Abstract
Most spinal cord injury (SCI) research programs focus only on the injured spinal cord with the goal of restoring locomotor function by overcoming mechanisms of cell death or axon regeneration failure. Given the importance of the spinal cord as a locomotor control center and the public perception that paralysis is the defining feature of SCI, this “spinal-centric” focus is logical. Unfortunately, such a focus likely will not yield new discoveries that reverse other devastating consequences of SCI including cardiovascular and metabolic disease, bladder/bowel dysfunction and infection. The current review considers how SCI changes the physiological interplay between the spinal cord, the gut and the immune system. A suspected culprit in causing many of the pathological manifestations of impaired spinal cord- gut-immune axis homeostasis is the gut microbiota. After SCI, the composition of the gut microbiota changes, creating a chronic state of gut “dysbiosis”. To date, much of what we know about gut dysbiosis was learned from 16S-based taxonomic profiling studies that reveal changes in the composition and abundance of various bacteria. However, this approach has limitations and creates taxonomic “blindspots”. Notably, only bacteria can be analyzed. Thus, in this review we also discuss how the application of emerging sequencing technologies can improve our understanding of how the broader ecosystem in the gut is affected by SCI. Specifically, metagenomics will provide researchers with a more comprehensive look at post-injury changes in the gut virome (and mycome). Metagenomics also allows changes in microbe population dynamics to be linked to specific microbial functions that can affect the development and progression of metabolic disease, immune dysfunction and affective disorders after SCI. As these new tools become more readily available and used across the research community, the development of an “ecogenomic” toolbox will facilitate an Eco-Systems Biology approach to study the complex interplay along the spinal cord-gut-immune axis after SCI.
1. Introduction
Traumatic spinal cord injury (SCI) affects approximately 1.4 million people living in the United States. Loss of motor and sensory function are visible consequences of SCI. Less appreciated, although no less important or debilitating, are the “hidden” pathologies that manifest after SCI due to permanent dysfunction of the autonomic nervous system (ANS).
Many autonomic neurons, including all that comprise the sympathetic branch, are found in the spinal cord. When the spinal cord is injured, especially at higher spinal levels (above mid-thoracic spinal cord), axons that normally descend from brain/brainstem regions to control spinal sympathetic neurons are lost or damaged. The subsequent loss of normal sympathetic tone throughout the body leads to the development of some of the most devastating consequences of SCI including cardiovascular disease, bladder/bowel dysfunction and immune dysfunction. In this review, we highlight key changes in the spinal cord-gut-immune axis that converge to cause various co-morbidities and neurological complications after SCI (Fig. 1).
Fig. 1.
Spinal cord injury (SCI) sets in a motion a systematic breakdown of communication between the nervous system, the immune system and the gastrointestinal system (1). As a result of SCI, normal sympathetic control over all body systems is lost, creating an imbalance in autonomic tone in most organs. Consequently, physiological control of hematopoiesis (bone marrow and spleen) and immune surveillance (controlled by bone marrow, spleen, lymph nodes, gastrointestinal- associated lymphoid tissue/GALT) (2) as well as gastrointestinal function (e.g., motility, transit, mucin production) and liver function are lost. This break in homeostasis causes chronic immune dysfunction and gut dysbiosis, i.e., a lasting change in the ecological balance of microorganisms found in the biofilm and gut lumen (3). The metabolites and cytokines produced by microbes and cells in the dysbiotic gut enter the circulation or activate the vagus nerve, creating a feedback loop by which functional changes in the gut-immune axis affect structure and function within the CNS (brain, brainstem and spinal cord) (4).
1.1. The spinal cord-gut axis and gut dysbiosis after spinal cord injury
The enteric nervous system of the gastrointestinal (GI) tract is innervated by the parasympathetic vagus nerve and sympathetic spinal nerves originating in the brainstem and spinal cord, respectively. After a SCI, an imbalance in autonomic tone develops because sympathetic control of the small bowel and colon is lost, causing acute and protracted GI dysfunction marked by impairments in gut motility, mucosal secretions, vascular tone and immune function. These physiological complications of a neurogenic bowel contribute to the constipation, bloating, nausea, impaired transit, incontinence and abdominal pain experienced by SCI individuals (Tate et al., 2016).
Spinal sympathetic nerves also innervate the gastrointestinal-associated lymphoid tissue (GALT), the immune system distributed throughout the GI tract. The GALT protects the body from microbes that attempt to invade the body through the mucosal surface of the GI tract (Hooper et al., 2012; Nicholson et al., 2012; Round and Mazmanian, (2009). This protective function of the GALT is remarkably specific and effective, especially if one considers that the mucosal surface of the gut is also home to trillions of microorganisms, i.e., the gut microbiota (Donaldson et al., 2016; Gu et al., 2013). The gut microbiota and their collective genomes (i.e., the microbiome), are critical for maintaining homeostasis both in the gut and throughout the body. In addition to exerting direct control over the GALT, the sympathetic nervous system also exerts indirect effects on the enteric microbiota through regulation of gut motility, mucosal secretions (e.g., mucin, acids) and epithelial permeability. In a neurogenic bowel, i.e., a functionally impaired bowel due to nerve injury, impaired intestinal transit limits the delivery of important nutrients to microbiota in the distal colon. Altered mucin production impairs production of the mucus layer, an important niche that is colonized by enteric microbiota creating a “biofilm”. Because of the distinct location of the biofilm relative to microorganisms in the gut lumen, these discrete ecological domains may be differentially affected by SCI (Fig. 1).
Gut microbiota contribute to host metabolism by breaking down complex molecules in food. During this process, commensal bacteria release metabolites that influence cellular function within and beyond the gut. In humans, correlations exist between metabolites in the blood and specific types of gut microbiota (Zhang et al., 2018). The range of cells that are known to be affected by gut-derived metabolites is extensive and includes intestinal epithelia, primary afferent neurons in the gut and immune cells in the GALT. These metabolites also diffuse into the circulation to influence cells in the liver, peripheral immune organs (e.g., bone marrow, spleen, lymph nodes) and the central nervous system (Clarke et al., 2014; Forsythe et al., 2014; Perez-Burgos et al., 2015; Tillisch, 2014; Wikoff et al., 2009; Yano et al., 2015). Changes in gut permeability brought on by chronic stress or trauma, can liberate commensal bacteria from the gut lumen, allowing the microbes themselves, not just the metabolites they produce, to enter the circulation. This increases the likelihood that gut-derived microbes will colonize and elicit inflammation in previously “sterile” tissues throughout the body.
In pre-clinical SCI models, intestinal epithelial cell permeability is increased and is associated with enhanced bacterial translocation and colonization of various organs including the lung (Kigerl et al., 2016). Whether bacteria or other gut-derived microbes can access the injured spinal cord is not known; however, it is possible since the permeability of the blood-spinal cord barrier and intestinal barrier increase at the same time after SCI (Noble and Wrathall, 1989; Popovich et al., 1996; Whetstone et al., 2003). Gut microbes and their metabolites may also influence the barrier functions of spinal cord endothelia, just as they do in the brain (Braniste et al., 2014).
Given the robust effects that the microbiota can have on host physiology, the changes that SCI causes in the ecological balance of microorganisms in the gut, i.e., gut dysbiosis, could broadly influence neurological function, overall health and quality of life. After SCI, gut dysbiosis has been documented in multiple clinical and preclinical studies (Gungor et al., 2016; Kigerl et al., 2016; Myers et al., 2019; O’Connor et al., 2018; Zhang et al., 2018). In most cases, 16s rRNA sequencing has been used to reveal the composition and relative abundance of bacteria in fecal samples or gut tissue samples after SCI. In mice, SCI causes robust and lasting gut dysbiosis and is characterized by inverse changes in the relative abundance of Bacteroidales and Clostridiales, the two most prevalent bacterial taxa in mouse gut: Bacteroidales decreases while Clostridiales increases after SCI (Eckburg et al., 2005; Kigerl et al., 2016; Krych et al., 2013). Minor taxa (Anaeroplasmatales, Turicibacterales and Lactobacillales) also are affected (Kigerl et al., 2016). The relative abundance of bacteria from the phylum Proteobacteria also increases in SCI mice (Myers et al., 2019). Chronic gut dysbiosis has also been described in rats after SCI, lasting at least 8 weeks post-injury (O’Connor et al., 2018). In humans, the abundance of butyrate-producing gut bacteria is reduced for at least 1 year after SCI (Gungor et al., 2016).
Much less appreciated than gut dysbiosis is the post-injury onset of urinary dysbiosis. In addition to developing a neurogenic bowel, SCI patients (and animals) also develop neurogenic bladders and the duration of neurogenic bladder (along with method and frequency of catheterization) can adversely affect the urinary microbiome. The pathophysiological effects of urinary dysbiosis are not known but there is diagnostic value in performing 16s-profiling of the urinary microbiome. Clinical data indicate that at baseline the bacterial composition of urine from healthy men and women is different and that after SCI, compositional changes in bacteria can differentiate between asymptomatic bacteriuria and urinary tract infection (UTI). (Kreydin et al., 2018; Evans et al., 2011; Morton et al., 2002; Skelton et al., 2019).
In addition to sex, other covariates will likely affect the magnitude, duration and pathophysiological impact of SCI-induced gut dysbiosis. For example, age affects experimental and clinical outcomes after SCI. Since gut microbial composition is affected by age and the average age at time of injury has steadily increased since the 1970s (Level, 2018), age should, when possible, be included as a covariate in all pre-clinical and clinical SCI studies (Arumugam et al., 2013; Guo et al., 2014; Jašarević et al., 2016; Markle et al., 2013; Sheng et al., 2017; Stilling et al., 2015; Thevaranjan et al., 2017). The spinal level at which an injury occurs also is an important covariate with implications for gut dysbiosis. Injuries at higher spinal levels will cause greater imbalance in autonomic control over the GI tract than injuries affecting lower spinal levels (Holmes and Blanke, 2019). SPNs controlling the small and large intestines are located primarily in the intermediolateral cell column in thoracic segments T5–10 and T10-S4, respectively (Browning and Travagli, 2014; Levatte et al., 1998; Mabon et al., 1997). Therefore, although autonomic control of the gut will be adversely affected by SCI occurring at any spinal level, higher level injuries (above T5) will remove most or all bulbospinal control over spinal autonomic networks that innervate the gut. To date, there have been no controlled studies designed to evaluate level-dependent effects on gut microbiota; however, data in a recent clinical report indicated that cervical SCI caused changes in gut microbiota composition that were distinct from those found in individuals with a thoracic or lumbar SCI (Zhang et al., 2018).
Currently, the functional consequences of SCI-induced dysbiosis are unknown but significant effects can be inferred from experimental studies. For example, changes in the relative abundance of certain gut bacteria correlate with worse or better locomotor function and also with immune function (Kigerl et al., 2016). Also, changes in GALT immune cell composition occur coincident with increased or decreased production of cytokines (TNFα, IL-1β, TGF-β, IL-10) for up to one month post-SCI (Kigerl et al., 2016) A similar relationship exists in SCI rats; at 8 weeks post-SCI, increased production of inflammatory cytokines in the intestines correlates with the relative abundance of particular types of gut bacteria (O’Connor et al., 2018). Precisely how gut dysbiosis develops after SCI, whether it persists indefinitely, and how it affects structure/function in the CNS and other organ systems is unknown. But, if causal relationships could be proved, many comorbidities that affect individuals with SCI might be treatable by targeting the gut microbiota or the metabolites that they produce. In this context it is encouraging that readily available therapeutics that influence the gut microbiota, including Lactobacillus-rich VSL#3 probiotics and melatonin, can restore gut microbiota homeostasis and improve locomotor recovery (Jing et al., 2019; Kigerl et al., 2016). In the paragraphs that follow, several systemic comorbidities that may contribute to or exacerbate SCI-induced gut dysbiosis are discussed.
1.2. Immune dysfunction
SCI disrupts normal sympathetic nervous system control of all major immune organs (Brommer et al., 2016; Lucin et al., 2007; Zhang et al., 2013). This causes SCI-induced immune depression syndrome (SCI- IDS), a profound and lasting deficiency in the immune system’s ability to fight infection. Indeed, SCI patients are at increased risk to develop infections and are 37x more likely to die of pneumonia than able-bodied individuals (DeVivo et al., 1993); the development of pneumonia and wound infections after SCI are associated with worse neurological recovery (Failli et al., 2012; Kopp et al., 2017). The higher incidence of infection after SCI may explain why survival rates have not improved for SCI patients over the past 30 years (Shavelle et al., 2015). What is needed to improve clinical care for SCI individuals is a better understanding of why infection develops more readily in this at-risk population. A likely culprit is a dysfunctional immune system that in turn alters the microbial ecology of gut commensals (Fig. 1).
SCI-IDS develops and persists indefinitely, in part, because maladaptive plasticity develops in the intraspinal circuitry located below the level of injury. This “new” circuitry, when activated by common recurring stimuli (e.g. bladder filling and other visceral/somatic input), triggers hyperactive neural-immune reflexes. As a result of excessive and uncontrolled reflex activity, leukocytes in peripheral immune organs are exposed to supra-physiological concentrations of hormones (e.g., glucocorticoids) and neurotransmitters (e.g., catecholamines) that elicit distinct intracellular signaling cascades that converge to kill immune cells (Lucin et al., 2007; Prüss et al., 2017; Ueno et al., 2016; Zhang et al., 2013). SCI-induced changes in autonomic tone to the GI tract may also cause or exacerbate SCI-IDS.
