ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Pathogenesis of Salmonella gastroenteritis

Pathogenesis of Salmonella gastroenteritis
Literature review current through: Jan 2024.
This topic last updated: Jun 07, 2023.

INTRODUCTION — Salmonellae cause a broad range of infections, including gastroenteritis, enteric fever, bacteremia, endovascular infections, and focal infections such as osteomyelitis and abscesses [1]. Salmonellae are facultative anaerobic gram-negative bacilli and usually enter the body via the gastrointestinal (GI) tract, where they can persist for long periods of time. Salmonellae can act as both commensals and pathogens and are found in the GI tracts of domestic and wild animals, including insects, reptiles, birds, and mammals.

Although there are many types of Salmonella, they can be divided into two broad categories: those that cause typhoid and enteric fever and those that primarily induce gastroenteritis:

The typhoidal Salmonella, such as Salmonella Typhi or Salmonella Paratyphi primarily colonize humans, are transmitted via the consumption of fecally contaminated food or water, and cause a systemic illness usually with little or no diarrhea. (See "Enteric (typhoid and paratyphoid) fever: Epidemiology, clinical manifestations, and diagnosis".)

The much broader group of nontyphoidal Salmonella usually results from improperly handled food that has been contaminated by animal or human fecal material. It can also be acquired via the fecal-oral route, either from other humans or farm or pet animals [2]. (See "Nontyphoidal Salmonella: Microbiology and epidemiology".)

The bacterial and host factors that contribute to Salmonella gastroenteritis will be reviewed here (figure 1). The approach to patients with Salmonella in a stool culture is discussed separately. (See "Nontyphoidal Salmonella: Gastrointestinal infection and asymptomatic carriage".)

GENERAL ISSUES — A number of different serovars of Salmonella cause human infection. The source of infection typically is from food. For every laboratory-confirmed case of Salmonella, there are many other undetected cases; one estimate is 29 infections for every 1 culture-confirmed case [3].

Serotypes — The 2000-plus serovars of Salmonella can be characterized by three major antigens: the somatic O antigen, which is derived from the LPS cell wall component; the flagellar H antigen, and the surface Vi antigen. The O-antigen is widely used by clinical laboratories to divide Salmonella into serogroups A, B, C1, C2, D, and E (table 1). Significant cross-reactivity can occur between serogroups. The serogroups cannot be used to divide enteric fever-causing strains from gastroenteritis-causing strains: group D contains both Salmonella enteritidis and S. Typhi, while group B contains Salmonella Typhimurium as well as some strains of S. Paratyphi. To investigate a possible outbreak, further identification must be done with bacteriophage typing, restriction fragment length polymorphism analysis, pulsed-field gel electrophoresis, and plasmid profile determination.

The host specificity of different Salmonella serotypes often determines the nature of the clinical illness. Classic typhoid or enteric fever is caused by S. Typhi and S. Paratyphi. These strains are highly adapted for humans and do not colonize or cause disease in animals, though S. Paratyphi has very rarely been associated with domestic animals (see "Pathogenesis of enteric (typhoid and paratyphoid) fever"). Those serotypes which cause enteric fever are frequently not associated with significant diarrheal symptoms, although enteric fever may be heralded by transient diarrhea prior to the onset of fever.

S. enteritidis and S. Typhimurium have broad host ranges and may produce colonization or gastroenteritis in humans, mice, and fowl (see "Nontyphoidal Salmonella: Microbiology and epidemiology"). These serotypes are most frequently associated with gastroenteritis in humans because of the large reservoirs of bacteria in domestic animals. Salmonella choleraesuis (adapted for swine) and Salmonella dublin (adapted for cattle) are associated with human septicemia and metastatic foci of infection but are uncommon causes of gastroenteritis [4]. Salmonella gallinarum-pullorum (adapted for fowl) causes relatively transient illness in humans only after very large inocula are ingested. The molecular bases of these species specificities is complex and currently under investigation.

