. Open Forum Infect. Dis. 2 (2), ofv074.
coprolites. Int. J. Parasitol. 36 (13), 1419 1425. Kolman, C.J., Centurion-Lara, A., Lukehart, S.A., Owsley, D.W.,
Inskip, S.A., Taylor, G.M., Zakrzewski, S.R., Mays, S.A., Pike, A.W., Tuross, N., 1999. Identification of Treponema pallidum subspecies
Llewellyn, G., et al., 2015. Osteological, biomolecular and geochem- pallidum in a 200-year-old skeletal sample. J. Infect. Dis. 180,
ical examination of an early anglo-saxon case of lepromatous lep- 2060 2063.
rosy. PLoS One 10 (5), e0124282. Krause-Kyora, B., Susat, J., Key, F.M., Kuhnert, D., Bosse, E., Immel,
Ivinson, A.J., Turnbull, L., David, R.A., Dyer, P.A., Elles, R., 1992. A., et al., 2018. Neolithic and Medieval virus genomes reveal com-
HLA Polymorphism in ancient Egyptian mummies. Eur. J. plex evolution of Hepatitis B. Elife 7, e36666. Available from:
Immunogen. 19, 461. https://doi.org/10.7554/eLife.36666.
206 Ortner’s Identification of Pathological Conditions in Human Skeletal Remains

Kubica, G.P., Kim, T.H., Dunbar, F.P., 1972. Designation of strain Malmstrom, H., Svensson, E.M., Gilbert, M.T., Willerslev, E.,
H37Rv as the neotype of Mycobacterium tuberculosis. Int. J. Syst. Gotherstrom, A., Holmlund, G., 2007. More on contamination: the
Evol. Microbiol. 22, 99 106. use of asymmetric molecular behavior to identify authentic ancient
Kurosaki, K., Matsushita, T., Ueda, S., 1993. Individual identification human DNA. Mol. Biol. Evol. 24 (4), 998 1004.
from ancient human remains. Am. J. Hum. Genet. 53, 638 643. Manchester, K., 1991. Tuberculosis and leprosy: evidence for interaction
Lawlor, D.A., Dickel, C.D., Hauswirth, W.W., Parham, P., 1991. of disease. In: Ortner, D.J., Aufderheide, A.C. (Eds.), Human
Ancient HLA genes from 7,500-year-old archaeological remains. Paleopathology: Current Syntheses and Future Options. Smithsonian
Nature 349, 785 788. Institution Press, Washington, DC, pp. 23 35.
LeBlanc, J.G., Milani, C., de Giori, G.S., Sesma, F., van Sinderen, D., Manchester, K.M., 1986. Tuberculosis and leprosy in antiquity: an inter-
Ventura, M., 2013. Bacteria as vitamin suppliers to their host: a pretation. Museum Appl. Sci. Center Archaeol. J. 4 (1), 22 30.
gut microbiota perspective. Curr. Opin. Biotechnol. 24 (2), Marciniak, S., Klunk, J., Devault, A., Enk, J., Poinar, H.N., 2015.
160 168. Ancient human genomics: the methodology behind reconstructing
Lederberg, J., McCray, P., 2001. ‘Ome Sweet’ Omics-a genealogical evolutionary pathways. J. Hum. Evol. 79, 21 34.
treasury of words. The Scientist 15, 8. Marciniak, S., Prowse, T.L., Herring, D.A., Klunk, J., Kuch, M.,
Leles, D., Araujo, A., Ferreira, L.F., Vicente, A.C., Iniguez, A.M., 2008. Duggan, A.T., et al., 2016. Plasmodium falciparum malaria in 1st-
Molecular paleoparasitological diagnosis of Ascaris sp. from copro- 2nd century CE southern Italy. Curr. Biol. 26 (23), R1220 R1222.
lites: new scenery of ascariasis in pre-Colombian South America Margaryan, A., Hansen, H.B., Rasmussen, S., Sikora, M., Moiseyev, V.,
times. Mem. Inst. Oswaldo Cruz 103 (1), 106 108. Khoklov, A., et al., 2018. Ancient pathogen DNA in human teeth
Leles, D., Cascardo, P., Freire Ados, S., Maldonado Jr., A., Sianto, L., and petrous bones. Ecol. Evol. 8 (6), 3534 3542.
et al., 2014. Insights about echinostomiasis by paleomolecular diag- Marota, I., Basile, C., Ubaldi, M., Rollo, F., 2002. DNA decay rate in
nosis. Parasitol. Int. 63 (4), 646 649. papyri and human remains from Egyptian archaeological sites. Am.
Leonard, J.A., Shanks, O., Hofreiter, M., Kreuz, E., Hodges, L., Ream, J. Phys. Anthropol. 117 (4), 310 318.
W., et al., 2007. Animal DNA in PCR reagents plagues ancient McCallum, H., Barlow, N., Hone, J., 2001. How should pathogen trans-
DNA research. J. Archaeol. Sci. 34 (9), 1361 1366. mission be modelled? Trends Ecol. Evol 16 (6), 295 300.