Sympathetic noradrenergic nerves innervate the vasculature and tissue parenchyma of the GALT, especially in regions of the GALT that are enriched with T and B lymphocytes (Straub et al., 2006). Generally, norepinephrine released by sympathetic post-ganglionic nerve terminals binds to beta-adrenergic receptors on innate and adaptive immune cells leading to suppression of their antimicrobial functions (Elenkov et al., 2000). Impaired sympathetic signaling may also alter the functional specialization of recently discovered macrophage networks found in the lamina propria and muscular layers of the intestine (Gabanyi et al., 2016). Overall, a break in immune homeostasis in GALT, caused by SCI-induced changes in sympathetic tone, will change how the mucosal immune system regulates the enteric microbiota.
After SCI, microbes that bypass the intestinal epithelial barrier could release peptides, metabolites and neurotransmitter-like molecules (e.g., quorum sensing molecules) that can directly activate vagal afferents located in the lamina propria. For those microbes that persist in the gut lumen, they also can elicit vago-vagal reflexes by signalling directly to enterochromaffin or enteroendocrine cells (EECs). EECs are sensor transducer cells that respond to nutrients, hormones, and microbe-derived factors produced in the gut lumen or the biofilm. In response to these stimuli, EECs release hormones, neuropeptides and neurotransmitters that activate neural networks via paracrine signaling. EECs also can directly activate sensory nerve terminals via neuropods, which are cytoplasmic extensions of the EECs that form functional synaptic contacts with sensory nerves in the lamina propria (Bohórquez et al., 2015; Liddle, 2019).
Gut microbiota also influence systemic immunity through modulation of vago-vagal reflexes (Borovikova et al., 2000; van Westerloo, 2010) and monocyte egress from the bone marrow. Not surprisingly, gut microbes have been implicated as potent regulators of immune responses to respiratory infections and the development of atopic and autoimmune diseases (Hill et al., 2012; Ichinohe et al., 2011; Ochoa-Repáraz et al., 2010; Shi et al., 2011; Wu et al., 2010). Emerging data also implicate the gut microbiota in regulating CNS glial homeostasis and neuroinflammation. Indeed, metabolites (e.g., short-chain fatty acids, tryptophan metabolites) produced by gut microbiota affect the maturation and function of CNS resident microglia and cross-talk between microglia and astrocytes (Brown et al., 2019; Erny et al., 2015; Rothhammer et al., 2016, 2018).
SCI elicits a systemic autoimmune response, i.e., trauma-induced autoimmunity (TIA) (Ankeny et al., 2006, 2009; Arevalo-Martin et al., 2018; Davies et al., 2007; Hayes et al., 2002; Hergenroeder et al., 2016; Jones et al., 2002). TIA develops when cells of the immune system recognize and mount an immune response against “self” antigens including proteins, carbohydrates, lipids and nucleic acids (Ankeny et al.,2009). Since SCI causes profound immune suppression, the onset of TIA seems paradoxical. However, it may be useful to think of TIA as a continuum of a non-resolving inflammatory response, dominated by innate immune cells, that is set in motion by SCI (Schwab et al., 2014). TIA may also be a secondary consequence of gut dysbiosis and bacterial translocation from the gut. Bacteria within the gut, notably segmented filamentous bacteria, can activate Th17 cells in GALT and trigger systemic and CNS autoimmunity (Flannigan and Denning, 2018; Ivanov et al., 2009; Kriegel et al., 2011). Bacterial translocation has been implicated as a trigger for the onset of systemic and CNS autoimmune diseases (Lee et al., 2011; Manfredo Vieira et al., 2018).
1.3. Mental and cognitive health
People living with a SCI and without concurrent brain injury develop anxiety and major depressive disorder or depression-like symptoms at higher rates than able-bodied individuals. Although the relationship between SCI and reduced mental health has been recognized for decades (Cao et al., 2017; Frank et al., 1992; Krause et al., 2000; Shin et al., 2012), most have attributed changes in psychiatric and mental health to environmental stressors including sudden loss of independence, high health care costs and the unique physical challenges associated with living with a SCI. However, physiological changes also occur after SCI that can directly affect emotional and mental health. Magnetic resonance imaging of cortical volume in SCI patients has revealed changes in brain areas that are important for information processing and emotional affect (Hawasli et al., 2018; Nicotra et al., 2006). In preclinical SCI models, higher indices of depression, anxiety and cognitive impairment have been documented (Craig et al., 2017; Davidoff et al., 1992; Luedtke et al., 2014; Roth et al., 1989; Wu et al.,2010) indicating that mental and cognitive impairments after SCI are likely stereotypical consequences of SCI and are caused by factors other than environmental stressors.
The gut-brain axis is now recognized as a powerful physiological regulator of mood and mental health. When the gut is intentionally colonized with discrete types of bacteria, signaling networks in the brain can be activated that elicit anxiety-like behaviors, whereas anxiety-like behaviors are reduced in germ-free mice when compared to specific pathogen free control mice (Diaz Heijtz et al., 2011; Neufeld et al., 2011). Vagal afferent-microbe interactions in the gut are implicated in modulation of mental health. In rats, vagal afferents are stimulated by microbially-derived fatty acids and vagotomy makes mice resistant to microbially-induced anxiety-like behaviors (Bravo et al.,2011; Lal et al., 2001).
Gut dysbiosis can also impair mental health. When the ecology of fecal samples obtained from people with major depressive disorder was analyzed, large population shifts were noted in the types of bacteria that produce neuroactive metabolites (Zheng et al., 2016). When these fecal samples were transplanted into germ-free mice, recipient mice developed anxiety-like and depressive-like behaviors (Zheng et al., 2016). In rats and mice, SCI also affects mood and increases anxiety-like behaviors (Craig et al., 2017; Davidoff et al., 1992; Luedtke et al., 2014; Roth et al., 1989; Wu et al., 2014). Recent data suggest that post-SCI dysbiosis may be the reason why these behaviors develop. Specifically, a fecal transplant prepared from healthy rats, when delivered orally to SCI rats, reverses SCI-induced dysbiosis and has potent anxiolytic effects (Fouad, K; personal communication). The molecular basis for this treatment effect is unknown, even though fecal transplants are now being considered as treatments for depression in people (Kurokawa et al., 2018).
1.4. Metabolic disease
Emerging data indicate that after SCI, loss of sympathetic tone and the development of aberrant spinal autonomic reflex control over immune organs (e.g., spleen) and other major organs that control metabolism (e.g., liver, adrenal gland, muscle, adipose tissue and gut) causes immune dysfunction and multi-organ pathology. Since immune and metabolic processes are normally tightly coupled and are essential for life (Hotamisligil, 2017a, 2017b), most, if not all, co-morbidities that affect SCI individuals (e.g., spontaneous infections in lung or skin, impaired wound healing, non-alcoholic fatty liver disease, chronic depression, atherosclerosis, type 2 diabetes, fatigue and anxiety), could be explained by impaired immunometabolism, which is caused or exacerbated by gut dysbiosis.
Metabolic function is significantly impaired in both paraplegics and tetraplegics; they have higher body fat content compared to able-bodied individuals (Gater, 2007; Gorgey et al., 2011, 2014). SCI individuals also have higher levels of intramuscular fat that contributes to insulin resistance and impaired glucose sensitivity (Boettcher et al., 2009; Elder et al., 2004). Changes in Bacteriodetes and Firmicutes, two of the largest populations of gut bacteria found in both mice and humans (Eckburg et al., 2005; Krych et al., 2013), could cause or contribute to chronic metabolic disturbances after SCI (Baothman et al., 2016; Ley, 2010; Tilg and Kaser, 2011; Turnbaugh and Gordon, 2009). 16s rRNA sequencing of fecal samples from SCI mice revealed that the relative abundance of Bacteroidales decreased as a function of time post-injury with a corresponding increase in Clostridiales, a class of Firmicutes (Kigerl et al., 2016). In obese able-bodied individuals and rodents, a similar reduction in the Bacteroidetes:Firmicutes ratio occurs and the associated metabolic dysfunction can be transmitted to naïve rats by colonizing their gut with fecal suspensions from obese animals or humans (Ley et al., 2006; Turnbaugh et al., 2006, 2008).
Signs of liver disease (e.g., nonalcoholic steatohepatitis), marked by an increase in liver adiposity and hepatic inflammation, also develop soon after SCI (Goodus et al., 2018; Sauerbeck et al., 2015). In SCI humans, liver adiposity is a prognostic indicator of metabolic disease (Rankin et al., 2017) and is associated with altered gut microbiota and increased bacterial translocation (Abu-Shanab and Quigley, 2010; Dumas et al., 2006). Similar to the GALT, the liver acts as a firewall between the gut and the body, filtering microbes that drain from the intestine into the hepatic portal vein (Balmer et al., 2014; Jenne and Kubes, 2013). In SCI animals and humans, an inflamed liver can limit hepatic filtration capacity, allowing gut microbes to pass through the liver and elicit systemic inflammation. These same microbes may also elicit or propagate liver inflammation.
1.5. Emerging topics and opportunities
Today, there is a growing appreciation in the field of SCI research and clinical care, that the gut microbiota represents an important but poorly understood biological variable that is capable of significantly affecting the health, recovery and well-being of SCI individuals. As a result, a growing number of pre-clinical and human subject research projects have been completed to better understand how SCI affects gut (and urine - see above) ecology. Below, we discuss technical aspects of gut microbiome research and emphasize the importance of understanding the limitations of commonly used techniques. We also introduce the gut virome as a novel and as yet understudied component of the gut microbiota, especially in the context of SCI.
Technical advances in microbiome science.
Recent technological advances have transformed the study of microbes throughout the life sciences. Early descriptions of the taxonomic profile, or enterotypes, of bacteria in the large intestine of mice were done using new (at the time) anaerobic culturing methods of fecal samples (Schaedler et al., 1965), where early studies characterized the relationship between mammalian gut commensal bacteria and development of the mammalian lymphatics (Bauer et al., 1963). With time, DNA-sequencing techniques improved and RNA experts transformed the study of microbial diversity. Now, rather than use culture-dependent techniques, they used gene markers (e.g., 16s rDNA) to survey naturally-occurring microbial communities (Lane et al., 1985; Woese and Fox, 1977). Over time, this evolution in technique revealed that culture-based methods missed most of the microbes that could be detected using molecular markers, often with far less than 1% of microbes being captured with culture-based techniques (Rappé and Giovannoni, 2003). Subsequently, large-scale screening of fecal and plasma samples from mammalian models, including humans and mice, provided comprehensive “who is there?” (compositional) databases using such molecular surveys rather than the less comprehensive in vitro culture-based methods. To date, 16S-based taxonomic profiling has enabled community-wide microbial surveys across diverse environments with unprecedented temporal and spatial scales. These emerging databases have transformed our understanding of such ecosystems, necessitating an eco-systems biology perspective to best understand the role of microbes in complex communities. For example, 16S studies in the human gut now implicate specific bacteria in determining whether you are obese or lean (Turnbaugh et al., 2008, 2006) or whether you will be susceptible to inflammatory bowel disease (Imhann et al., 2018). The same tools have also revealed associations between the gut microbiota and other ‘ecosystems’ found in humans such as the oral cavity (Olsen and Yamazaki, 2019; Segata et al., 2012).
As sequencing costs have decreased, gene-targeted sequencing is rapidly being overtaken by metagenomic sequencing (i.e., shotgun sequencing or community genomic sequencing). In this approach, whole communities of organisms - including bacteria, archaea, fungi and their viruses - can be surveyed simultaneously. The 16S-based approach is commonly biased against archaea, and misses entirely fungi and viruses, as they lack this gene marker. Fungi can be targeted using 18S primer sets (Banos et al., 2018) but there are no universal gene markers to capture viruses (Sullivan, 2015). In many ecosystems, fungi remain virtually unstudied, but viruses are now credited with drastically impacting microbial communities through lysis, gene transfer and metabolic reprogramming during infection (Brum et al., 2015). Thus, to gain a fuller ecosystem perspective, it is critical to advance beyond 16S-based surveys so as to avoid known taxonomic blind spots in bacteria and archaea, while also capturing other entities such as viruses or fungi.