The epidemiology of Salmonella has evolved over time. A detailed atlas of Salmonella infections from 1968 to 2011 in the United States is available through the Centers for Disease Control and Prevention (CDC) [5]. During the 2000s, the incidence of S. Typhimurium decreased, while serotypes Newport, Mississippi, and Javiana increased in incidence. Specific control programs focusing on contaminated eggs might have led to the reduction of S. enteritidis infections. Rates of antibiotic resistance among several serotypes have been increasing and are an area of significant concern; a substantial proportion of S. Typhimurium and Salmonella newport isolates are multidrug resistant [6]. In the United States, an estimated 6200 culture-confirmed Salmonella infections with clinically important resistance occurred annually from 2004 to 2012 [7]. Most cases of human salmonellosis are almost entirely limited to the serotypes of subspecies Salmonella enterica, with the top five serotypes responsible for human disease being S. Enteritidis, S. Typhimurium, S. Infantis, S. Stanley, and S. Newport; most human infections due to non-enterica subspecies develop in adults with weakened immune systems [8].

Sources of infection — There are an estimated two to four million cases of Salmonellosis per year in the United States, with gastroenteritis as the most common clinical presentation. Nontyphoidal Salmonella account for 9.7 percent of all bacterial foodborne illnesses and 30.6 percent of deaths as of 1999 [9]; between the time periods 1996 to 1998 and 2005, the number of Salmonella cases decreased by 9 percent [10]. Salmonella gastroenteritis usually results from improperly handled food. It can also be acquired via the fecal-oral route, either from other humans or farm or pet animals. Sources of infection are discussed in detail elsewhere. (See "Nontyphoidal Salmonella: Microbiology and epidemiology", section on 'Modes of transmission'.)

Infectious dose — Data on the number of Salmonella organisms required for clinical illness have been obtained from studies on human volunteers and outbreaks in which the vehicle of inoculation was quantitatively cultured [11]. Several general statements about infectious doses can be made:

Large inocula (>104) produce higher rates of illness after shorter incubation periods than small inocula (≤103).

Asymptomatic excretion may occur after ingestion of small inocula, although even very small inocula (5 to 100 organisms) may cause disease in susceptible hosts.

Antibiotic use can reduce the infectious dose necessary to cause disease by diminishing the normally protective indigenous flora (see below).

The infectious dose is lower in patients with clinical conditions associated with a reduction in gastric acidity such as neonates [12], achlorhydric states [13], gastric surgery [14,15], and the use of antacids or H2 blockers [15,16].

Water supplies are contaminated at lower levels than food, resulting in lower attack rates and longer incubation periods in waterborne outbreaks.

Salmonella may persist for months in cheese, frozen meat, or ice cream.

PATHOGENESIS — A number of factors related both to the pathogen and the host influence the pathogenesis of Salmonella gastroenteritis.

Interaction of Salmonellae with enteric host defenses — Ingested microorganisms must traverse the acidic barrier of the stomach in order to establish enteric infection (table 2). Although Salmonella and other enteric pathogens survive poorly at the very low pH encountered in the stomach, Salmonella exhibit increased tolerance to "acid shock" if first exposed to a moderately acidic environment (pH 4 to 5). The organism's ability to adapt to low pH has been called the acid tolerance response [17].

Salmonellae that survive passage through the stomach must then compete with the normal intestinal microbial flora. The indigenous flora is an important but often overlooked barrier to infection with enteric pathogens. Elegant mouse studies performed in the 1960s showed that a single injection of streptomycin reduced the oral infectious dose of S. Typhimurium by over 100,000-fold [18]. Studies in animals have shown that the protective effect of indigenous microbes is due to a variety of factors including competition for nutrients, maintenance of a low luminal pH, and production of inhibitory compounds such as volatile fatty acids [19,20].