Levy, S.E., Myers, R.M., 2016. Advancements in next-generation Mendum, T.A., Schuenemann, V.J., Roffey, S., Taylor, G.M., Wu, H.,
sequencing. Annu. Rev. Genomics Hum. Genet. 17, 95 115. Singh, P., et al., 2014. Mycobacterium leprae genomes from a
Li, Y., Carroll, D.S., Gardner, S.N., Walsh, M.C., Vitalis, E.A., Damon, British medieval leprosy hospital: towards understanding an ancient
I.K., 2007. On the origin of smallpox: correlating variola phylo- epidemic. BMC Genomics 15 (1), 270.
genics with historical smallpox records. Proc. Natl. Acad. Sci. U.S. Meyer, M., Kircher, M., Gansauge, M.T., Li, H., Racimo, F., Mallick,
A. 104 (40), 15787 15792. S., et al., 2012. A high-coverage genome sequence from an archaic
Lindahl, T., 1993. Instability and decay of the primary structure of Denisovan individual. Science 338, 222 226.
DNA. Nature 362 (6422), 709 715. Meyer, M., Arsuaga, J.L., de Filippo, C., Nagel, S., Aximu-Petri, A.,
Little, L.K. (Ed.), 2008. Plague and the End of Antiquity. Cambridge Nickel, B., et al., 2016. Nuclear DNA sequences from the Middle
University Press, Cambridge. Pleistocene Sima de los Huesos hominins. Nature 531 (7595),
Loreille, O., Roumat, E., Verneau, O., Bouchet, F., Hanni, C., 2001. 504 507.
Ancient DNA from Ascaris: extraction amplification and sequences Millar, C.D., Lambert, D.M., 2013. Ancient DNA: towards a million-
from eggs collected in coprolites. Int. J. Parasitol. 31 (10), year-old genome. Nature 499 (7456), 34 35.
1101 1106. Milos, A., Selmanovic, A., Smajlovic, L., Huel, R.L., Katzmarzyk, C.,
Louvel, G., Der Sarkissian, C., Hanghoj, K., Orlando, L., 2016. Rizvic, A., et al., 2007. Success rates of nuclear short tandem repeat
metaBIT, an integrative and automated metagenomic pipeline for typing from different skeletal elements. Croat. Med. J. 48 (4),
analysing microbial profiles from high-throughput sequencing shot- 486 493.
gun data. Mol. Ecol. Resour. 16 (6), 1415 1427. Moeller, A.H., Peeters, M., Ndjango, J.B., Li, Y., Hahn, B.H., Ochman,
Loy, D.E., Liu, W., Li, Y., Learn, G.H., Plenderleith, L.J., H., 2013. Sympatric chimpanzees and gorillas harbor convergent gut
Sundararaman, S.A., et al., 2017. Out of Africa: origins and evolu- microbial communities. Genome Res. 23 (10), 1715 1720.
tion of the human malaria parasites Plasmodium falciparum and Moeller, A.H., Li, Y., Mpoudi Ngole, E., Ahuka-Mundeke, S., Lonsdorf,
Plasmodium vivax. Int. J. Parasitol. 47 (2 3), 87 97. E.V., Pusey, A.E., et al., 2014. Rapid changes in the gut microbiome
Lugli, G.A., Milani, C., Mancabelli, L., Turroni, F., Ferrario, C., during human evolution. Proc. Natl. Acad. Sci. U.S.A. 111 (46),
Duranti, S., et al., 2017. Ancient bacteria of the Otzi’s microbiome: 16431 16435.
a genomic tale from the Copper Age. Microbiome 5 (1), 5. Moeller, A.H., Caro-Quintero, A., Mjungu, D., Georgiev, A.V.,
Maixner, F., Thomma, A., Cipollini, G., Widder, S., Rattei, T., Zink, A., Lonsdorf, E.V., Muller, M.N., et al., 2016. Cospeciation of gut
2014. Metagenomic analysis reveals presence of Treponema dentico- microbiota with hominids. Science 353 (6297), 380 382.
la in a tissue biopsy of the Iceman. PLoS One 9 (6), e99994. Montiel, R., Solorzano, E., Diaz, N., Alvarez-Sandoval, B.A., Gonzalez-
Maixner, F., Krause-Kyora, B., Turaev, D., Herbig, A., Hoopmann, M. Ruiz, M., Canadas, M.P., et al., 2012. Neonate human remains: a
R., Hallows, J.L., et al., 2016. The 5300-year-old Helicobacter window of opportunity to the molecular study of ancient syphilis.
pylori genome of the Iceman. Science 351 (6269), 162 165. PLoS One 7 (5), e36371.
Malmstrom, H., Stora, J., Dalen, L., Holmlund, G., Gotherstrom, A., Moodley, Y., Linz, B., Bond, R.P., Nieuwoudt, M., Soodyall, H.,
2005. Extensive human DNA contamination in extracts from ancient Schlebusch, C.M., et al., 2012. Age of the association between
dog bones and teeth. Mol. Biol. Evol. 22 (10), 2040 2047. Helicobacter pylori and man. PLoS Pathog. 8 (5), e1002693.