Fortunately, concomitant with the rise of new sequencing technologies that have enabled metagenomic data generation, legions of researchers have developed analytics and community-available tools to interpret these large-scale datasets (Fig. 2; Table 1). Beyond expanding our abilities to answer “who is there?” to non-bacteria, metagenomics also enables functional analyses to help answer “what are they doing, and with whom?”. Such advances are gaining recent attention in human microbiome studies (e.g., (Quince et al., 2017)), but they have long been transforming our understanding of the oceans (DeLong et al., 2006) and other complex systems (Tringe et al., 2005; Tyson et al., 2004). For the oceans in particular, global scale surveys have now been conducted for viruses (Brum et al., 2015; Gregory et al., 2019b; Roux et al., 2016), prokaryotes (Sunagawa et al., 2015) and microbial eukaryotes(de Vargas et al., 2015). Such surveys have become the basis for analytics that have revealed global “interactomes” (the hypothesized interactions between hundreds of thousands of organisms)(Lima-Mendez et al., 2015), as well as genes-to-ecosystems based modeling approaches that take such data and identify those that best predict a feature of interest (Guidi et al., 2016). In oceans, this feature of interest was carbon flux and surprisingly identified viruses as the best predictor (Guidi et al., 2016). Still, the scalable analytical approach is generalizable to any large-scale dataset, such that it can be used to identify which tens of thousands of different microbes best predict disease. Given the importance of microbes and their viruses to ‘ecosystem properties’ in so many other environments, it is now well-accepted that anywhere microbes are abundant they are likely major players. Here, we will briefly consider what is known about the human gut virome in health and various disease states, as well as the potential role for the human gut virome in SCI.
Fig. 2.
The metagenomic pipeline. Raw sequencing reads are provided by the sequencing center. The first step is to quality control (QC) the reads (Bolger et al., 2014; Bushnell, 2019; Joshi and Fass, 2011), which involves removal of reads that map to the model organism from which samples were collected, removal of adaptors and common sequencing center contaminants, and removal of low-quality reads. The final result of this step is a set of clean reads. Next, the clean reads are assembled to form long contigs (Kulikov et al., 2012; Li et al., 2015; Peng et al., 2012). Ideally, these contigs correspond to all or part of the genomes of the microbial taxa (bacteria and viruses) within the sample. In the microbial identification step, bacteria can be grouped (called binning) by operational taxonomic unit (OTU) with a variety of tools (Alneberg et al., 2014; Dick et al., 2009; Graham et al., 2017; Imelfort et al., 2014; Kang et al., 2015; Lu et al., 2016; Parks, 2017; Sieber et al., 2018; Uritskiy et al., 2018; Wu et al., 2016)(ggkbase.berkeley.edu). Viral OTUs (vOTUs) are similarly identified with a variety of tools (.Akhter et al., n.d; Amgarten et al., 2018; Arndt et al., 2016; Ren et al., 2018; Roux et al., 2015; Vik et al., 2017; Zheng et al., 2019). At this point, a user possesses two sets of contigs corresponding to complete or near-complete genomes of the viruses and bacteria present in the original sample. Here, analysis can diverge. Common analyses include read mapping (Bushnell, 2019; Langmead and Salzberg, 2012; Li and Durbin, 2009; Rabosky, 2014; Woodcroft, 2019) to calculate abundances, taxonomy classification (Doulcier et al., 2017; Parks et al., 2018), and functional annotation (identification of genes present within contigs) (Finn et al., 2014; Huerta-Cepas et al., 2018; Kanehisa et al., 2015; Tatusov et al., 2000). For phages (viruses that infect bacteria), host prediction (Altschup et al., 1990; Bland et al., 2007; Galiez et al., 2017; Laslett and Canback, 2004; Lowe and Eddy, 1997; Villarroel et al., 2016) to identify the bacterial host is a common downstream analysis. For bacteria, pathway analysis (Boyd, 2019; Caspi et al., 2012) may be of interest.
Table 1.
Tools for Metagenomic Analysis. Lists the tool on the left, with the title of associated paper on the right. If no published paper is available, a short description is provided with corresponding code repository as requested by the tool’s creator.
| ABAWACA | A tool for binning metagenomic contigs. Available at: https://github.com/CK7/abawaca |
| ARAGORN | ARAGORN, a program to detect tRNA genes and tmRNA genes in nucleotide sequences |
| BamM | Automatic Detection of Key Innovations, Rate Shifts, and Diversity-Dependence on Phylogenetic Trees |
| BBTools | BBTools is a suite of fast, multithreaded bioinformatics tools designed for analysis of DNA and RNA sequence data, including bbduk, which filters and trims reads for adapters and contaminants using k-mers and bbmap, a short-read aligner for DNA and RNA-seq data. Available at: https://sourceforge.net/projects/bbmap/ |
| BinSanity | BinSanity: unsupervised clustering of environmental microbial assemblies using coverage and affinity propagation. |
| BLAST/prophage blast | Basic Local Alignment Search Tool |
| Bowtie2 | Fast gapped-read alignment with Bowtie 2 |
| BWA | Fast and accurate short read alignment with Burrows-Wheeler transform |
| CD-HIT | Cd-hit: a fast program for clustering and comparing large sets of protein or nucleotide sequences |
| CheckM | CheckM: assessing the quality of microbial genomes recovered from isolates, single cells, and metagenomes |
| ClusterGenomes | A tool to remove redundancies from contigs or genomes by clustering them, using the longest contig as the representative for the population. Available at: https://bitbucket.org/MAVERICLab/docker-clustergenomes |
| COCACOLA | COCACOLA: binning metagenomic contigs using sequence COmposition, read CoverAge, CO-alignment and paired-end read LinkAge |
| COG | The COG database: a tool for genome-scale analysis of protein functions and evolution |
| CONCOCT | Binning metagenomic contigs by coverage and composition |
| CoverM | A configurable, easy to use and fast DNA read coverage and relative abundance calculator focused on metagenomics applications. Available at: https://github.com/wwood/CoverM/ |
| CRT/MinCED | CRISPR recognition tool (CRT): a tool for automatic detection of clustered regularly interspaced palindromic repeats. |
| DAS Tool | Recovery of genomes from metagenomes via a dereplication, aggregation and scoring strategy |
| DeepVirFinder | Identifying viruses from metagenomic data by deep learning |
| dRep | dRep: a tool for fast and accurate genomic comparisons that enables improved genome recovery from metagenomes through de-replication |
| eggNOG | eggNOG 5.0: a hierarchical, functionally and phylogenetically annotated orthology resource based on 5090 organisms and 2502 viruses |
| EnrichM | A tool for MAG annotation and de novo MAG identification, as well as metabolic pathway analysis and functional analysis. Available at: https://github.com/geronimp/enrichM |
| ggKbase | A suite of tools, including a popular binning tool which uses phylogeny, coverage, and GC content to separate a set of contigs into bins. Available at: https://ggkbase.berkeley.edu/ |
| GroopM | GroopM: an automated tool for the recovery of population genomes from related metagenomes |
| GTDB | A standardized bacterial taxonomy based on genome phylogeny substantially revises the tree of life. |
| HostPhinder | HostPhinder: A Phage Host Prediction Tool |
| IDBA-UD | IDBA-UD: a de novo assembler for single-cell and metagenomic sequencing data with highly uneven depth. |
| KEGG | KEGG as a reference resource for gene and protein annotation |
| MArVD | Putative archaeal viruses from the mesopelagic ocean |
| MARVEL | MARVEL, a Tool for Prediction of Bacteriophage Sequences in Metagenomic Bins |
| MaxBin | MaxBin 2.0: an automated binning algorithm to recover genomes from multiple metagenomic datasets. |
| Megahit | MEGAHIT: an ultra-fast single-node solution for large and complex metagenomics assembly via succinct de Bruijn graph |
| MetaBAT | MetaBAT, an efficient tool for accurately reconstructing single genomes from complex microbial communities |
| MetaCyc | The MetaCyc database of metabolic pathways and enzymes and the BioCyc collection of pathway/genome databases |
| MetaWRAP | MetaWRAP—a flexible pipeline for genome-resolved metagenomic data analysis |
| Pfam | Pfam: the protein families database |
| PHASTER | PHASTER: a better, faster version of the PHAST phage search tool |
| PhiSpy | PhiSpy: a novel algorithm for finding prophages in bacterial genomes that combines similarity- and composition-based strategies |
| Sickle | A sliding-window, adaptive, quality-based trimming tool for FastQ files. Available at: github.com/najoshi/sickle |
| SPAdes | SPAdes: A New Genome Assembly Algorithm and Its Applications to Single-Cell Sequencing |
| tetraESOM | Community-wide analysis of microbial genome sequence signatures |
| Trimmomatic | Trimmomatic: a flexible trimmer for Illumina sequence data |
| tRNA-scan-SE | tRNAscan-SE: a program for improved detection of transfer RNA genes in genomic sequence |
| UniteM | UniteM implements different ensemble binning strategies to produce a non-redundant set ofbins from the output of multiple binning methods. Available at: https://github.com/dparks1134/UniteM |
| VconTACT2 | vConTACT: an iVirus tool to classify double-stranded DNA viruses that infect Archaea and Bacteria |
| VirMiner | Mining, analyzing, and integrating viral signals from metagenomic data |
| VirSorter | VirSorter: mining viral signal from microbial genomic data |
| WIsH | WIsH: who is the host? Predicting prokaryotic hosts from metagenomic phage contigs |
The Virome.
In contrast to pathogenic eukaryotic viruses, the gut virome is dominated by viruses that infect bacteria (bacteriophages, or phages). Within the human gut, most known bacteriophages are in the order Caudovirales (containing the families myoviridae, podoviridae, and siphoviridae) or the floating family, microviridae. While there are highly conserved phages across individuals (Dutilh et al., 2014; Yutin et al., 2018), the gut virome is largely unique between individuals and within an individual and is largely stable over time (Manrique et al., 2017; Reyes et al., 2010). Further, it has been shown that the composition of the gut virome can be altered by development, the immune system, diet, cohabitation, pharmacologic factors and disease (Abeles et al., 2015; Ly et al., 2016; Manrique et al., 2017; Minot et al., 2011; Scarpellini et al., 2015). Bacteriophages, like eukaryotic viruses, reproduce using a lytic (causing death of host bacteria) or lysogenic (non-lytic) life cycle, and metagenomic sequence data suggest that most phages in the human gut are lysogenic (Minot et al., 2011; Monaco et al., 2016). While the dynamics of the human gut virome are not well characterized, a system dominated by lysogenic vs lytic phages allows persistence of individual bacteria over longer time spans. Additionally, lysogenic viruses can be activated by stimuli, such as changes in diet, to rapidly alter gut bacterial community structure (Duerkop et al., 2018; Minot et al., 2011).
The human gut virome has now been examined in a handful of diseases: colorectal cancer, HIV, inflammatory bowel disease, hypertension, obesity, type 1 diabetes, Clostridium difficile infection, and autism. Though this is a nascent field of study, new data are revealing the dynamics of the microbiome in response to disease, viral interactions with the host immune system, and the classificatory power of the enteric virome. In obesity, mouse models have demonstrated an expansion of Caudovirales phages (Kim and Bae, 2016). In HIV, enteric eukaryotic viruses, including adenovirus and anelloviruses, can be found in blood, suggesting immune dysfunction and translocation across a compromised gut epithelium. At the same time, abundance of these viruses in the gut increases, while bacterial diversity decreases (Handley et al., 2012; Monaco et al., 2016). In inflammatory bowel disease, patient gut viral composition was sufficient to distinguish patients with ulcerative colitis from those with Crohn’s disease. Further, as in HIV, it was observed that viral diversity increased as bacterial diversity decreased (Norman et al., 2015). While viral and bacterial diversity are calculated using different methods that are not directly comparable, it is unusual that they would move in opposite directions, raising the possibility that gut virome dynamics are not simply mirroring expansion or reduction in bacterial communities.
The role of phages is not limited to interactions with their host bacteria. In ulcerative colitis, there is evidence that phages directly interact with the immune system through stimulation of dendritic TLR9, inducing IFN-gamma production by CD4+ T-cells and exacerbating symptoms (Gogokhia et al., 2019). Phages also have high bio-marker potential: in colorectal cancer, differences in the gut virome and bacteriome could be used to distinguish human patients with cancer from healthy controls (Handley and Devkota, 2019; Hannigan et al., 2018). In hypertension, it has been shown that gut viruses have higher discriminatory power than gut bacteria in distinguishing between healthy, prehypertension and hypertension samples (Han et al., 2018). Likewise, in children with type 1 diabetes, changes in gut viral community structure were found to precede autoimmunity and changes in gut bacterial communities, suggesting that the gut virome is a sensitive predictor of disease risk (Kostic et al., 2015; Zhao et al., 2018). In fact, phages may be more sensitive than bacteria in predicting at-risk individuals (Moreno-Gallego et al., 2019) and also as biomarkers of disease and pathogenesis (Flores et al., 2013; Gregory et al., 2018). Finally, commensal phages may have therapeutic potential. In C. difficile infection, where fecal microbiota transplant (FMT) has been growing as an alternative to antibiotic therapy, it has been shown that bacteria are not necessary for therapeutic success, raising the possibility that bacteriophages, i.e., the virome that is transferred in FMT, is responsible for recovery from C. difficile infection (Ott et al., 2017). Indeed, successful bacteriophage transfer in FMT has been associated with treatment outcome in C. difficile (Zuo et al., 2018). In a small open-label study, sustained FMT treatment suggested that viruses and microbes from healthy donors could mitigate autism symptoms in children (Kang et al., 2017). To date, the human virome in SCI remains unexplored, but it is clear that phages are altered in diseases ranging from infectious to metabolic to inflammatory and beyond. One key challenge in interpreting such “human virome” studies is that non-quantitative sample preparations are commonly used, which minimizes the ecological interpretations that can be made from the data (Gregory et al., 2019).