These findings appear to be applicable to humans, as can be illustrated by the following observations:

Prophylactic antimicrobial therapy increases the frequency of salmonellosis among travelers [21].

Patients found to be infected during an outbreak of multidrug-resistant S. Typhimurium enteritis used antimicrobials in the month before the onset of illness much more frequently than controls (30 versus 6 percent) [22]. A similar association with prior antimicrobial use was noted in a multistate outbreak of illness caused by Salmonella havana which was pansensitive to antimicrobial agents (30 versus 13 percent) [23]. The association persisted even when controlled for the presence of underlying illness or immunosuppression.

S. Typhimurium has a marked and unique growth advantage over other bacteria when using a specific nutrient, ethanolamine, which is released from host tissue in the lumen of the inflamed intestine and is not utilizable by competing bacteria [24]. Thus, by inducing intestinal inflammation, S. Typhimurium sidesteps nutritional competition and creates a significant survival advantage for itself.

In addition to competition from the indigenous bacterial flora, Salmonella must withstand a gamut of enteric defenses including bile salts, pancreatic enzymes, Paneth cell antimicrobial peptides [25], and secretory IgA [26].

The stringent response (SR) system, mediated by guanosine pentaphosphate ([p]ppGpp), has been implicated in the regulation of invasion, virulence, persistence, and latency of several bacterial pathogens and intracellular obligate parasites; similar S. Typhi mutants show severely impaired virulence-associated traits, such as motility, chemotaxis, and Vi antigenic capsule formation, leading to markedly attenuated adhesion, invasion of the intestinal epithelial cells, and intracellular survival within macrophages [27].

Mechanisms of adherence and invasion — There are two main components to Salmonella infection of the GI tract: adherence and subsequent invasion. Adherence is complex and is mediated by multiple genes. Fimbriae are very important in adhering to and adapting to a eukaryotic cell surface [28]; several fimbrial operons may facilitate adherence [29]. Biofilms may play a role as well [28]. The invasion operon (inv) can also induce adherence [30].

Invasion may be achieved by several different mechanisms:

Salmonella can selectively attach to specialized epithelial cells overlying Peyer's patches in the colon known as M (microfold) cells. These cells are an important portal of entry of Salmonellae and other pathogens into the submucosal lymphoid system [31]. M cells are highly endocytic and can rapidly transfer material from their luminal side to their basal side, where the T cells and antigen-presenting cells reside, ready to elicit an immune response.

It has been suggested that the cells of the columnar epithelium may also be an important common portal of entry for Salmonella, particularly since they greatly outnumber the M cells [32].

Salmonellae can also induce nonphagocytic cells such as enterocytes to internalize them. This process of bacterial-mediated endocytosis has been extensively studied in vitro and appears to be important in the pathogenesis of Salmonella gastroenteritis [33,34].

Invasion can also occur via the dendritic cells that intercalate between epithelial cells by extending protrusions into the gut lumen.

Numerous small foci of solitary intestinal lymphoid tissues (SILTs) with a strong inflammatory response can be found in the murine small intestine; Salmonella can be observed within these SILTs at early stages of infection, and the SILTs may act as portals of entry [35].

Following invasion into the cell, the bacteria remain within a modified phagosome known as the Salmonella-containing vacuole (SCV), within which they will survive and replicate. Type III secretion systems are used to translocate bacterial effector proteins into the host cell, mediating both invasion and vacuole biogenesis. Vacuole biogenesis is a complex and dynamic process involving extensive membrane remodeling, interactions with the endolysosomal pathway, actin rearrangements, and microtubule-based movement and tubule extension [36]. Interferon 1-mediated signaling and control of lysosomal function appear to modulate intestinal epithelial cell death and systemic spread of Salmonella [37].

The mechanisms of persistent Salmonella fecal shedding of Salmonella are under investigation. The shdA gene, which encodes an outer membrane autotransporter protein that binds fibronectin, is required for persistent shedding of S. Typhimurium in mice [38]. This raises the question of whether this locus might mediate adherence of Salmonella at other clinically important structures known to be foci of metastatic infection, such as arteries and bone.