Ancient DNA in the Study of Ancient Disease Chapter | 8 207

Muller, R., Roberts, C.A., Brown, T.A., 2014. Genotyping of ancient Palakuru, S.K., Lakshman, V.K., Bhat, K.G., 2012. Microbiological anal-
Mycobacterium tuberculosis strains reveals historic genetic diversity. ysis of oral samples for detection of Mycobacterium tuberculosis by
Proc. Biol. Sci. 281 (1781), 20133236. nested polymerase chain reaction in tuberculosis patients with peri-
Müller, R., Roberts, C.A., Brown, T.A., 2015. Complications in the odontitis. Dent. Res. J. (Isfahan) 9 (6), 688 693.
study of ancient tuberculosis: non-specificity of IS6110 PCRs. Sci. Patterson Ross, Z., Klunk, J., Fornaciari, G., Giuffra, V., Duchene, S.,
Technol. Archaeol. Res. 1 (1), 1 8. Duggan, A.T., et al., 2018. The paradox of HBV evolution as
Mullis, K., Faloona, F., Scharf, S., Saiki, R., Horn, G., Erlich, H., 1986. revealed from a 16th century mummy. PLoS Pathog. 14 (1),
Specific enzymatic amplification of DNA in vitro: the polymerase e1006750.
chain reaction. Cold Spring Harb. Symp. Quant. Biol. 51 (Pt 1), Pedersen, M.W., Overballe-Petersen, S., Ermini, L., Sarkissian, C.D.,
263 273. Haile, J., Hellstrom, M., et al., 2015. Ancient and modern environ-
Mullis, K.B., Faloona, F.A., 1987. Specific synthesis of DNA in vitro mental DNA. Philos. Trans. R. Soc. Lond. B Biol. Sci. 370 (1660),
via a polymerase-catalyzed chain reaction. Methods Enzymol. 155, 20130383.
335 350. Peltzer, A., Jager, G., Herbig, A., Seitz, A., Kniep, C., Krause, J., et al.,
Mutolo, M.J., Jenny, L.L., Buszek, A.R., Fenton, T.W., Foran, D.R., 2016. EAGER: efficient ancient genome reconstruction. Genome
2012. Osteological and molecular identification of brucellosis in Biol. 17, 60.
ancient Butrint, Albania. Am. J. Phys. Anthropol. 147 (2), 254 263. Pilli, E., Modi, A., Serpico, C., Achilli, A., Lancioni, H., Lippi, B.,
Nagler, C., Hyžný, M., Haug, J.T., 2017. 168 million years old “marine et al., 2013. Monitoring DNA contamination in handled vs. directly
lice” and the evolution of parasitism within isopods. BMC Evol. excavated ancient human skeletal remains. PLoS One 8 (1),
Biol. 17 (1), 76. e52524.
Ng, T.F., Chen, L.F., Zhou, Y., Shapiro, B., Stiller, M., Heintzman, P.D., Pinhasi, R., Fernandes, D., Sirak, K., Novak, M., Connell, S., Alpaslan-
et al., 2014. Preservation of viral genomes in 700-y-old caribou Roodenberg, S., et al., 2015. Optimal Ancient DNA yields from the
feces from a subarctic ice patch. Proc. Natl. Acad. Sci. U.S.A. 111 inner ear part of the human petrous bone. PLoS One 10 (6),
(47), 16842 16847. e0129102.
Nguyen-Hieu, T., Aboudharam, G., Drancourt, M., 2012. Heat degrada- Poinar, H., Hofreiter, M., Spaulding, W.G., Martin, P.S., Stankiewicz, B.
tion of eukaryotic and bacterial DNA: an experimental model for A., Bland, H., et al., 1998. Molecular coproscopy: dung and diet of
paleomicrobiology. BMC Res. Notes 5, 528. the extinct ground sloth Nothrotheriops shastensis. Science 281,
O’Neill, Y.V., 1993. Diseases of the middle ages. In: Kiple, K.F. (Ed.), 402 406.
The Cambridge World History of Human Disease. Cambridge Poinar, H.N., Schwarz, C., Qi, J., Shapiro, B., Macphee, R.D., Buigues,
University Press, Cambridge, pp. 270 279. B., et al., 2006. Metagenomics to paleogenomics: large-scale
O’Rourke, D.H., Carlyle, S.W., Parr, R.L., 1996. Ancient DNA: meth- sequencing of mammoth DNA. Science 311 (5759), 392 394.
ods, progress and perspectives. Am. J. Hum. Biol. 8, 557 571. Porter, A.F., Duggan, A.T., Poinar, H.N., Holmes, E.C., 2017. Comment:
Obregon-Tito, A.J., Tito, R.Y., Metcalf, J., Sankaranarayanan, K., Characterization of two historic smallpox specimens from a Czech
Clemente, J.C., Ursell, L.K., et al., 2015. Subsistence strategies in tradi- Museum. Viruses 9 (10), 276. Available from: https://doi.org/
tional societies distinguish gut microbiomes. Nat. Commun. 6, 6505. 10.3390/v9100276.
Oh, C.S., Seo, M., Hong, J.H., Chai, J.Y., Oh, S.W., Park, J.B., et al., Potter, C.W., 2001. A history of influenza. J. Appl. Microbiol. 91 (4),
2015. Ancient mitochondrial DNA analyses of Ascaris eggs discov- 572 579.
ered in coprolites from Joseon tomb. Korean J. Parasitol. 53 (2), Power, R.C., Salazar-Garcia, D.C., Wittig, R.M., Freiberg, M., Henry, A.
237 242. G., 2015. Dental calculus evidence of Tai Forest Chimpanzee plant
Orlando, L., Ginolhac, A., Zhang, G., Froese, D., Albrechtsen, A., consumption and life history transitions. Sci. Rep. 5, 15161.