In summary, there are numerous critical interactions between phages and their bacterial hosts: phage-host dynamics alter commensal bacterial communities and determine community assembly trajectory early in life (Lim et al., 2015), provide bacteria with additional survival and virulence genes (De Smet et al., 2016; Torres-Barceló, 2018), and reprogram their bacterial hosts in ways that alter metabolic outputs or confer protection from invasion by pathogenic bacteria at mucosal surfaces (Barr et al., 2019; Mirzaei and Maurice, 2017; Reyes et al., 2012). Like bacteria, gut phage populations change in disease, and sometimes these changes precede disease onset, highlighting the possibility that gut phage profiles could be used as biomarkers to predict disease risk. Like bacteria, phages can also interact with the immune system to promote inflammation or strengthen mucosal defenses and protect the host from disease (Kernbauer et al., 2014). Emerging data suggest that therapeutic phages can work in tandem with the host immune system to resolve bacterial infections in skin (Abedon et al.,2011) and the lung (Roach et al., 2017). Together, these findings suggest that phages likely play an integral role in the microbe-gut-brain axis in disease and should be considered as part of any Eco-Systems biology study.
2. Conclusions
Like so many other human diseases, research in SCI has for decades overlooked the possible impact of microbes and their viruses, largely due to technological limitations that prevented us from seeing these “hidden” movers and shakers in the human body. However, there are now numerous examples where microbes likely play supporting, if not causal, roles in disease pathogenesis. As new tools and approaches become more readily available to the broader research community, the “ecogenomic” toolbox described above will enable a more wholistic, Eco-Systems Biology approach to study SCI that should help unravel the complex interplay along the virus/microbe-gut-brain axis.
Acknowledgments
The authors would like to acknowledge Jingjie Du and Ahmed Zayed for their contributions to this work. Personnel and data referred to in this report are supported in part by the Belford Spinal Cord Injury Center (PGP), The Ray W. Poppleton endowment (PGP) and NINDS grant R35NS111582 (PGP).
References
- Abedon ST, Kuhl SJ, Blasdel BG, Kutter EM, 2011. Phage treatment of human infections. Bacteriophage 1, 66–85. 10.4161/bact.1.2.15845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Abeles SR, Ly M, Santiago-Rodriguez TM, Pride DT, 2015. Effects of long term antibiotic therapy on human oral and fecal viromes. PLoS One 10, e0134941. 10.1371/journal.pone.0134941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Abu-Shanab A, Quigley EM, 2010. The role of the gut microbiota in nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol 7, 691–701. 10.1038/nrgastro.2010.172. [DOI] [PubMed] [Google Scholar]
- Akhter S, Aziz RK, Edwards RA, n.d. PhiSpy: a novel algorithm for finding prophages in bacterial genomes that combines similarity-and composition-based strategies. doi: 10.1093/nar/gks406 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alneberg J, Bjarnason BS, de Bruijn I, Schirmer M, Quick J, Ijaz UZ, Lahti L, Loman NJ, Andersson AF, Quince C, 2014. Binning metagenomic contigs by coverage and composition. Nat Methods 11, 1144–1146. 10.1038/nmeth.3103. [DOI] [PubMed] [Google Scholar]
- Altschup SF, Gish W, Miller W, Myers EW, Lipman DJ, 1990. Basic local alignment search tool. J Mol Biol 215 (3), 403–410. [DOI] [PubMed] [Google Scholar]
- Amgarten D, Braga LPP, da Silva AM, Setubal JC, 2018. MARVEL, a tool for prediction of bacteriophage sequences in metagenomic bins. Front Genet 9, 1–8. 10.3389/fgene.2018.00304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ankeny DP, Lucin KM, Sanders VM, McGaughy VM, Popovich PG, 2006. Spinal cord injury triggers systemic autoimmunity: evidence for chronic B lymphocyte activation and lupus-like autoantibody synthesis. J Neurochem 99, 1073–1087. 10.1111/j.1471-4159.2006.04147.x. [DOI] [PubMed] [Google Scholar]
- Ankeny DP, Guan Z, Popovich PG, 2009. B cells produce pathogenic antibodies and impair recovery after spinal cord injury in mice. J Clin Invest 119, 2990–2999. 10.1172/JCI39780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arevalo-Martin A, Grassner L, Garcia-Ovejero D, Paniagua-Torija B, Barroso-Garcia G, Arandilla AG, Mach O, Turrero A, Vargas E, Alcobendas M, Rosell C, Alcaraz MA, Ceruelo S, Casado R, Talavera F, Palazón R, Sanchez-Blanco N, Maier D, Esclarin A, Molina-Holgado E, 2018. Elevated autoantibodies in sub-acute human spinal cord injury are naturally occurring antibodies. Front Immunol 9, 2365 10.3389/fimmu.2018.02365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arndt D, Grant JR, Marcu A, Sajed T, Pon A, Liang Y, Wishart DS, 2016PHASTER: a better, faster version of the PHAST phage search tool. Nucl Acids Res 44 10.1093/nar/gkw387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arumugam M, Raes J, Pelletier E, Le Paslier D, Batto J, Bertalan M, Borruel N, Casellas F, Costea PI, Hildebrand F, Manimozhiyan A, Bäckhed F, Blaser MJ, Bushman FD, De Vos WM, Ehrlich SD, Fraser CM, Hattori M, Huttenhower C, Jeffery IB, Knights D, Lewis JD, Ley RE, Ochman H, O’Toole PW, Quince C, Relman DA, Shanahan F, Sunagawa S, Wang J, Weinstock GM, Wu GD, Zeller G, Zhao L, Raes J, Knight R, Bork P, Gorvitovskaia A, Holmes SP, Huse SM, 2013. Enterotypes in the landscape of gut microbial community composition. Nature 3, 1–12. 10.1038/nature09944.Enterotypes. [DOI] [Google Scholar]
- Balmer ML, Slack E, de Gottardi A, Lawson MA, Hapfelmeier S, Miele L, Grieco A, Van Vlierberghe H, Fahrner R, Patuto N, Bernsmeier C, Ronchi F, Wyss M, Stroka D, Dickgreber N, Heim MH, McCoy KD, Macpherson AJ, 2014. The liver may act as a firewall mediating mutualism between the host and its gut commensal microbiota. Sci Transl Med 6, 237ra66. 10.1126/scitranslmed.3008618. [DOI] [PubMed] [Google Scholar]
- Banos S, Lentendu G, Kopf A, Wubet T, Glöckner FO, Reich M, 2018. A comprehensive fungi-specific 18S rRNA gene sequence primer toolkit suited for diverse research issues and sequencing platforms. BMC Microbiol 18, 190 10.1186/s12866-018-1331-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baothman OA, Zamzami MA, Taher I, Abubaker J, Abu-Farha M, 2016. The role of Gut Microbiota in the development of obesity and diabetes. Lipids Heal Dis 15, 108 10.1186/s12944-016-0278-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barr JJ, Auro R, Furlan M, Whiteson KL, Erb ML, Pogliano J, Stotland A, Wolkowicz R, Cutting AS, Doran KS, Salamon P, Youle M, Rohwer F, 2019. Bacteriophage adhering to mucus provide a non-host-derived immunity. 10.1073/pnas.1305923110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BAUER H, HOROWITZ RE, LEVENSON SM, POPPER H, 1963. The response of the lymphatic tissue to the microbial flora. Studies on germfree mice. Am J Pathol 42, 471–483. [PMC free article] [PubMed] [Google Scholar]
- Bland C, Ramsey TL, Sabree F, Lowe M, Brown K, Kyrpides NC, Hugenholtz P, 2007. CRISPR Recognition Tool (CRT): a tool for automatic detection of clustered regularly interspaced palindromic repeats. BMC Bioinformatics 8, 209 10.1186/1471-2105-8-209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boettcher M, Machann J, Stefan N, Thamer C, Häring HU, Claussen CD, Fritsche A, Schick F, 2009. Intermuscular adipose tissue (IMAT): association with other adipose tissue compartments and insulin sensitivity. J Magn Reson Imaging 29, 1340–1345. 10.1002/jmri.21754. [DOI] [PubMed] [Google Scholar]
- Bohórquez DV, Shahid RA, Erdmann A, Kreger AM, Wang Y, Calakos N, Wang F, Liddle RA, 2015. Neuroepithelial circuit formed by innervation of sensory enteroendocrine cells. J Clin Invest 125, 782–786. 10.1172/JCI78361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bolger AM, Lohse M, Usadel B, 2014. Trimmomatic: A flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120. 10.1093/bioinformatics/btu170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Borovikova LV, Ivanova S, Nardi D, Zhang M, Yang H, Ombrellino M, Tracey KJ, 2000. Role of vagus nerve signaling in CNI-1493-mediated suppression of acute inflammation. Aut Neurosci 85, 141–147. 10.1016/S1566-0702(00)00233-2. [DOI] [PubMed] [Google Scholar]
- Boyd JA, 2019. EnrichM [WWW Document]. URL. https://github.com/geronimp/enrichM.
- Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Toth M, Korecka A, Bakocevic N, Guan NL, Kundu P, Gulyas B, Halldin C, Hultenby K, Nilsson H, Hebert H, Volpe BT, Diamond B, Pettersson S, 2014. The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med 6, 263ra158. 10.1126/scitranslmed.3009759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM, Dinan TG, Bienenstock J, Cryan JF, 2011. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci USA 108, 16050–16055. 10.1073/pnas.1102999108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brommer B, Engel O, Kopp MA, Watzlawick R, Müller S, Prüss H, Chen Y, DeVivo MJ, Finkenstaedt FW, Dirnagl U, Liebscher T, Meisel A, Schwab JM,2016. Spinal cord injury-induced immune deficiency syndrome enhances infection susceptibility dependent on lesion level. Brain 139, 692–707. 10.1093/brain/awv375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brown DG, Soto R, Yandamuri S, Stone C, Dickey L, Gomes-Neto JC, Pastuzyn ED, Bell R, Petersen C, Buhrke K, Fujinami RS, O’Connell RM, Stephens WZ, Shepherd JD, Lane TE, Round JL, 2019. The microbiota protects from viral-induced neurologic damage through microglia-intrinsic TLR signaling. Elife 8 10.7554/elife.47117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Browning KN, Travagli RA, 2014. Central nervous system control of gastrointestinal motility and secretion and modulation of gastrointestinal functions. Compr Physiol 4, 1339–1368. 10.1002/cphy.c130055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brum JR, Ignacio-Espinoza JC, Roux S, Doulcier G, Acinas SG, Alberti A,Chaffron S, Cruaud C, de Vargas C, Gasol JM, Gorsky G, Gregory AC, Guidi L, Hingamp P, Iudicone D, Not F, Ogata H, Pesant S, Poulos BT, Schwenck SM, Speich S, Dimier C, Kandels-Lewis S, Picheral M, Searson S, Bork P, Bowler C, Sunagawa S, Wincker P, Karsenti E, Sullivan MB, Coordinators TO, 2015. Ocean plankton. Patterns and ecological drivers of ocean viral communities. Science (80) 348, 1261498. 10.1126/science.1261498. [DOI] [PubMed] [Google Scholar]
- Bushnell B, 2019. BBTools [WWW Document]. URL. https://sourceforge.net/projects/bbmap/.