Pathogenicity islands — Two Salmonella "pathogenicity islands" (SPI) have been found, termed SPI-1 and 2, each about 40 kilobases of DNA and located at centrosomes 63 and 30 of the chromosome, respectively [39-44]. Both SPIs encode multiple virulence factors, including Type III secretions systems (TTSS). The TTSS creates a hypodermic needle-like apparatus (figure 2) and injects proteins into the cells, facilitating uptake of the bacteria into those cells. SPI-1 encodes genes needed for nonphagocytic cell invasion via a ruffling mechanism and initiation of intestinal secretory and inflammatory responses. SPI-2 is induced within the cell and contains genes needed for survival and replication within macrophages [45]. Some of these genes induce secretory and inflammatory responses [46,47]. Some of these genes on SPI-1 encode proteins homologous to Shigella proteins which direct the export and translocation of signaling molecules capable of interacting with eukaryotic cells [39,40]. The secreted proteins of both Salmonella and Shigella induce ruffled projections in eukaryotic cell membranes which subsequently invaginate and internalize the invading bacterium within a membrane bound vacuole, called macropinocytosis [48].

These TTSSs facilitate bacterial entry, replication within cells, and target cell transcriptional reprogramming via effector proteins that, although highly conserved across different serovars, have enough differences to explain variations in clinical presentations. Analyzing transcriptional responses of cultured epithelial cells infected with S. Typhi and S. Typhimurium, studies have found serovar-specific transcriptional fingerprints that correlated with activation of specific signal transduction pathways and effector proteins, which may help explain the variations in the pathogenic mechanisms and clinical illnesses [49].

Mutants lacking these genes display reduced virulence. In one study avirulent environmental isolates of Salmonella senftenberg and Salmonella litchfield contained deletions in SPI-1, while clinical isolates of the same serotypes retained the gene cluster [50].

Inflammatory response mechanisms — In addition to facilitating their own uptake, virulent strains of Salmonella are able to induce migration of subepithelial neutrophils across polarized epithelial cells in vitro [51]. A substantial neutrophil infiltration into the intestine occurs in S. Typhimurium-induced colitis in humans; whether this is due to effectors from Salmonella or innate pathways of inflammation triggered by pathogen recognition receptors on cells in the lamina propria is a matter of debate [52]. The paracellular traffic of neutrophils has been hypothesized to induce diarrhea by causing paracellular fluid and electrolyte fluxes. This theory is supported by the observation that strains which do not usually cause enteritis, such as S. Typhi and S. gallinarum, also do not induce neutrophil transmigration [53].

In addition to IL-8, Salmonella induce other proinflammatory cytokines such as GM-CSF, monocyte chemotactic protein-1, and tumor necrosis factor-alpha in colonic epithelial cells, which in turn mediate further immune responses [54]. Macrophages respond to S. Typhimurium infection via flagellin-mediated activation of Ipaf, a NACHT-leucine-rich repeat family member that activates caspase-1 [55]. Activated caspase-1 is required for the secretion of proinflammatory cytokines, such as interleukin (IL)-1beta and IL-18, and is important in host defense against a variety of pathogens; caspase-1-deficient mice are more susceptible to several pathogens including, Shigella, Listeria, and Francisella tularensis [56-58], as well as Salmonella [59].