Stiller, M., et al., 2013. Recalibrating Equus evolution using the Preus, H.R., Marvik, O.J., Selvig, K.A., Bennike, P., 2011. Ancient bac-
genome sequence of an early Middle Pleistocene horse. Nature 499 terial DNA (aDNA) in dental calculus from archaeological human
(7456), 74 78. remains. J. Archaeol. Sci. 38, 1827 1831.
Ozga, A.T., Nieves-Colon, M.A., Honap, T.P., Sankaranarayanan, K., Prüfer, K., Racimo, F., Patterson, N., Jay, F., Sankararaman, S., Sawyer,
Hofman, C.A., Milner, G.R., et al., 2016. Successful enrichment and S., et al., 2014. The complete genome sequence of a Neanderthal
recovery of whole mitochondrial genomes from ancient human den- from the Altai Mountains. Nature 505 (7481), 43 49.
tal calculus. Am. J. Phys. Anthropol. 160, 220 228. Pruvost, M., Schwarz, R., Correia, V.B., Champlot, S., Braguier, S.,
Pääbo, S., 1985. Molecular cloning of ancient Egyptian mummy DNA. Morel, N., et al., 2007. Freshly excavated fossil bones are best for
Nature 314, 644 645. amplification of ancient DNA. Proc. Natl. Acad. Sci. U.S.A. 104
Pääbo, S., 1989. Ancient DNA: extraction, characterization, molecular (3), 739 744.
cloning, and enzymatic amplification. Proc. Natl. Acad. Sci. U.S.A. Qadri, F., Islam, T., Clemens, J.D., 2017. Cholera in Yemen an
86, 1939 1943. old foe rearing its ugly head. N. Engl. J. Med. 377 (21),
Pääbo, S., Gifford, J.A., Wilson, A.C., 1988. Mitochondrial DNA 2005 2007.
sequences from a 7000-year old brain. Nucleic Acids Res. 16 (20), Radini, A., Buckley, S., Rosas, A., Estalrrich, A., de la Rasilla, M.,
9775 9787. Hardy, K., 2016. Neanderthals, trees and dental calculus: new evi-
Pajer, P., Dresler, J., Kabickova, H., Pisa, L., Aganov, P., Fucik, K., dence from El Sidron. Antiquity 90 (350), 290 301.
et al., 2017. Characterization of two historic smallpox specimens Rafi, A., Spigelman, M., Stanford, J., Lemma, E., Donoghue, H., Zias,
from a Czech Museum. Viruses 9 (8), 200. Available from: https:// J., 1994. Mycobacterium leprae DNA from ancient bone detected by
doi.org/10.3390/v9080200. PCR. Lancet 343, 1360 1361.
208 Ortner’s Identification of Pathological Conditions in Human Skeletal Remains

Raoult, D., Reed, D.L., Dittmar, K., Kirchman, J.J., Rolain, J.M., Sallares, R., 2007. Ecology, evolution, and epidemiology of plague.
Guillen, S., et al., 2008. Molecular identification of lice from pre- In: Little, L. (Ed.), Plague and the End of Antiquity. Cambridge
Columbian mummies. J. Infect. Dis. 197 (4), 535 543. University Press, Cambridge, pp. 231 289.
Rasmussen, M., Li, Y., Lindgreen, S., Pedersen, J.S., Albrechtsen, A., Sallares, R., Gomzi, S., Anderung, C., Bouwman, A., Brown, T.A.,
Moltke, I., et al., 2010. Ancient human genome sequence of an Robson-Brown, K.A., 2003. Identification of a malaria epidemic in
extinct Palaeo-Eskimo. Nature 463 (7282), 757 762. antiquity using ancient DNA. In: Robson-Brown, K.A. (Ed.),
Rasmussen, M., Guo, X., Wang, Y., Lohmueller, K.E., Rasmussen, S., Proceedings of the Archaeological Sciences ‘99 Conference, S1111
Albrechtsen, A., et al., 2011. An Aboriginal Australian genome reveals ed. Bristol University, Archaeopress, pp. 120 125.
separate human dispersals into Asia. Science 334 (6052), 94 98. Sallares, R., Bouwman, A., Anderung, C., 2004. The spread of malaria
Rasmussen, S., Allentoft, M.E., Nielsen, K., Orlando, L., Sikora, M., to Southern Europe in antiquity: new approaches to old problems.
Sjogren, K.G., et al., 2015. Early divergent strains of Yersinia pestis Med. Hist. 48 (3), 311 328.
in Eurasia 5,000 years ago. Cell 163 (3), 571 582. Salo, W.L., Aufderheide, A.C., Buikstra, J., Holcomb, T.A., 1994.
Reed, D.L., Smith, V.S., Hammond, S.L., Rogers, A.R., Clayton, D.H., Identification of Mycobacterium tuberculosis DNA in a pre-
2004. Genetic analysis of lice supports direct contact between mod- Columbian Peruvian mummy. Proc. Natl. Acad. Sci. U.S.A. 91,
ern and archaic humans. PLoS Biol. 2 (11), e340. 2091 2094.