- Cao Y, Li C, Gregory A, Charlifue S, Krause JS, 2017. Depressive symptomatology after spinal cord injury: A multi-center investigation of multiple racial-ethnic groups. J Spinal Cord Med 40, 85–92. 10.1080/10790268.2016.1244314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caspi R, Altman T, Dreher K, Fulcher CA, Subhraveti P, Keseler IM, Kothari A, Krummenacker M, Latendresse M, Mueller LA, Ong Q, Paley S, Pujar A, Shearer AG, Travers M, Weerasinghe D, Zhang P, Karp PD, 2012. The MetaCyc database of metabolic pathways and enzymes and the BioCyc collection of pathway/genome databases. Nucleic Acids Res 40, D742–D753. 10.1093/nar/gkr1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clarke G, Stilling RM, Kennedy PJ, Stanton C, Cryan JF, Dinan TG, 2014. Gut Microbiota: the neglected endocrine organ. Mol Endocrinol. 10.1210/me.2014-1108.me20141108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Craig A, Guest R, Tran Y, Middleton J, 2017. Cognitive impairment and mood states after spinal cord injury. J Neurotrauma 34, 1156–1163. 10.1089/neu.2016.4632. [DOI] [PubMed] [Google Scholar]
- Davidoff GN, Roth EJ, Richards JS, 1992. Cognitive deficits in spinal cord injury: epidemiology and outcome. Arch Phys Med Rehabil 73, 275–284. [PubMed] [Google Scholar]
- Davies AL, Hayes KC, Dekaban GA, 2007. Clinical correlates of elevated serum concentrations of cytokines and autoantibodies in patients with spinal cord injury. Arch Phys Med Rehabil 88, 1384–1393. 10.1016/j.apmr.2007.08.004. [DOI] [PubMed] [Google Scholar]
- De Smet J, Zimmermann M, Kogadeeva M, Ceyssens P-J, Vermaelen W, Blasdel B, Bin Jang H, Sauer U, Lavigne R, 2016. High coverage metabolomics analysis reveals phage-specific alterations to Pseudomonas aeruginosa physiology during infection. ISME J 10, 1823–1835. 10.1038/ismej.2016.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Vargas C, Audic S, Henry N, Decelle J, Mahé F, Logares R, Lara E, Berney C, Le Bescot N, Probert I, Carmichael M, Poulain J, Romac S, Colin S, Aury JM, Bittner L, Chaffron S, Dunthorn M, Engelen S, Flegontova O, Guidi L, Horák A, Jaillon O, Lima-Mendez G, Lukeš J, Malviya S, Morard R, Mulot M, Scalco E, Siano R, Vincent F, Zingone A, Dimier C, Picheral M, Searson S, Kandels-Lewis S, Acinas SG, Bork P, Bowler C, Gorsky G, Grimsley N, Hingamp P, Iudicone D, Not F, Ogata H, Pesant S, Raes J, Sieracki ME, Speich S, Stemmann L, Sunagawa S, Weissenbach J, Wincker P, Karsenti E, Coordinators TO, 2015. Ocean plankton. Eukaryotic plankton diversity in the sunlit ocean. Science (80-) 348, 1261605. 10.1126/science.1261605. [DOI] [PubMed] [Google Scholar]
- DeLong EF, Preston CM, Mincer T, Rich V, Hallam SJ, Frigaard NU, Martinez A, Sullivan MB, Edwards R, Brito BR, Chisholm SW, Karl DM, 2006. Community genomics among stratified microbial assemblages in the ocean’s interior. Science (80-) 311, 496–503. 10.1126/science.1120250. [DOI] [PubMed] [Google Scholar]
- DeVivo MJ, Black KJ, Stover SL, 1993. Causes of death during the first 12 years after spinal cord injury. Arch Phys Med Rehabil 74, 248–254. [PubMed] [Google Scholar]
- Diaz Heijtz R, Wang S, Anuar F, Qian Y, Björkholm B, Samuelsson A, Hibberd ML, Forssberg H, Pettersson S, 2011. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A 108, 3047–3052. 10.1073/pnas.1010529108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dick GJ, Andersson AF, Baker BJ, Simmons SL, Thomas BC, Pepper Yelton A, Banfield JF, 2009. Community-wide analysis of microbial genome sequence signatures. Genome Biol 10, 85 10.1186/gb-2009-10-8-r85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Donaldson GP, Lee SM, Mazmanian SK, 2016. Gut biogeography of the bacterial microbiota. Nat Rev Microbiol 14, 20–32. 10.1038/nrmicro3552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doulcier G, Bolduc B, Jang H. Bin, Sullivan MB, Roux S, You Z-Q, 2017. vConTACT: an iVirus tool to classify double-stranded DNA viruses that infect Archaea and Bacteria. PeerJ 5, e3243. 10.7717/peerj.3243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duerkop BA, Kleiner M, Paez-Espino D, Zhu W, Bushnell B, Hassell B, Winter SE, Kyrpides NC, Hooper LV, 2018. Murine colitis reveals a disease-associated bacteriophage community. Nat Microbiol 3, 1023–1031. 10.1038/s41564-018-0210-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dumas ME, Barton RH, Toye A, Cloarec O, Blancher C, Rothwell A, Fearnside J, Tatoud R, Blanc V, Lindon JC, Mitchell SC, Holmes E, McCarthy MI, Scott J, Gauguier D, Nicholson JK, 2006. Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice. Proc Natl Acad Sci USA 103, 12511–12516. 10.1073/pnas.0601056103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dutilh BE, Cassman N, McNair K, Sanchez SE, Silva GGZ, Boling L, Barr JJ, Speth DR, Seguritan V, Aziz RK, Felts B, Dinsdale EA, Mokili JL, Edwards RA, 2014. A highly abundant bacteriophage discovered in the unknown sequences of human faecal metagenomes. Nat Commun 5, 4498 10.1038/ncomms5498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, Gill SR, Nelson KE, Relman DA, 2005. Diversity of the human intestinal microbial flora. Science (80-) 308, 1635–1638. 10.1126/science.1110591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elder CP, Apple DF, Bickel CS, Meyer RA, Dudley GA, 2004. Intramuscular fat and glucose tolerance after spinal cord injury - A cross-sectional study. Spinal Cord 42, 711–716. 10.1038/sj.sc.3101652. [DOI] [PubMed] [Google Scholar]
- Elenkov IJ, Wilder RL, Chrousos GP, Vizi ES, 2000. The sympathetic nerve-an integrative interface between two supersystems: the brain and the immune system. Pharmacol Rev 52, 595–638. [PubMed] [Google Scholar]
- Erny D, de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, Keren-Shaul H, Mahlakoiv T, Jakobshagen K, Buch T, Schwierzeck V, Utermöhlen O, Chun E, Garrett WS, McCoy KD, Diefenbach A, Staeheli P, Stecher B, Amit I, Prinz M, 2015. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci 18, 965–977. 10.1038/nn.4030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Evans CT, Rogers TJ, Burns SP, Lopansri B, Weaver FM, 2011. Knowledge and use of antimicrobial stewardship resources by spinal cord injury providers. PM R 3, 619–623. 10.1016/j.pmrj.2011.03.021. [DOI] [PubMed] [Google Scholar]
- Failli V, Kopp MA, Gericke C, Martus P, Klingbeil S, Brommer B, Laginha I, Chen Y, DeVivo MJ, Dirnagl U, Schwab JM, 2012. Functional neurological recovery after spinal cord injury is impaired in patients with infections. Brain 135, 3238–3250. 10.1093/brain/aws267. [DOI] [PubMed] [Google Scholar]
- Finn RD, Bateman A, Clements J, Coggill P, Eberhardt RY, Eddy SR, Heger A, Hetherington K, Holm L, Mistry J, Sonnhammer ELL, Tate J, Punta M, 2014. Pfam: the protein families database. Nucleic Acids Res 42, D222–D230. 10.1093/nar/gkt1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flannigan KL, Denning TL, 2018. Segmented filamentous bacteria-induced immune responses: a balancing act between host protection and autoimmunity. Immunology 10.1111/imm.12950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flores SC, Campbell TB, Bassis CM, Beck JM, Weinstock GM, Schmidt TM, Twigg H, Curtis JL, Lynch SV, Ghedin E, Kleerup E, Schloss PD, Jablonski K, Sodergren E, Venkataraman A, Crothers K, Young VB, Morris A, Fontenot AP, Huang L, 2013. Comparison of the respiratory microbiome in healthy non-smokers and smokers. Am J Respir Crit Care Med 187, 1067–1075. 10.1164/rccm.201210-1913oc. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Forsythe P, Bienenstock J, Kunze WA, 2014. Vagal pathways for microbiome-brain-gut axis communication. Adv Exp Med Biol 817, 115–133. 10.1007/978-1-4939-0897-4_5. [DOI] [PubMed] [Google Scholar]
- Frank RG, Chaney JM, Clay DL, Shutty MS, Niels CB, Kay DR, Elliott TR, Grambling S, 1992. Dysphoria: A major symptom factor in persons with disability or chronic illness. Psychiatry Res 43, 231–241. 10.1016/0165-1781(92)90056-9. [DOI] [PubMed] [Google Scholar]
- Gabanyi I, Muller PA, Feighery L, Oliveira TY, Costa-Pinto FA, Mucida D, 2016. Neuro-immune interactions drive tissue programming in intestinal macrophages. Cell 164, 378–391. 10.1016/j.cell.2015.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Galiez C, Siebert M, Enault F, Vincent J, Sö Ding J, 2017. WIsH: who is the host? Predicting prokaryotic hosts from metagenomic phage contigs. Bioinformatics 33, 3113–3114. 10.1093/bioinformatics/btx383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gater DR, 2007. Obesity after spinal cord injury. Phys Med Rehabil Clin N Am 18 (333–51), vii 10.1016/j.pmr.2007.03.004. [DOI] [PubMed] [Google Scholar]
- Gogokhia L, Buhrke K, Bell R, Casjens SR, Longman RS, Round Correspondence JL, 2019. Expansion of bacteriophages is linked to aggravated intestinal inflammation and colitis. Cell Host Microbe 25, 285–299. 10.1016/j.chom.2019.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goodus MT, Sauerbeck AD, Popovich PG, Bruno RS, McTigue DM, 2018. Dietary green tea extract prior to spinal cord injury prevents hepatic iron overload but does not improve chronic hepatic and spinal cord pathology in rats. J Neurotrauma 35, 2872–2882. 10.1089/neu.2018.5771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gorgey AS, Mather KJ, Poarch HJ, Gater DR, 2011. Influence of motor complete spinal cord injury on visceral and subcutaneous adipose tissue measured by multiaxial magnetic resonance imaging. J Spinal Cord Med 34, 99–109. 10.1179/107902610X12911165975106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gorgey AS, Dolbow DR, Dolbow JD, Khalil RK, Castillo C, Gater DR, 2014. Effects of spinal cord injury on body composition and metabolic profile - part I. J Spinal Cord Med 37, 693–702. 10.1179/2045772314Y.0000000245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Graham ED, Heidelberg JF, Tully BJ, 2017. BinSanity: unsupervised clustering of environmental microbial assemblies using coverage and affinity propagation. PeerJ 5, e3035. 10.7717/peerj.3035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gregory AC, Sullivan MB, Segal LN, Keller BC, 2018. Smoking is associated with quantifiable differences in the human lung DNA virome and metabolome. Respir Res 19, 1–13. 10.1186/s12931-018-0878-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gregory AC, Zablocki O, Howell A, Bolduc B, Sullivan MB, 2019. The human gut virome database. bioRxiv 655910. 10.1101/655910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gregory AC, Zayed AA, Conceição-Neto N, Temperton B, Bolduc B, Alberti A, Ardyna M, Arkhipova K, Carmichael M, Cruaud C, Dimier C, Domínguez-Huerta G, Ferland J, Kandels S, Liu Y, Marec C, Pesant S, Picheral M, Pisarev S, Poulain J, Tremblay J, Vik D, Babin M, Bowler C, Culley AI, de Vargas C, Dutilh BE, Iudicone D, Karp-Boss L, Roux S, Sunagawa S, Wincker P, Sullivan MB, Coordinators TO, 2019b. Marine DNA viral macro- and microdiversity from pole to pole. Cell 177, 1109–1123.e14. 10.1016/j.cell.2019.03.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gu S, Chen D, Zhang JN, Lv X, Wang K, Duan LP, Nie Y, Wu XL, 2013. Bacterial community mapping of the mouse gastrointestinal tract. PLoS One 8, e74957. 10.1371/journal.pone.0074957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guidi L, Chaffron S, Bittner L, Eveillard D, Larhlimi A, Roux S, Darzi Y, Audic S, Berline L, Brum JR, Coelho LP, Espinoza JCI, Malviya S, Sunagawa S, Dimier C, Kandels-Lewis S, Picheral M, Poulain J, Searson S, Coordinators TOC, Stemmann L, Not F, Hingamp P, Speich S, Follows M, Karp-Boss L, Boss E, Ogata H, Pesant S, Weissenbach J, Wincker P, Acinas SG, Bork P, de Vargas C, Ludicone D, Sullivan MB, Raes J, Karsenti E, Bowler C, Gorsky G,2016. Plankton networks driving carbon export in the oligotrophic ocean. Nature 532, 465 10.1038/nature16942.5327600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gungor B, Adiguzel E, Gursel I, Yilmaz B, Gursel M, 2016. Intestinal microbiota in patients with spinal cord injury. PLoS One 11, 1–10. 10.1371/journal.pone.0145878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo L, Karpac J, Tran SL, Jasper H, 2014. PGRP-SC2 promotes gut immune homeostasis to limit commensal dysbiosis and extend lifespan. Cell 156, 109–122. 10.1016/j.cell.2013.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han M, Yang P, Zhong C, Ning K, 2018. The human gut virome in hypertension. Front Microbiol 9, 1–10. 10.3389/fmicb.2018.03150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Handley SA, Devkota S, 2019. Going Viral: A Novel Role for Bacteriophage in Colorectal Cancer. 10.1128/mBio.02248-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Handley SA, Thackray LB, Zhao G, Presti R, Miller AD, Droit L, Abbink P, Maxfield LF, Kambal A, Duan E, Stanley K, Kramer J, Macri SC, Permar SR, Schmitz JE, Mansfield K, Brenchley JM, Veazey RS, Stappenbeck TS, Wang D, Barouch DH, Virgin HW, 2012. Pathogenic simian immunodeficiency virus infection is associated with expansion of the enteric virome. Cell 151, 253–266. 10.1016/j.cell.2012.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hannigan GD, Duhaime MB, Ruffin MT, Koumpouras CC, Schloss PD, 2018. Diagnostic Potential and Interactive Dynamics of the Colorectal Cancer Virome. 10.1128/mBio. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hawasli AH, Rutlin J, Roland JL, Murphy RKJ, Song SK, Leuthardt EC,Shimony JS, Ray WZ, 2018. Spinal cord injury disrupts resting-state networks in the human brain. J Neurotrauma 35, 864–873. 10.1089/neu.2017.5212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hayes KC, Hull TCL, Delaney GA, Potter PJ, Sequeira KAJ, Campbell K, Popovich PG, 2002. Elevated serum titers of proinflammatory cytokines and CNS autoantibodies in patients with chronic spinal cord injury. J Neurotrauma 19, 753–761. 10.1089/08977150260139129. [DOI] [PubMed] [Google Scholar]