Lipid A is the biologically active component of lipopolysaccharide (LPS) found in the cell wall of Salmonella and other gram-negative bacteria. Lipid A is toxic to mammalian cells and is a potent immunomodulator. Certain features of the lipid A in Salmonella may correlate with virulence or with activation of host inflammation [60,61]. Lipid A induces toll-like receptor 4 (TLR4)-mediated responses, which are important for host defense against Salmonella infection, and modifications in lipid A as part of Salmonella's adaptation to host environments reduce this signaling [62]. Death in mice from Salmonella may be related to the toxic effect of lipid A, which triggers further production of TNF-alpha and IL-1 beta. S. Typhimurium mutants with a defective lipid A molecule have greatly attenuated virulence in mice [63]. Structural modifications of lipid A are influenced by the Salmonella virulence regulatory locus (phoP/phoQ) which responds to a variety of host intracellular environmental signals [64]. For example, antimicrobial peptides have been shown to be part of the first step in signal transduction across the bacterial membrane, resulting in activation of phoQ and promotion of bacterial virulence [65]. PhoP has also been found to bind a promoter region of a drug efflux system, thus connecting virulence with possible drug resistance [66].

Enterotoxins may also play a role in Salmonella gastroenteritis. An enterotoxin, encoded by the stn gene and antigenically similar to cholera toxin has been identified [67-69]. While many Salmonellae carry the stn gene, only a fraction express the gene, as assessed by CHO cell assay [70]. While the tropism of typhoid toxin to immune cells likely alters innate and adaptive immune responses in Salmonella Typhi pathogenesis, the non-typhoid ortholog Javiana toxin does not cause the same toxicity despite the high degree of amino acid sequence similarity between these proteins. They have markedly different glycan-binding preferences and virulence [71]. Vaccination with S. Javiana or its toxin results in cross-reactive protection against lethal-dose typhoid toxin challenge in mice, which may help advance vaccine development.

Additional characterization of the enterotoxin is needed to assess its role in human pathogenesis.

Survival within phagocytes — Salmonellae have long been known to persist within the reticuloendothelial system. Macrophages may be the main cell type to support bacterial growth in vivo, and this growth is regulated by both the host and the Salmonella [72]. The ability of Salmonella to survive within macrophages contributes to the dissemination of the microorganism from the submucosa to the circulation and the reticuloendothelial system. Intracellular bacterial growth within phagocytes is limited by mechanisms requiring reactive oxygen intermediates, magnesium transporters [73], reactive nitrogen intermediates, lysosomal enzymes, and defensins. Itaconate, an antimicrobial metabolite produced within mitochondria also contributes [74]. S. Typhimurium has been shown to induce succinate accumulation [75], which fuels its own virulence by promoting resistance of antimicrobials and type III secretion of virulence factors. Both the phoP/phoQ virulence regulatory locus [76-78] and the SPI-2 contain genes that are important for survival within macrophages. The importance of this process is illustrated by studies that showed that phoP/phoQ-deleted S. Typhimurium, when used as a vaccine vector, caused no bacteremia or late sequelae in a limited number of subjects who received a large oral inoculum [79]. Isolates of S. Typhimurium that are unable to survive within macrophages in vitro are avirulent in mice [80].

Role of virulence plasmids — Nontyphoidal Salmonellae also carry a variety of virulence plasmids [81]. A highly conserved 8 kilobase region of DNA contained within these plasmids has been associated with the ability of strains to induce bacteremia and persist within the reticuloendothelial system [81,82]. S. Typhimurium has a self-transmissible [83] 90 kilobase virulence plasmid that contains the genes spvRABCD and while it increases the growth rate of Salmonellae in macrophages, it may have a limited contribution to virulence [84]. The virulence plasmid stimulates IL-12 production in a mouse model, which may lead to attenuated T cell proliferation [85].

Immune response to Salmonella infection — Because of the frequency and nonspecific nature of the symptoms of Salmonella gastroenteritis, little is known about the rates of reinfection and correlates of immunity. The innate immune system, cell-mediated immunity, and humoral immunity all play important roles in limiting Salmonella infection.