Reed, D.L., Light, J.E., Allen, J.M., Kirchman, J.J., 2007. Pair of lice Santiago-Rodriguez, T.M., Fornaciari, G., Luciani, S., Dowd, S.E.,
lost or parasites regained: the evolutionary history of anthropoid pri- Toranzos, G.A., Marota, I., et al., 2015. Gut microbiome of an 11th
mate lice. BMC Biol. 5, 7. century A.D. pre-Columbian Andean Mummy. PLoS One 10 (9),
Reich, D., Green, R.E., Kircher, M., Krause, J., Patterson, N., Durand, E. e0138135.
Y., et al., 2010. Genetic history of an archaic hominin group from Santiago-Rodriguez, T.M., Fornaciari, G., Luciani, S., Dowd, S.E.,
Denisova Cave in Siberia. Nature 468 (7327), 1053 1060. Toranzos, G.A., Marota, I., et al., 2016a. Natural mummification of
Reid, A.H., Fanning, T.G., Hultin, J.V., Taubenberger, J.K., 1999. Origin the human gut preserves bacteriophage DNA. FEMS Microbiol.
and evolution of the 1918 “Spanish” influenza virus hemagglutinin Lett. 363 (1), fnv219.
gene. Proc. Natl. Acad. Sci. U.S.A. 96, 1651 1656. Santiago-Rodriguez, T.M., Fornaciari, G., Luciani, S., Dowd, S.E.,
Reid, A.H., Fanning, T.G., Janczewski, T.A., Taubenberger, J.K., 2000. Toranzos, G.A., Marota, I., et al., 2016b. Taxonomic and predicted
Characterization of the 1918 “Spanish” influenza virus neuramini- metabolic profiles of the human gut microbiome in pre-Columbian
dase gene. Proc. Natl. Acad. Sci. U.S.A. 97 (12), 6785 6790. mummies. FEMS Microbiol. Ecol. 92 (11), 182. Available from:
Reid, A.H., Fanning, T.G., Janczewski, T.A., Lourens, R.M., https://doi.org/10.1093/femsec/fiw182.
Taubenberger, J.K., 2004. Novel origin of the 1918 pandemic influ- Santiago-Rodriguez, T.M., Narganes-Storde, Y., Chanlatte-Baik, L.,
enza virus nucleoprotein gene. J. Virol. 78 (22), 12462 12470. Toranzos, G.A., Cano, R.J., 2017. Insights of the dental calculi
Richards, M.B., Sykes, B.C., Hedges, R.E.M., 1995. Authenticating DNA microbiome of pre-Columbian inhabitants from Puerto Rico. PeerJ
extracted from ancient skeletal remains. J. Archaeol. Sci. 22, 291 299. 5, e3277.
Roberts, C., Buikstra, J.E., 2003. The Bioarchaeology of Tuberculosis. A Sawafuji, R., Cappellini, E., Nagaoka, T., Fotakis, A.K., Jersie-
Global View on a Reemerging Disease. University Press of Florida, Christensen, R.R., Olsen, J.V., et al., 2017. Proteomic profiling of
Gainesville, FL, 343 pp. archaeological human bone. R. Soc. Open Sci. 4 (6), 161004.
Roberts, C.A., 1986. Leprosy and leprosaria in Medieval Britain. Schnorr, S.L., Candela, M., Rampelli, S., Centanni, M., Consolandi, C.,
Museum Appl. Sci. Center Archaeol. J. 4, 15 21. Basaglia, G., et al., 2014. Gut microbiome of the Hadza hunter-
Rohland, N., Hofreiter, M., 2007. Comparison and optimization of gatherers. Nat. Commun. 5, 3654.
ancient DNA extraction. BioTechniques 42 (3), 343 352. Schnorr, S.L., Sankaranarayanan, K., Lewis Jr, C.M., Warinner, C.,
Rohland, N., Reich, D., 2012. Cost-effective, high-throughput DNA 2016. Insights into human evolution from ancient and contemporary
sequencing libraries for multiplexed target capture. Genome Res. 22 microbiome studies. Curr. Opin. Genet. Dev. 41, 14 26.
(5), 939 946. Schubert, M., Ermini, L., Der Sarkissian, C., Jonsson, H., Ginolhac, A.,
Rohland, N., Siedel, H., Hofreiter, M., 2004. Nondestructive DNA Schaefer, R., et al., 2014. Characterization of ancient and modern
extraction method for mitochondrial DNA analyses of museum spe- genomes by SNP detection and phylogenomic and metagenomic
cimens. BioTechniques 36 (5), 814 816. , 818 821. analysis using PALEOMIX. Nat. Protoc. 9 (5), 1056 1082.
Rohland, N., Harney, E., Mallick, S., Nordenfelt, S., Reich, D., 2015. Schuenemann, V.J., Bos, K., DeWitte, S., Schmedes, S., Jamieson, J.,
Partial uracil-DNA-glycosylase treatment for screening of ancient Mittnik, A., et al., 2011. Targeted enrichment of ancient pathogens
DNA. Philos. Trans. R. Soc. Lond. B Biol. Sci. 370 (1660), yielding the pPCP1 plasmid of Yersinia pestis from victims of the
20130624. Black Death. Proc. Natl. Acad. Sci. U.S.A. 108 (38), E746 E752.
Rossetti, C.A., Arenas-Gamboa, A.M., Maurizio, E., 2017. Caprine bru- Schuenemann, V.J., Singh, P., Mendum, T.A., Krause-Kyora, B., Jager,
cellosis: a historically neglected disease with significant impact on G., Bos, K.I., et al., 2013. Genome-wide comparison of medieval
public health. PLoS Negl. Trop. Dis. 11 (8), e0005692. and modern Mycobacterium leprae. Science 341 (6142), 179 183.