- Hergenroeder GW, Moore AN, Schmitt KM, Redell JB, Dash PK, 2016. Identification of autoantibodies to glial fibrillary acidic protein in spinal cord injury patients. Neuroreport 27, 90–93. 10.1097/WNR.0000000000000502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hill DA, Siracusa MC, Abt MC, Kim BS, Kobuley D, Kubo M, Kambayashi T, Larosa DF, Renner ED, Orange JS, Bushman FD, Artis D, 2012. Commensal bacteria-derived signals regulate basophil hematopoiesis and allergic inflammation. Nat Med 18, 538–546. 10.1038/nm.2657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holmes GM, Blanke EN, 2019. Gastrointestinal dysfunction after spinal cord injury. Exp Neurol. 10.1016/j.expneurol.2019.113009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hooper LV, Littman DR, Macpherson AJ, 2012. Interactions between the microbiota and the immune system. Science (80-) 336, 1268–1273. 10.1126/science.1223490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hotamisligil GS, 2017a. Inflammation, metaflammation and immunometabolic disorders. Nature 542, 177–185. 10.1038/nature21363. [DOI] [PubMed] [Google Scholar]
- Hotamisligil GS, 2017b. Foundations of immunometabolism and implications for metabolic health and disease. Immunity 47, 406–420. 10.1016/j.immuni.2017.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huerta-Cepas J, Szklarczyk D, Heller D, Hernández-Plaza A, Forslund SK, Cook H, Mende DR, Letunic I, Rattei T, Jensen LJ, Von Mering C, Bork P, 2018. eggNOG 5.0: a hierarchical, functionally and phylogenetically annotated orthology resource based on 5090 organisms and 2502 viruses. Nucleic Acids Res 47, 309–314. 10.1093/nar/gky1085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ichinohe T, Pang IK, Kumamoto Y, Peaper DR, Ho JH, Murray TS, Iwasaki A,2011. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc Natl Acad Sci U S A 108, 5354–5359. 10.1073/pnas.1019378108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Imelfort M, Parks D, Woodcroft BJ, Dennis P, Hugenholtz P, Tyson GW, 2014. GroopM: an automated tool for the recovery of population genomes from related metagenomes. PeerJ 2, e603. 10.7717/peerj.603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Imhann F, Vich Vila A, Bonder MJ, Fu J, Gevers D, Visschedijk MC, Spekhorst LM, Alberts R, Franke L, van Dullemen HM, Ter Steege RWF, Huttenhower C, Dijkstra G, Xavier RJ, Festen EAM, Wijmenga C, Zhernakova A, Weersma RK, 2018. Interplay of host genetics and gut microbiota underlying the onset and clinical presentation of inflammatory bowel disease. Gut 67, 108–119. 10.1136/gutjnl-2016-312135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, Tanoue T, Imaoka A, Itoh K, Takeda K, Umesaki Y, Honda K, Littman DR, 2009. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139, 485–498. 10.1016/j.cell.2009.09.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jašarević E, Morrison KE, Bale TL, 2016. Sex differences in the gut microbiome - Brain axis across the lifespan. Philos Trans R Soc B Biol Sci. 10.1098/rstb.2015.0122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jenne CN, Kubes P, 2013. Immune surveillance by the liver. Nat Immunol 14, 996–1006. 10.1038/ni.2691. [DOI] [PubMed] [Google Scholar]
- Jing Y, Yang D, Bai F, Zhang C, Qin C, Li D, Wang L, Yang M, Chen Z, Li J, 2019. Melatonin treatment alleviates spinal cord injury-induced gut dysbiosis in mice. J. Neurotrauma 36, 2646–2664. 10.1089/neu.2018.6012. [DOI] [PubMed] [Google Scholar]
- Jones TB, Basso DM, Sodhi A, Pan JZ, Hart RP, MacCallum RC, Lee S, Whitacre CC, Popovich PG, 2002. Pathological CNS autoimmune disease triggered by traumatic spinal cord injury: implications for autoimmune vaccine therapy. J Neurosci 22, 2690–2700 doi:20026267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joshi NA, Fass JN, 2011. Sickle: A Sliding-Window, Adaptive, Quality-Based Trimming Tool for FastQ Files (Version 1.33) [Software]. [WWW Document]. URL. https://github.com/najoshi/sickle. [Google Scholar]
- Kanehisa M, Sato Y, Kawashima M, Furumichi M, Tanabe M, 2015. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res 44, 457–462. 10.1093/nar/gkv1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kang DD, Froula J, Egan R, Wang Z, 2015. MetaBAT, an efficient tool for accurately reconstructing single genomes from complex microbial communities. PeerJ 3, e1165. 10.7717/peerj.1165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kang DW, Adams JB, Gregory AC, Borody T, Chittick L, Fasano A, Khoruts A, Geis E, Maldonado J, McDonough-Means S, Pollard EL, Roux S, Sadowsky MJ, Lipson KS, Sullivan MB, Caporaso JG, Krajmalnik-Brown R, 2017. Microbiota transfer therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: An open-label study. Microbiome 5, 10 10.1186/s40168-016-0225-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kernbauer E, Ding Y, Cadwell K, 2014. An enteric virus can replace the beneficial function of commensal bacteria. Nature 516, 94 10.1038/nature13960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kigerl KA, Hall JCE, Wang L, Mo X, Yu Z, Popovich PG, 2016. Gut dysbiosis impairs recovery after spinal cord injury. J Exp Med 213, 2603–2620. 10.1084/jem.20151345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim M-S, Bae J-W, 2016. Spatial disturbances in altered mucosal and luminal gut viromes of diet-induced obese mice. Environ Microbiol 18, 1498–1510. 10.1111/1462-2920.13182. [DOI] [PubMed] [Google Scholar]
- Kopp MA, Watzlawick R, Martus P, Failli V, Finkenstaedt FW, Chen Y, DeVivo MJ, Dirnagl U, Schwab JM, 2017. Long-term functional outcome in patients with acquired infections after acute spinal cord injury. Neurology 88, 892–900. 10.1212/WNL.0000000000003652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kostic AD, Gevers D, Siljander H, Vatanen T, Hyötyläinen T, Hämäläinen A-M, Peet A, Tillmann V, Pöhö P, Mattila I, Lähdesmäki H, Franzosa EA, Vaarala O, de Goffau M, Harmsen H, Ilonen J, Virtanen SM, Clish CB, Orešič M, Huttenhower C, Knip M, Xavier RJ, 2015. The dynamics of the human infant gut microbiome in development and in progression toward Type 1 diabetes. Cell Host Microbe 17, 260–273. 10.1016/j.chom.2015.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krause JS, Kemp B, Coker J, 2000. Depression after spinal cord injury: relation to gender, ethnicity, aging, and socioeconomic indicators. Arch Phys Med Rehabil 81, 1099–1109. [DOI] [PubMed] [Google Scholar]
- Kreydin E, Welk B, Chung D, Clemens Q, Yang C, Danforth T, Gousse A, Kielb S, Kraus S, Mangera A, Reid S, Szell N, Cruz F, Chartier-Kastler E, Ginsberg DA, 2018. Surveillance and management of urologic complications after spinal cord injury. World J Urol. 10.1007/s00345-018-2345-0. [DOI] [PubMed] [Google Scholar]
- Kriegel MA, Sefik E, Hill JA, Wu HJ, Benoist C, Mathis D, 2011. Naturally transmitted segmented filamentous bacteria segregate with diabetes protection in nonobese diabetic mice. Proc Natl Acad Sci USA 108, 11548–11553. 10.1073/pnas.1108924108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krych L, Hansen CH, Hansen AK, van den Berg FW, Nielsen DS, 2013. Quantitatively different, yet qualitatively alike: a meta-analysis of the mouse core gut microbiome with a view towards the human gut microbiome. PLoS One 8, e62578. 10.1371/journal.pone.0062578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kulikov AS, Prjibelski AD, Tesler G, Vyahhi N, Sirotkin AV, Pham S, Dvorkin M, Pevzner PA, Bankevich A, Nikolenko SI, Pyshkin AV, Nurk S, Gurevich AA, Antipov D, Alekseyev MA, Lesin VM, 2012. SPAdes: a new genome assembly algorithm and its applications to single-cell sequencing. J Comput Biol 19, 455–477. 10.1089/cmb.2012.0021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kurokawa S, Kishimoto T, Mizuno S, Masaoka T, Naganuma M, Liang KC,Kitazawa M, Nakashima M, Shindo C, Suda W, Hattori M, Kanai T, Mimura M, 2018. The effect of fecal microbiota transplantation on psychiatric symptoms among patients with irritable bowel syndrome, functional diarrhea and functional constipation: An open-label observational study. J Affect Disord 235, 506–512. 10.1016/j.jad.2018.04.038. [DOI] [PubMed] [Google Scholar]
- Lal S, Kirkup AJ, Brunsden AM, Thompson DG, Grundy D, 2001. Vagal afferent responses to fatty acids of different chain length in the rat. Am J Physiol Gastrointest Liver Physiol 281, G907–G915. 10.1152/ajpgi.2001.281.4.G907. [DOI] [PubMed] [Google Scholar]
- Lane DJ, Pace B, Olsen GJ, Stahl DA, Sogin ML, Pace NR, 1985. Rapid determination of 16S ribosomal RNA sequences for phylogenetic analyses. Proc Natl Acad Sci U S A 82, 6955–6959. 10.1073/pnas.82.20.6955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Langmead B, Salzberg SL, 2012. Fast gapped-read alignment with Bowtie 2. Nat Methods 9, 357–359. 10.1038/nmeth.1923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Laslett D, Canback B, 2004. ARAGORN, a program to detect tRNA genes and tmRNA genes in nucleotide sequences. Nucleic Acids Res 32, 11–16. 10.1093/nar/gkh152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee YK, Menezes JS, Umesaki Y, Mazmanian SK, 2011. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A 108 (Suppl), 4615–4622. 10.1073/pnas.1000082107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levatte MA, Mabon PJ, Weaver LC, Dekaban GA, 1998. Simultaneous identification of two populations of sympathetic preganglionic neurons using recombinant herpes simplex virus type 1 expressing different reporter genes. Neuroscience 82, 1253–1267. [DOI] [PubMed] [Google Scholar]
- Level N, 2018. Spinal Cord Injury Facts and Figures at a Glance Re-Hospitalization Cause of Death. [Google Scholar]
- Ley RE, 2010. Obesity and the human microbiome. Curr Opin Gastroenterol 26, 5–11. 10.1097/MOG.0b013e328333d751. [DOI] [PubMed] [Google Scholar]
- Ley RE, Turnbaugh PJ, Klein S, Gordon JI, 2006. Microbial ecology: human gut microbes associated with obesity. Nature 444, 1022–1023. 10.1038/4441022a. [DOI] [PubMed] [Google Scholar]
- Li H, Durbin R, 2009. Fast and Accurate Short Read Alignment with Burrows-Wheeler Transform. 25 pp. 1754–1760. 10.1093/bioinformatics/btp324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li D, Liu C-M, Luo R, Sadakane K, Lam T-W, 2015. MEGAHIT: an ultra-fast single-node solution for large and complex metagenomics assembly via succinct de Bruijn graph. Bioinformatics 31, 1674–1676. 10.1093/bioinformatics/btv033. [DOI] [PubMed] [Google Scholar]