Innate immune system — The innate immune system plays a critical role in the initial response to Salmonella infection. It may be the determining factor in whether the infection is subclinical or more aggressive. The importance of macrophages and polymorphonuclear (PMN) leukocyte response to Salmonella has been well described. Neutropenic mice have a high mortality in experimental Salmonella infection [86], and depressed PMN function increases the incidence of Salmonella infection in humans. These conditions most commonly include sickle cell anemia [87], malaria [88,89], schistosomiasis [90], and histoplasmosis [91]. In addition, Toll receptor agonists such as LPS, lipoprotein, and flagellin stimulate proinflammatory cytokine production, resulting in TNF-alpha and interleukin responses [32]. Activation of Toll-like receptor 5 and Ipaf by Salmonella flagellin has been a significant finding [92].

Cell-mediated immunity — Cell-mediated immunity plays an important role in clearing infection and protecting against subsequent Salmonella infection. Clinical vigilance in the diagnosis and management of Salmonella infections should be increased in settings associated with cellular immunosuppression. As an example, infection is more severe and prolonged in patients with depressed cellular immunity due to glucocorticoids [93], AIDS [94-96], and malignancy [97]. In one study, nontyphoidal Salmonella (14 percent) were the second most frequent bacterial isolate after Staphylococcus aureus (29 percent) in a study of 249 bacteremias in HIV-infected individuals [98]. Nude mice and mice deficient in alpha-beta T cells are more susceptible to Salmonella infections. Murine models suggest that CD4+ T cells are more contributory than CD8+ T cells [32].

Humoral immune responses — The humoral response to Salmonella infection is complex and may be protective or maladaptive. Its importance in containing Salmonella infection is illustrated by the protective immunity induced in vaccination studies with S. Typhi. Mice deficient in B cells due to a targeted deletion of the Ig-mu gene show increased susceptibility to Salmonella infection and are unable to mount a significant convalescent immune response [99]. In addition, mucosal humoral immune responses may play a contributory role. Murine studies have shown that secretion of large amounts of a single monoclonal IgA directed against S. Typhimurium lipopolysaccharide (LPS) into the intestinal lumen provides significant protection against systemic disease [40]. Furthermore, mucosal antibodies to nontyphoidal Salmonellae appear to inhibit the "take" of the oral live attenuated typhoid fever vaccine Ty21a [100].

However, in a study that evaluated the effect of serum from HIV-infected patients on the killing of S. Typhimurium in vitro, a high serum concentration of antibodies against the LPS of non-typhoidal Salmonellae paradoxically prevented the destruction of Salmonella [101]. The authors postulated that inhibitory antibodies in the serum of HIV-infected individuals could be responsible for this effect by blocking the action of bactericidal antibodies. These results suggest that the preferred vaccine target should be the bacterial outer-membrane proteins of non-typhoidal Salmonellae as opposed to LPS, since LPS could potentially elicit inhibitory rather than the desired bactericidal antibodies.

Interaction with intestinal flora — Both pathogen transmission and pathogen clearance from the gut lumen can be mediated by the microbiota. In one study, mice with low complexity gut flora failed to clear S. Typhimurium from the gut lumen, and pathogen clearance was achieved by transferring a normal complex microbiota [102]. The normal microbiota were able to grow from low numbers in abnormal conditions and gradually clear even very high pathogen loads from the gut lumen, a site inaccessible to most "classical" immune effector mechanisms. Thus, besides preventing pathogen transmission by an intact microbiota (also known as colonization resistance), the microbiota also facilitates pathogen clearance, a second, novel protective function. More recent work suggests this may be mediated by microbiota-enhanced antibacterial interferon-gamma responses [103], which can be augmented by transfer of specific commensal communities. Increasingly, other specific mechanisms are being elucidated. For example, Enterococcus faecium's secreted antigen A (SagA) is sufficient to protect Caenorhabditis elegans against Salmonella and other pathogens. SagA's peptidoglycan hydrolase activity generates muramyl-peptide fragments that enhance epithelial barrier integrity.