Rubio, L., Martinez, L.J., Martinez, E., Martin de las Heras, S., 2009. Schuenemann, V.J., Avanzi, C., Krause-Kyora, B., Seitz, A., Herbig, A.,
Study of short- and long-term storage of teeth and its influence on Inskip, S., et al., 2018. Ancient genomes reveal a high diversity of
DNA. J. Forensic Sci. 54 (6), 1411 1413. Mycobacterium leprae in medieval Europe. PLoS Pathog. 14 (5),
Saiki, R.K., Gelfand, D.H., Stoffel, S., Scharf, S.J., Higuchi, R., Horn, e1006997.
G.T., et al., 1988. Primer-directed enzymatic amplification of DNA Schwarz, C., Debruyne, R., Kuch, M., McNally, E., Schwarcz, H.,
with a thermostable DNA polymerase. Science 239, 487 491. Aubrey, A.D., et al., 2009. New insights from old bones: DNA
Ancient DNA in the Study of Ancient Disease Chapter | 8 209

preservation and degradation in permafrost preserved mammoth Stiller, M., 2012. Case study: Targeted high-throughput sequencing of
remains. Nucleic Acids Res. 37 (10), 3215 3229. mitochondrial genomes from extinct cave bears via direct multi-
Scollard, D.M., 2016. Infection with Mycobacterium lepromatosis. Am. plex PCR sequencing (DMPS). Methods Mol. Biol. 840,
J. Trop. Med. Hyg. 95 (3), 500 501. 171 176.
Seifert, L., Wiechmann, I., Harbeck, M., Thomas, A., Grupe, G., Stone, A.C., Stoneking, M., 1993. Ancient DNA from a pre-Columbian
Projahn, M., et al., 2016. Genotyping Yersinia pestis in historical Amerindian population. Am. J. Phys. Anthropol. 92, 463 471.
plague: evidence for long-term persistence of Y. pestis in Europe Stone, A.C., Stoneking, M., 1999. Analysis of ancient DNA from a pre-
from the 14th to the 17th century. PLoS One 11 (1), e0145194. historic Amerindian cemetery. Philos. Trans. R. Soc. Lond. B Biol.
Shapiro, B., Drummond, A.J., Rambaut, A., Wilson, M.C., Matheus, P. Sci. 354 (1379), 153 159.
E., Sher, A.V., et al., 2004. Rise and fall of the Beringian steppe Stone, A.C., Starrs, J.E., Stoneking, M., 2001. Mitochondrial DNA anal-
bison. Science 306 (5701), 1561 1565. ysis of the presumptive remains of Jesse James. J. Forensic Sci. 46
Shendure, J., Ji, H., 2008. Next-generation DNA sequencing. Nature (1), 173 176.
Biotechnol. 26 (10), 1135 1145. Stone, A.C., Wilbur, A.K., Buikstra, J.E., Roberts, C.A., 2009.
Shendure, J., Balasubramanian, S., Church, G.M., Gilbert, W., Rogers, Tuberculosis and leprosy in perspective. Am. J. Phys. Anthropol.
J., Schloss, J.A., et al., 2017. DNA sequencing at 40: past, present 140 (Suppl. 49), 66 94.
and future. Nature 550 (7676), 345 353. Supply, P., Lesjean, S., Savine, E., Kremer, K., van Soolingen, D.,
Shin, D.H., Chai, J.Y., Park, E.A., Lee, W., Lee, H., Lee, J.S., et al., Locht, C., 2001. Automated high-throughput genotyping for study of
2009. Finding ancient parasite larvae in a sample from a male living global epidemiology of Mycobacterium tuberculosis based on myco-
in late 17th century Korea. J. Parasitol. 95 (3), 768 771. bacterial interspersed repetitive units. J. Clin. Microbiol. 39 (10),
Shin, D.H., Oh, C.S., Hong, J.H., Lee, H., Lee, S.D., Lee, E., 2018. 3563 3571.
Helicobacter pylori DNA obtained from the stomach specimens of Swanston, T., Haakensen, M., Deneer, H., Walker, E.G., 2011. The char-
two 17(th) century Korean mummies. Anthropol. Anz. 75, 75 87. acterization of Helicobacter pylori DNA associated with ancient
Singh, P., Benjak, A., Schuenemann, V.J., Herbig, A., Avanzi, C., human remains recovered from a Canadian glacier. PLoS One 6 (2),
Busso, P., et al., 2015. Insight into the evolution and origin of lep- e16864.
rosy bacilli from the genome sequence of Mycobacterium leproma- Taubenberger, J.K., Reid, A.H., Krafft, A.E., Bijwaard, K.E., Fanning,
tosis. Proc. Natl. Acad. Sci. U.S.A. 112 (14), 4459 4464. T.G., 1997. Initial genetic characterization of the 1918 “Spanish”
Siveter David, J., Briggs Derek, E.G., Siveter Derek, J., Sutton Mark, D., influenza virus. Science 275, 1793 1796.