- Liddle RA, 2019. Neuropods. Cell Mol Gastroenterol Hepatol 7, 739–747. 10.1016/j.jcmgh.2019.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lim ES, Zhou Y, Zhao G, Bauer IK, Droit L, Ndao IM, Warner BB, Tarr PI, Wang D, Holtz LR, 2015. Early life dynamics of the human gut virome and bacterial microbiome in infants. Nat Med 21, 1228–1234. 10.1038/nm.3950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lima-Mendez G, Faust K, Henry N, Decelle J, Colin S, Carcillo F, Chaffron S, Ignacio-Espinosa JC, Roux S, Vincent F, Bittner L, Darzi Y, Wang J, Audic S, Berline L, Bontempi G, Cabello AM, Coppola L, Cornejo-Castillo FM, d’Ovidio F, De Meester L, Ferrera I, Garet-Delmas MJ, Guidi L, Lara E, Pesant S, Royo- Llonch M, Salazar G, Sánchez P, Sebastian M, Souffreau C, Dimier C, Picheral M, Searson S, Kandels-Lewis S, Gorsky G, Not F, Ogata H, Speich S, Stemmann L, Weissenbach J, Wincker P, Acinas SG, Sunagawa S, Bork P, Sullivan MB, Karsenti E, Bowler C, de Vargas C, Raes J, coordinators TO, 2015. Ocean plankton. Determinants of community structure in the global plankton interactome. Science (80-) 348, 1262073. 10.1126/science.1262073. [DOI] [PubMed] [Google Scholar]
- Lowe TM, Eddy SR, 1997. tRNAscan-SE: a program for improved detection of transfer RNA genes in genomic sequence. Nucleic Acids Res 25 (5), 955–964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu YY, Chen T, Fuhrman JA, Sun F, 2016. COCACOLA: binning metagenomic contigs using sequence COmposition, read CoverAge, CO-alignment and paired-end read LinkAge. Bioinformatics 33, btw290. 10.1093/bioinformatics/btw290. [DOI] [PubMed] [Google Scholar]
- Lucin KM, Sanders VM, Jones TB, Malarkey WB, Popovich PG, 2007. Impaired antibody synthesis after spinal cord injury is level dependent and is due to sympathetic nervous system dysregulation. Exp Neurol 207, 75–84. 10.1016/j.expneurol.2007.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luedtke K, Bouchard SM, Woller SA, Funk MK, Aceves M, Hook MA, 2014. Assessment of depression in a rodent model of spinal cord injury. J Neurotrauma 31, 1107–1121. 10.1089/neu.2013.3204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ly M, Jones MB, Abeles SR, Santiago-Rodriguez TM, Gao J, Chan IC, Ghose C, Pride DT, 2016. Transmission of viruses via our microbiomes. Microbiome 4, 64 10.1186/s40168-016-0212-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mabon PJ, LeVatte MA, Dekaban GA, Weaver LC, 1997. Identification of sympathetic preganglionic neurons controlling the small intestine in hamsters using a recombinant herpes simplex virus type-1. Brain Res 753, 245–250. [DOI] [PubMed] [Google Scholar]
- Manfredo Vieira S, Hiltensperger M, Kumar V, Zegarra-Ruiz D, Dehner C, Khan N, Costa FRC, Tiniakou E, Greiling T, Ruff W, Barbieri A, Kriegel C, Mehta SS, Knight JR, Jain D, Goodman AL, Kriegel MA, 2018. Translocation of a gut pathobiont drives autoimmunity in mice and humans. Science (80-) 359, 1156–1161. 10.1126/science.aar7201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Manrique P, Dills M, Young M, Young MJ, 2017. The human gut phage community and its implications for health and disease. Viruses 9, 141 10.3390/v9060141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Markle JGM, Frank DN, Mortin-Toth S, Robertson CE, Feazel LM, Rolle-Kampczyk U, Von Bergen M, McCoy KD, Macpherson AJ, Danska JS, 2013. Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science (80-) 339, 1084–1088. 10.1126/science.1233521. [DOI] [PubMed] [Google Scholar]
- Minot S, Sinha R, Chen J, Li H, Keilbaugh SA, Wu GD, Lewis JD, Bushman D, 2011. The human gut virome: Inter-individual variation and dynamic response to diet. Genome Res 21, 1616 10.1101/gr.122705.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mirzaei MK, Maurice CF, 2017. Ménage à trois in the human gut: interactions between host, bacteria and phages. Nat Rev Microbiol 15, 397–408. 10.1038/nrmicro.2017.30. [DOI] [PubMed] [Google Scholar]
- Monaco CL, Gootenberg DB, Zhao G, Handley SA, Ghebremichael MS, Lim ES, Lankowski A, Baldridge MT, Wilen CB, Flagg M, Norman JM, Keller BC, Luévano JM, Wang D, Boum Y, Martin JN, Hunt PW, Bangsberg DR, Siedner MJ, Kwon DS, Virgin HW, 2016. Altered virome and bacterial microbiome in human immunodeficiency virus-associated acquired immunodeficiency syndrome. Cell Host Microbe 19, 311–322. 10.1016/j.chom.2016.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moreno-Gallego JL, Chou SP, Di Rienzi SC, Goodrich JK, Spector TD, Bell JT, Youngblut ND, Hewson I, Reyes A, Ley RE, 2019. Virome diversity correlates with intestinal microbiome diversity in adult monozygotic twins. Cell Host Microbe 25, 261–272.e5. https://doi.Org/10.1016/j.chom.2019.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morton SC, Shekelle PG, Adams JL, Bennett C, Dobkin BH, Montgomerie J, Vickrey BG, 2002. Antimicrobial prophylaxis for urinary tract infection in persons with spinal cord dysfunction. Arch Phys Med Rehabil 83, 129–138. 10.1053/apmr.2002.26605. [DOI] [PubMed] [Google Scholar]
- Myers SA, Gobejishvili L, Saraswat Ohri S, Garrett Wilson C, Andres KR, Riegler AS, Donde H, Joshi-Barve S, Barve S, Whittemore SR, 2019. Following spinal cord injury, PDE4B drives an acute, local inflammatory response and a chronic, systemic response exacerbated by gut dysbiosis and endotoxemia. Neurobiol Dis 124, 353–363. 10.1016/j.nbd.2018.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neufeld KM, Kang N, Bienenstock J, Foster JA, 2011. Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterol Motil 23, 255–64, e119. 10.1111/j.1365-2982.2010.01620.x. [DOI] [PubMed] [Google Scholar]
- Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, Pettersson S,2012. Host-gut microbiota metabolic interactions. Science (80-) 336, 1262–1267. 10.1126/science.1223813. [DOI] [PubMed] [Google Scholar]
- Nicotra A, Critchley HD, Mathias CJ, Dolan RJ, 2006. Emotional and autonomic consequences of spinal cord injury explored using functional brain imaging. Brain 129, 718–728. 10.1093/brain/awh699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Noble LJ, Wrathall JR, 1989. Distribution and time course of protein extravasation in the rat spinal cord after contusive injury. Brain Res 482, 57–66. 10.1016/0006-8993(89)90542-8. [DOI] [PubMed] [Google Scholar]
- Norman JM, Handley SA, Baldridge MT, Droit L, Liu CY, Keller BC, Kambal A, Monaco CL, Zhao G, Fleshner P, Stappenbeck TS, McGovern DPB, Keshavarzian A, Mutlu EA, Sauk J, Gevers D, Xavier RJ, Wang D, Parkes M, Virgin HW, 2015. Disease-specific alterations in the enteric virome in inflammatory bowel disease. Cell 160, 447–460. 10.1016/j.cell.2015.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Connor G, Jeffrey E, Madorma D, Marcillo A, Abreu MT, Deo SK, Dietrich WD, Daunert S, 2018. Investigation of microbiota alterations and intestinal inflammation post-spinal cord injury in rat model. J Neurotrauma. 10.1089/neu.2017.5349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ochoa-Repáraz J, Mielcarz DW, Haque-Begum S, Kasper LH, 2010. Induction of a regulatory B cell population in experimental allergic encephalomyelitis by alteration of the gut commensal microflora. Gut Microbes 1, 103–108. 10.4161/gmic.1.2.11515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olsen I, Yamazaki K, 2019. Can oral bacteria affect the microbiome of the gut? J Oral Microbiol 11, 1586422. 10.1080/20002297.2019.1586422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ott SJ, Waetzig GH, Rehman A, Moltzau-Anderson J, Bharti R, Grasis JA, Cassidy L, Tholey A, Fickenscher H, Seegert D, Rosenstiel P, Schreiber S,2017. Efficacy of sterile fecal filtrate transfer for treating patients with Clostridium difficile infection. Gastroenterology 152, 799–811.e7. 10.1053/j.gastro.2016.11.010. [DOI] [PubMed] [Google Scholar]
- Parks D, 2017. UniteM [WWW Document]. URL. https://github.com/dparks1134/UniteM.
- Parks DH, Chuvochina M, Waite DW, Rinke C, Skarshewski A, Chaumeil P-A, Hugenholtz P, 2018. A standardized bacterial taxonomy based on genome phylogeny substantially revises the tree of life. Nat Biotechnol 36, 996 10.1038/nbt.4229. [DOI] [PubMed] [Google Scholar]
- Peng Y, Leung HCM, Yiu SM, Chin FYL, 2012. IDBA-UD: a de novo assembler for single-cell and metagenomic sequencing data with highly uneven depth. Bioinformatics 28, 1420–1428. 10.1093/bioinformatics/bts174. [DOI] [PubMed] [Google Scholar]
- Perez-Burgos A, Wang L, McVey Neufeld KA, Mao YK, Ahmadzai M, Janssen LJ, Stanisz AM, Bienenstock J, Kunze WA, 2015. The TRPV1 channel in rodents is a major target for antinociceptive effect of the probiotic Lactobacillus reuteri DSM 17938. J Physiol 593, 3943–3957. 10.1113/JP270229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Popovich PG, Horner PJ, Mullin BB, Stokes BT, 1996. A quantitative spatial analysis of the blood-spinal cord barrier I. Permeability changes after experimental spinal contusion injury. Exp Neurol 142, 258–275. 10.1006/exnr.1996.0196. [DOI] [PubMed] [Google Scholar]
- Prüss H, Tedeschi A, Thiriot A, Lynch L, Loughhead SM, Stutte S, Mazo IB, Kopp MA, Brommer B, Blex C, Geurtz LC, Liebscher T, Niedeggen A, Dirnagl U, Bradke F, Volz MS, DeVivo MJ, Chen Y, von Andrian UH, Schwab JM,2017. Spinal cord injury-induced immunodeficiency is mediated by a sympathetic-neuroendocrine adrenal reflex. Nat Neurosci 20, 1549–1559. 10.1038/nn.4643. [DOI] [PubMed] [Google Scholar]
- Quince C, Walker AW, Simpson JT, Loman NJ, Segata N, 2017. Shotgun meta-genomics, from sampling to analysis. Nat Biotechnol 35, 833–844. 10.1038/nbt.3935. [DOI] [PubMed] [Google Scholar]
- Rabosky DL, 2014. Automatic detection of key innovations, rate shifts, and diversity-dependence on phylogenetic trees. Cit Rabosky DL 9, 89543 10.1371/journal.pone.0089543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rankin KC, O’Brien LC, Segal L, Khan MR, Gorgey AS, 2017. Liver adiposity and metabolic profile in individuals with chronic spinal cord injury. Biomed Res Int 2017, 1364818. 10.1155/2017/1364818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rappé MS, Giovannoni SJ, 2003. The uncultured microbial majority. Annu Rev Microbiol 57, 369–394. 10.1146/annurev.micro.57.030502.090759. [DOI] [PubMed] [Google Scholar]
- Ren J, Song K, Deng C, Ahlgren NA, Fuhrman JA, Li Y, Xie X, Sun F, 2018. Identifying Viruses from Metagenomic Data by Deep Learning. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reyes A, Haynes M, Hanson N, Angly FE, Heath AC, Rohwer F, Gordon JI, 2010. Viruses in the faecal microbiota of monozygotic twins and their mothers. Nature 466, 334–338. 10.1038/nature09199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reyes A, Semenkovich NP, Whiteson K, Rohwer F, Gordon JI, 2012. Going viral: next-generation sequencing applied to phage populations in the human gut. Nat Rev Microbiol 10, 607–617. 10.1038/nrmicro2853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roach DR, Leung CY, Henry M, Morello E, Singh D, Di Santo JP, Weitz JS, Debarbieux L, 2017. Synergy between the host immune system and bacteriophage is essential for successful phage therapy against an acute respiratory pathogen. Cell Host Microbe 22, 38–47.e4. https://doi.Org/10.1016/j.chom.2017.06.018. [DOI] [PubMed] [Google Scholar]
- Roth E, Davidoff G, Thomas P, Doljanac R, Dijkers M, Berent S, Morris J,Yarkony G, 1989. A controlled study of neuropsychological deficits in acute spinal cord injury patients. Paraplegia 27, 480–489. 10.1038/sc.1989.75. [DOI] [PubMed] [Google Scholar]
- Rothhammer V, Mascanfroni ID, Bunse L, Takenaka MC, Kenison JE, Mayo L, Chao CC, Patel B, Yan R, Blain M, Alvarez JI, Kébir H, Anandasabapathy N, Izquierdo G, Jung S, Obholzer N, Pochet N, Clish CB, Prinz M, Prat A, Antel J, Quintana FJ, 2016. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat Med 22, 586–597. 10.1038/nm.4106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rothhammer V, Borucki DM, Tjon EC, Takenaka MC, Chao CC, Ardura-Fabregat A, De Lima KA, Gutiérrez-Vazquez C, Hewson P, Staszewski O, Blain M, Healy L, Neziraj T, Borio M, Wheeler M, Dragin LL, Laplaud DA, Antel J, Alvarez JI, Prinz M, Quintana FJ, 2018. Microglial control of astrocytes in response to microbial metabolites. Nature 557, 724–728. 10.1038/s41586-018-0119-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Round JL, Mazmanian SK, 2009. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol 9, 313–323. 10.1038/nri2515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roux S, Enault F, Hurwitz BL, Sullivan MB, 2015. VirSorter: mining viral signal from microbial genomic data. PeerJ 3, e985. 10.7717/peerj.985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roux S, Brum JR, Dutilh BE, Sunagawa S, Duhaime MB, Loy A, Poulos BT, Solonenko N, Lara E, Poulain J, Pesant S, Kandels-Lewis S, Dimier C, Picheral M, Searson S, Cruaud C, Alberti A, Duarte CM, Gasol JM, Vaqué D, Bork P, Acinas SG, Wincker P, Sullivan MB, Coordinators TO, 2016. Ecogenomics and potential biogeochemical impacts of globally abundant ocean viruses. Nature 537, 689–693. 10.1038/nature19366. [DOI] [PubMed] [Google Scholar]