In a trial of the oral live-attenuated typhoid vaccine Ty21a, bacterial 16S rRNA pyrosequencing was employed to evaluate whether oral immunization resulted in alterations of the microbiota and whether a given microbiota composition is associated with defined S. Typhi-specific immunological responses [104]. No discernible perturbations of the bacterial assemblage were found after vaccine administration. Among individuals able to mount a positive humoral response, no differences in microbial composition, diversity, or temporal stability were observed; however, individuals displaying broader (multiphasic) cell mediated immune responses harbored more diverse, complex communities. Further studies of the immune-modulatory properties of the human microbiota and the impact on disease transmission, pathogen clearance, and the response to vaccination is warranted.

Persisters — Some non-replicating Salmonella display a reversible, antibiotic-tolerant phenotype and can persist following antibiotic treatment and then revert to growth phase. This is of particular importance for S. Typhi, which has a chronic phase. Research on Salmonella growth and survival during antibiotic treatment has shown that antibiotic killing correlated with single-cell division rates, such that non-dividing Salmonella had the highest survival rates but were rare [105]. Most surviving bacteria were from the more common moderately growing, partially tolerant Salmonella. Using fluorescent single-cell analysis, other studies have shown that Salmonella persisters were part of a nonreplicating population that formed immediately after uptake by macrophages and were induced by vacuolar acidification and nutritional deprivation, similar to conditions that induce Salmonella virulence gene expression [106]. After phagocytosis by naïve macrophages, some persisters were subsequently able to resume intracellular growth, which could be a reservoir for relapsing infection. In response to the gallbladder environment, Salmonella can undergo genetic changes resulting in hyper-biofilm isolates with improved persistence, especially in the presence of gallstones, albeit at the cost of decreased virulence [107].

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on “patient info” and the keyword(s) of interest.)

Basics topics (see "Patient education: Salmonella infection (The Basics)")

SUMMARY

The 2000-plus serovars of Salmonella can be characterized by three major antigens: the somatic O antigen, which is derived from the lipopolysaccharide cell wall component, the flagellar H antigen, and the surface Vi antigen. (See 'Serotypes' above.)

The size of the inoculum is proportional to the frequency and rapidity of disease onset, but even small inocula can cause disease. Enteric host factors that defend against Salmonella infection include the acidic environment of the stomach and the normal intestinal microbial flora. The infectious dose necessary to cause disease can be lower in settings of antibiotic use and reduction of gastric acid. (See 'Infectious dose' above and 'Interaction of Salmonellae with enteric host defenses' above.)

Salmonellae adhere to and invade the gastrointestinal tract and submucosal lymphoid system through several different mechanisms. Bacterial entry into and survival within host cells are facilitated by multiple virulence factors. (See 'Mechanisms of adherence and invasion' above and 'Pathogenicity islands' above.)

The ability of Salmonella to survive within macrophages contributes to the dissemination of the microorganism from the submucosa to the circulation and the reticuloendothelial system. (See 'Survival within phagocytes' above.)

Virulent strains of Salmonella induce multiple host inflammatory responses and cytokines. This is in part mediated by lipid A, a component of lipopolysaccharide in the cell wall. (See 'Inflammatory response mechanisms' above.)

The host innate immune response may determine whether Salmonella infection is subclinical or more aggressive. Adaptive cell-mediated immunity plays an important role in clearing and protecting against subsequent infection. The humoral response is complex and may be protective or maladaptive. (See 'Immune response to Salmonella infection' above.)

An intact host intestinal flora, or microbiome, plays a role in preventing infection through the gastrointestinal route and also facilitates pathogen clearance. (See 'Interaction with intestinal flora' above.)