2015. A 425-million-year-old silurian pentastomid parasitic on ostra- Taubenberger, J.K., Reid, A.H., Lourens, R.M., Wang, R., Jin, G.,
cods. Curr. Biol. 25 (12), 1632 1637. Fanning, T.G., 2005. Characterization of the 1918 influenza virus
Skoglund, P., Malmstrom, H., Raghavan, M., Stora, J., Hall, P., polymerase genes. Nature 437 (7060), 889 893.
Willerslev, E., et al., 2012. Origins and genetic legacy of Neolithic Taylor, G.M., Crossey, M., Saldanha, J., Waldron, T., 1996. DNA from
farmers and hunter-gatherers in Europe. Science 336 (6080), Mycobacterium tuberculosis identified in mediaeval human skeletal
466 469. remains using polymerase chain reaction. J. Archaeol. Sci. 23,
Slon, V., Hopfe, C., Weiss, C.L., Mafessoni, F., de la Rasilla, M., 789 798.
Lalueza-Fox, C., et al., 2017. Neandertal and Denisovan DNA from Thierry, D., Chavarot, P., Marchal, G., Le Thi, K.T., Ho, M.L., Nguyen,
Pleistocene sediments. Science 356 (6338), 605 608. N.L., et al., 1995. Mycobacterium tuberculosis strains unidentified
Smith, C.I., Chamberlain, A.T., Riley, M.S., Cooper, A., Stringer, C.B., using the IS6110 probe can be detected by oligonucleotides derived
Collins, M.J., 2001. Neanderthal DNA. Not just old but old and from the Mt308 sequence. Res. Microbiol. 146 (4), 325 328.
cold? Nature 410 (6830), 771 772. Tito, R.Y., Macmil, S., Wiley, G., Najar, F., Cleeland, L., Qu, C., et al.,
Smith, C.I., Chamberlain, A.T., Riley, M.S., Stringer, C., Collins, M.J., 2008. Phylotyping and functional analysis of two ancient human
2003. The thermal history of human fossils and the likelihood of microbiomes. PLoS One 3 (11), e3703.
successful DNA amplification. J. Hum. Evol. 45 (3), 203 217. Tito, R.Y., Knights, D., Metcalf, J., Obregon-Tito, A.J., Cleeland, L.,
Smithson, C., Imbery, J., Upton, C., 2017. Re-assembly and analysis of Najar, F., et al., 2012. Insights from characterizing extinct human
an ancient variola virus genome. Viruses 9 (9), 253. Available from: gut microbiomes. PLoS One 7 (12), e51146.
https://doi.org/10.3390/v9090253. Tremaroli, V., Backhed, F., 2012. Functional interactions between the
Spigelman, M., Lemma, E., 1993. The use of the polymerase chain reac- gut microbiota and host metabolism. Nature 489 (7415), 242 249.
tion (PCR) to detect Mycobacterium tuberculosis in ancient skele- Truman, R.W., Singh, P., Sharma, R., Busso, P., Rougemont, J., Paniz-
tons. Int. J. Osteoarchaeol. 3, 137 143. Mondolfi, A., et al., 2011. Probable zoonotic leprosy in the southern
Sprent, J.F.A., 1962. Parasitism, immunity and evolution. In: Leeper, G. United States. N. Engl. J. Med. 364 (17), 1626 1633.
S. (Ed.), The Evolution of Living Organisms. Melbourne University Tumpey, T.M., Basler, C.F., Aguilar, P.V., Zeng, H., Solorzano, A.,
Press, Melbourne, pp. 149 165. Swayne, D.E., et al., 2005. Characterization of the reconstructed
Spyrou, M.A., Tukhbatova, R.I., Feldman, M., Drath, J., Kacki, S., 1918 Spanish influenza pandemic virus. Science 310 (5745), 77 80.
Beltran de Heredia, J., et al., 2016. Historical Y. pestis genomes Tuross, N., 1994. The biochemistry of ancient DNA in bone. Experientia
reveal the European black death as the source of ancient and modern 50, 530 535.
plague pandemics. Cell Host Microbe 19 (6), 874 881. Vagene, A.J., Herbig, A., Campana, M.G., Robles Garcia, N.M.,
Steinbock, R.T., 1976. Leprosy. Paleopathological Diagnosis and Warinner, C., Sabin, S., et al., 2018. Salmonella enterica genomes
Interpretation: Bone Diseases in Ancient Human Populations. from victims of a major sixteenth-century epidemic in Mexico. Nat.
Charles C. Thomas, Springfield, IL, pp. 192 212. Ecol. Evol. 2, 520 528.
210 Ortner’s Identification of Pathological Conditions in Human Skeletal Remains

van Soolingen, D., de Haas, P.E., Hermans, P.W., Groenen, P.M., van Wesolowski, V., de Souza, S.M.F.M., Reinhard, K.J., Ceccantini, G.,
Embden, J.D., 1993. Comparison of various repetitive DNA ele- 2010. Evaluating microfossil content of dental calculus from
ments as genetic markers for strain differentiation and epidemiology Brazilian sambaquis. J. Archaeol. Sci. 37 (6), 1326 1338.
of Mycobacterium tuberculosis. J. Clin. Microbiol. 31 (8), Weyrich, L.S., Dobney, K., Cooper, A., 2015. Ancient DNA analysis of
1987 1995. dental calculus. J. Hum. Evol. 79, 119 124.