- Sauerbeck AD, Laws JL, Bandaru VV, Popovich PG, Haughey NJ, McTigue DM, 2015. Spinal cord injury causes chronic liver pathology in rats. J Neurotrauma 32, 159–169. 10.1089/neu.2014.3497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scarpellini E, Ianiro G, Attili F, Bassanelli C, De Santis A, Gasbarrini A, 2015. The human gut microbiota and virome: potential therapeutic implications. Dig Liver Dis 47, 1007–1012. 10.1016/j.dld.2015.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schaedler RW, Dubos R, Costello R, 1965. The development of the bacterial flora in the gastrointestinal tract of mice. J Exp Med 122, 59–66. 10.1084/jem.122.1.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schwab JM, Zhang Y, Kopp MA, Brommer B, Popovich PG, 2014. The paradox of chronic neuroinflammation, systemic immune suppression, autoimmunity after traumatic chronic spinal cord injury. Exp Neurol 258, 121–129. 10.1016/j.expneurol.2014.04.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Segata N, Haake SK, Mannon P, Lemon KP, Waldron L, Gevers D, Huttenhower C, Izard J, 2012. Composition of the adult digestive tract bacterial microbiome based on seven mouth surfaces, tonsils, throat and stool samples. Genome Biol 13, R42. 10.1186/gb-2012-13-6-r42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shavelle RM, DeVivo MJ, Brooks JC, Strauss DJ, Paculdo DR, 2015. Improvements in long-term survival after spinal cord injury? Arch Phys Med Rehabil 96, 645–651. 10.1016/j.apmr.2014.11.003. [DOI] [PubMed] [Google Scholar]
- Sheng L, Jena PK, Liu HX, Kalanetra KM, Gonzalez FJ, French SW, Krishnan VV, Mills DA, Wan YJY, 2017. Gender differences in bile acids and microbiota in relationship with gender dissimilarity in steatosis induced by diet and FXR in-activation. Sci Rep 7, 1–12. 10.1038/s41598-017-01576-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi C, Jia T, Mendez-Ferrer S, Hohl TM, Serbina NV, Lipuma L, Leiner I, Li MO, Frenette PS, Pamer EG, 2011. Bone marrow mesenchymal stem and progenitor cells induce monocyte emigration in response to circulating toll-like receptor ligands. Immunity 34, 590–601. 10.1016/j.immuni.2011.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shin JC, Goo HR, Yu SJ, Kim DH, Yoon SY, 2012. Depression and quality of life in patients within the first 6 months after the spinal cord injury. Ann Rehabil Med 36, 119–125. 10.5535/arm.2012.36.1.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sieber CMK, Probst AJ, Sharrar A, Thomas BC, Hess M, Tringe SG, Banfield JF, 2018. Recovery of genomes from metagenomes via a dereplication, aggregation and scoring strategy. Nat Microbiol 3, 836–843. 10.1038/s41564-018-0171-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Skelton F, Martin LA, Evans CT, Kramer J, Grigoryan L, Richardson P, Kunik ME, Poon IO, Holmes SA, Trautner BW, 2019. Determining best practices for management of bacteriuria in spinal cord injury: Protocol for a mixed-methods study. J Med Internet Res 21, e12272. 10.2196/12272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stilling RM, Ryan FJ, Hoban AE, Shanahan F, Clarke G, Claesson MJ, Dinan TG, Cryan JF, 2015. Microbes & neurodevelopment - Absence of microbiota during early life increases activity-related transcriptional pathways in the amygdala. Brain Behav Immunol 50, 209–220. 10.1016/j.bbi.2015.07.009. [DOI] [PubMed] [Google Scholar]
- Straub RH, Wiest R, Strauch UG, Härle P, Schölmerich J, 2006. The role of the sympathetic nervous system in intestinal inflammation. Gut 55, 1640–1649. 10.1136/gut.2006.091322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sullivan MB, 2015. Viromes, not gene markers, for studying double-stranded DNA virus communities. J Virol 89, 2459–2461. 10.1128/JVI.03289-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sunagawa S, Coelho LP, Chaffron S, Kultima JR, Labadie K, Salazar G,Djahanschiri B, Zeller G, Mende DR, Alberti A, Cornejo-Castillo FM, Costea PI, Cruaud C, d’Ovidio F, Engelen S, Ferrera I, Gasol JM, Guidi L, Hildebrand F, Kokoszka F, Lepoivre C, Lima-Mendez G, Poulain J, Poulos BT, Royo-Llonch M, Sarmento H, Vieira-Silva S, Dimier C, Picheral M, Searson S, Kandels-Lewis S, Bowler C, de Vargas C, Gorsky G, Grimsley N, Hingamp P, Iudicone D, Jaillon O, Not F, Ogata H, Pesant S, Speich S, Stemmann L, Sullivan MB, Weissenbach J, Wincker P, Karsenti E, Raes J, Acinas SG, Bork P, coordinators TO, 2015. Ocean plankton. Structure and function of the global ocean microbiome. Science (80-) 348, 1261359. 10.1126/science.1261359. [DOI] [PubMed] [Google Scholar]
- Tate DG, Forchheimer M, Rodriguez G, Chiodo A, Cameron AP, Meade M,Krassioukov A, 2016. Risk factors associated with neurogenic bowel complications and dysfunction in spinal cord injury. Arch Phys Med Rehabil 97, 1679–1686. 10.1016/j.apmr.2016.03.019. [DOI] [PubMed] [Google Scholar]
- Tatusov RL, Galperin MY, Natale DA, Koonin EV, 2000. The COG database: a tool for genome-scale analysis of protein functions and evolution. Nucleic Acids Res 28, 33–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thevaranjan N, Puchta A, Schulz C, Naidoo A, Szamosi JC, Verschoor CP, Loukov D, Schenck LP, Jury J, Foley KP, Schertzer JD, Larché MJ, Davidson DJ, Verdu EF, Surette MG, Bowdish DME, 2017. Age-associated microbial dysbiosis promotes intestinal permeability, systemic inflammation, and macrophage dysfunction. Cell Host Microbe 21, 455–466.e4. 10.1016/j.chom.2017.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tilg H, Kaser A, 2011. Gut microbiome, obesity, and metabolic dysfunction. J Clin Invest 121, 2126–2132. 10.1172/JCI58109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tillisch K, 2014. The effects of gut microbiota on CNS function in humans. Gut Microbes 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Torres-Barceló C, 2018. The disparate effects of bacteriophages on antibiotic-resistant bacteria. Emerg Microbes Infect 7, 1–12. 10.1038/s41426-018-0169-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tringe SG, von Mering C, Kobayashi A, Salamov AA, Chen K, Chang HW, Podar M, Short JM, Mathur EJ, Detter JC, Bork P, Hugenholtz P, Rubin EM, 2005. Comparative metagenomics of microbial communities. Science (80-) 308, 554–557. 10.1126/science.1107851. [DOI] [PubMed] [Google Scholar]
- Turnbaugh PJ, Gordon JI, 2009. The core gut microbiome, energy balance and obesity. J Physiol 587, 4153–4158. 10.1113/jphysiol.2009.174136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI, 2006. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444, 1027–1031. 10.1038/nature05414. [DOI] [PubMed] [Google Scholar]
- Turnbaugh PJ, Bäckhed F, Fulton L, Gordon JI, 2008. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe 3, 213–223. 10.1016/j.chom.2008.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tyson GW, Chapman J, Hugenholtz P, Allen EE, Ram RJ, Richardson PM, Solovyev VV, Rubin EM, Rokhsar DS, Banfield JF, 2004. Community structure and metabolism through reconstruction of microbial genomes from the environment. Nature 428, 37–43. 10.1038/nature02340. [DOI] [PubMed] [Google Scholar]
- Ueno M, Ueno-Nakamura Y, Niehaus J, Popovich PG, Yoshida Y, 2016. Silencing spinal interneurons inhibits immune suppressive autonomic reflexes caused by spinal cord injury. Nat Neurosci 19, 784–787. 10.1038/nn.4289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uritskiy GV, DiRuggiero J, Taylor J, 2018. MetaWRAP—a flexible pipeline for genome-resolved metagenomic data analysis. Microbiome 6, 158 10.1186/s40168-018-0541-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Westerloo DJ, 2010. The vagal immune reflex: a blessing from above. Wien Med Wochenschr 160, 112–117. 10.1007/s10354-010-0761-x. [DOI] [PubMed] [Google Scholar]
- Vik DR, Roux S, Brum JR, Bolduc B, Emerson JB, Padilla CC, Stewart FJ, Sullivan MB, 2017. Putative archaeal viruses from the mesopelagic ocean. PeerJ 5, e3428 10.7717/peerj.3428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Villarroel J, Kleinheinz K, Jurtz V, Zschach H, Lund O, Nielsen M, Larsen M, Villarroel J, Kleinheinz KA, Jurtz VI, Zschach H, Lund O, Nielsen M, Larsen MV, 2016. HostPhinder: a phage host prediction tool. Viruses 8, 116 10.3390/v8050116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whetstone WD, Hsu JYC, Eisenberg M, Werb Z, Noble-Haeusslein LJ, 2003. Blood-spinal cord barrier after spinal cord injury: relation to revascularization and wound healing. J Neurosci Res 74, 227–239. 10.1002/jnr.10759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wikoff WR, Anfora AT, Liu J, Schultz PG, Lesley SA, Peters EC, Siuzdak G, 2009. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci USA 106, 3698–3703. 10.1073/pnas.0812874106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woese CR, Fox GE, 1977. Phylogenetic structure of the prokaryotic domain: the primary kingdoms. Proc Natl Acad Sci U S A 74, 5088–5090. 10.1073/pnas.74.11.5088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woodcroft B, 2019. CoverM [WWW Document]. URL. https://github.com/wwood/CoverM/.
- Wu HJ, Ivanov II, Darce J, Hattori K, Shima T, Umesaki Y, Littman DR, Benoist C, Mathis D, 2010. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 32, 815–827. 10.1016/j.immuni.2010.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu J, Zhao Z, Sabirzhanov B, Stoica BA, Kumar A, Luo T, Skovira J, Faden AI, 2014. Spinal cord injury causes brain inflammation associated with cognitive and affective changes: role of cell cycle pathways. J Neurosci 34, 10989–11006. 10.1523/JNEUR0SCI.5110-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu Y-W, Simmons BA, Singer SW, 2016. MaxBin 2.0: an automated binning algorithm to recover genomes from multiple metagenomic datasets. Bioinformatics 32, 605–607. 10.1093/bioinformatics/btv638. [DOI] [PubMed] [Google Scholar]
- Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, Nagler CR, Ismagilov RF, Mazmanian SK, Hsiao EY, 2015. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 161, 264–276. 10.1016/j.cell.2015.02.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yutin N, Makarova KS, Gussow AB, Krupovic M, Segall A, Edwards RA, Koonin V, 2018. Discovery of an expansive bacteriophage family that includes the most abundant viruses from the human gut. Nat Microbiol 3, 38–46. 10.1038/s41564-017-0053-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y, Guan Z, Reader B, Shawler T, Mandrekar-Colucci S, Huang K, Weil Z, Bratasz A, Wells J, Powell ND, Sheridan JF, Whitacre CC, Rabchevsky AG, Nash MS, Popovich PG, 2013. Autonomic dysreflexia causes chronic immune suppression after spinal cord injury. J Neurosci 33, 12970–12981. 10.1523/JNEUR0SCI.1974-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang C, Zhang W, Zhang J, Jing Y, Yang M, Du L, Gao F, Gong H, Chen L, Li J, Liu H, Qin C, Jia Y, Qiao J, Wei B, Yu Y, Zhou H, Liu Z, Yang D, 2018. Gut microbiota dysbiosis in male patients with chronic traumatic complete spinal cord injury. J Transl Med 16, 353 10.1186/s12967-018-1735-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao G, Vatanen T, Droit L, Park A, Kostic AD, Poon TW, Vlamakis H, Siljander H, Härkönen T, Hämäläinen A-M, Peet A, Tillmann V, Ilonen J, Wang D, Knip M, 2018. Intestinal virome changes precede autoimmunity in type I diabetes-susceptible children. Proc Natl Acad Sci 115 10.1073/pnas.1706359114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng P, Zeng B, Zhou C, Liu M, Fang Z, Xu X, Zeng L, Chen J, Fan S, Du X, Zhang X, Yang D, Yang Y, Meng H, Li W, Melgiri ND, Licinio J, Wei H, Xie P, 2016. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Mol Psychiatry 21, 786–796. 10.1038/mp.2016.44. [DOI] [PubMed] [Google Scholar]
- Zheng T, Li J, Ni Y, Kang K, Misiakou M-A, Imamovic L, Chow BKC, Rode AA, Bytzer P, Sommer M, Panagiotou G, 2019. Mining, analyzing, and integrating viral signals from metagenomic data. Microbiome 7, 42 10.1186/s40168-019-0657-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zuo T, Wong SH, Lam K, Lui R, Cheung K, Tang W, Ching JYL, Chan PKS, Chan MCW, Wu JCY, Chan FKL, Yu J, Sung JJY, Ng SC, 2018. Bacteriophage transfer during faecal microbiota transplantation in Clostridium difficile infection is associated with treatment outcome. Gut 67, 634–643. 10.1136/gutjnl-2017-313952. [DOI] [PMC free article] [PubMed] [Google Scholar]