Velsko, I.M., Overmyer, K.A., Speller, C., Klaus, L., Collins, M.J., Loe, Weyrich, L.S., Duchene, S., Soubrier, J., Arriola, L., Llamas, B., Breen,
L., et al., 2017. The dental calculus metabolome in modern and his- J., et al., 2017. Reconstructing Neandertal behavior diet, and disease
toric samples. Metabolomics 13 (11), 134. using ancient DNA from dental calculus. Nature 544, 357 361.
Verrelli, B.C., McDonald, J.H., Argyropoulos, G., Destro-Bisol, G., Willerslev, E., Hansen, A.J., Poinar, H.N., 2004. Isolation of nucleic acids and
Froment, A., Drousiotou, A., et al., 2002. Evidence for balancing cultures from fossil ice and permafrost. Trends Ecol. Evol. 19 (3), 141 147.
selection from nucleotide sequence analyses of human G6PD. Am. Woese, C.R., Fox, G.E., 1977. Phylogenetic structure of the prokaryotic
J. Hum. Genet. 71 (5), 1112 1128. domain: the primary kingdoms. Proc. Natl. Acad. Sci. U.S.A. 74
Vigano, C., Haas, C., Ruhli, F.J., Bouwman, A., 2017. 2,000 Year (11), 5088 5090.
old beta-thalassemia case in Sardinia suggests malaria was Worobey, M., Gemmel, M., Teuwen, D.E., Haselkorn, T., Kunstman, K.,
endemic by the Roman period. Am. J. Phys. Anthropol. 164, Bunce, M., et al., 2008. Direct evidence of extensive diversity of
362 370. HIV-1 in Kinshasa by 1960. Nature 455 (7213), 661 664.
von Bubnoff, A., 2005. The 1918 flu virus is resurrected. Nature 437, Xiao, Y.L., Kash, J.C., Beres, S.B., Sheng, Z.M., Musser, J.M.,
794 795. Taubenberger, J.K., 2013. High-throughput RNA sequencing of a
von Hunnius, T.E., Yang, D., Eng, B., Waye, J.S., Saunders, S.R., 2007. formalin-fixed, paraffin-embedded autopsy lung tissue sample from
Digging deeper into the limits of ancient DNA research on syphilils. the 1918 influenza pandemic. J. Pathol. 229 (4), 535 545.
J. Archaeol. Sci. 34, 2091 2100. Yang, D.Y., Eng, B., Waye, J.S., Dudar, J.C., Saunders, S.R., 1998.
Wagner, D.M., Klunk, J., Harbeck, M., Devault, A., Waglechner, N., Improved DNA extraction from ancient bones using silica-based
Sahl, J.W., et al., 2014. Yersinia pestis and the plague of Justinian spin columns. Am. J. Phys. Anthropol. 105 (4), 539 543.
541-543 AD: a genomic analysis. Lancet Infect. Dis. 14 (4), Yatsunenko, T., Rey, F.E., Manary, M.J., Trehan, I., Dominguez-Bello,
319 326. M.G., Contreras, M., et al., 2012. Human gut microbiome viewed
Warinner, C., 2016. Dental calculus and the evolution of the human oral across age and geography. Nature 486 (7402), 222 227.
microbiome. J. Calif. Dent. Assoc. 44 (7), 411 420. Ziesemer, K.A., Mann, A.E., Sankaranarayanan, K., Schroeder, H.,
Warinner, C., Hendy, J., Speller, C., Cappellini, E., Fischer, R., Ozga, A.T., Brandt, B.W., et al., 2015. Intrinsic challenges in
Trachsel, C., et al., 2014a. Direct evidence of milk consumption ancient microbiome reconstruction using 16S rRNA gene amplifica-
from ancient human dental calculus. Sci. Rep. 4, 7104. tion. Sci. Rep. 5, 16498.
Warinner, C., Rodrigues, J.F., Vyas, R., Trachsel, C., Shved, N., Zink, A., Haas, C.J., Reischl, U., Szeimies, U., Nerlich, A.G., 2001.
Grossmann, J., et al., 2014b. Pathogens and host immunity in the Molecular analysis of skeletal tuberculosis in an ancient Egyptian
ancient human oral cavity. Nat. Genet. 46 (4), 336 344. population. J. Med. Microbiol. 50 (4), 355 366.
Warinner, C., Speller, C., Collins, M.J., 2015. A new era in palaeomicro- Zink, A.R., Reischl, U., Wolf, H., Nerlich, A.G., 2002. Molecular analy-
biology: prospects for ancient dental calculus as a long-term record sis of ancient microbial infections. FEMS Microbiol. Lett. 213 (2),
of the human oral microbiome. Philos. Trans. R. Soc. Lond. B Biol. 141 147.
Sci. 370 (1660), 20130376. Zink, A.R., Sola, C., Reischl, U., Grabner, W., Rastogi, N., Wolf, H.,
Watson, C.L., Lockwood, D.N., 2009. Single nucleotide polymorphism et al., 2003. Characterization of Mycobacterium tuberculosis com-
analysis of European archaeological M. leprae DNA. PLoS One 4 plex DNAs from Egyptian mummies by spoligotyping. J. Clin.
(10), e7547. Microbiol. 41 (1), 359 367.

You might also like