  1. Qamar FN, Hussain W, Qureshi S. Salmonellosis Including Enteric Fever. Pediatr Clin North Am 2022; 69:65.
  2. Hohmann EL. Nontyphoidal salmonellosis. Clin Infect Dis 2001; 32:263.
  3. Scallan E, Hoekstra RM, Angulo FJ, et al. Foodborne illness acquired in the United States--major pathogens. Emerg Infect Dis 2011; 17:7.
  4. Blaser MJ, Feldman RA. From the centers for disease control. Salmonella bacteremia: reports to the Centers for Disease Control, 1968-1979. J Infect Dis 1981; 143:743.
  5. United States Centers for Disease Control and Prevention. An Atlas of Salmonella in the United States, 1968-2011; 2013. https://www.cdc.gov/salmonella/pdf/salmonella-atlas-508c.pdf (Accessed on June 06, 2019).
  6. McNabb SJ, Jajosky RA, Hall-Baker PA, et al. Summary of notifiable diseases--United States, 2006. MMWR Morb Mortal Wkly Rep 2008; 55:1.
  7. Medalla F, Gu W, Mahon BE, et al. Estimated Incidence of Antimicrobial Drug-Resistant Nontyphoidal Salmonella Infections, United States, 2004-2012. Emerg Infect Dis 2016; 23:29.
  8. Lamas A, Miranda JM, Regal P, et al. A comprehensive review of non-enterica subspecies of Salmonella enterica. Microbiol Res 2018; 206:60.
  9. Mead PS, Slutsker L, Dietz V, et al. Food-related illness and death in the United States. Emerg Infect Dis 1999; 5:607.
  10. Centers for Disease Control and Prevention (CDC). Preliminary FoodNet data on the incidence of infection with pathogens transmitted commonly through food--10 States, United States, 2005. MMWR Morb Mortal Wkly Rep 2006; 55:392.
  11. Blaser MJ, Newman LS. A review of human salmonellosis: I. Infective dose. Rev Infect Dis 1982; 4:1096.
  12. Agunod M, Yamaguchi N, Lopez R, et al. Correlative study of hydrochloric acid, pepsin, and intrinsic factor secretion in newborns and infants. Am J Dig Dis 1969; 14:400.
  13. Riley LW, Cohen ML, Seals JE, et al. Importance of host factors in human salmonellosis caused by multiresistant strains of Salmonella. J Infect Dis 1984; 149:878.
  14. KUNZ LJ, WADDELL WR. Association of Salmonella enteritis with operations on the stomach. N Engl J Med 1956; 255:555.
  15. Neal KR, Briji SO, Slack RC, et al. Recent treatment with H2 antagonists and antibiotics and gastric surgery as risk factors for Salmonella infection. BMJ 1994; 308:176.
  16. Giannella RA, Broitman SA, Zamcheck N. Gastric acid barrier to ingested microorganisms in man: studies in vivo and in vitro. Gut 1972; 13:251.
  17. Foster JW. Low pH adaptation and the acid tolerance response of Salmonella typhimurium. Crit Rev Microbiol 1995; 21:215.
  18. BOHNHOFF M, MILLER CP, MARTIN WR. RESISTANCE OF THE MOUSE'S INTESTINAL TRACT TO EXPERIMENTAL SALMONELLA INFECTION. I. FACTORS WHICH INTERFERE WITH THE INITIATION OF INFECTION BY ORAL INOCULATION. J Exp Med 1964; 120:805.
  19. Que JU, Casey SW, Hentges DJ. Factors responsible for increased susceptibility of mice to intestinal colonization after treatment with streptomycin. Infect Immun 1986; 53:116.
  20. Tannock GW, Savage DC. Indigenous microorganisms prevent reduction in cecal size induced by Salmonella typhimurium in vaccinated gnotobiotic mice. Infect Immun 1976; 13:172.
  21. Mentzing LO, Ringertz O. Salmonella infection in tourists. 2. Prophylaxis against salmonellosis. Acta Pathol Microbiol Scand 1968; 74:405.
  22. Ryan CA, Nickels MK, Hargrett-Bean NT, et al. Massive outbreak of antimicrobial-resistant salmonellosis traced to pasteurized milk. JAMA 1987; 258:3269.
  23. Pavia AT, Shipman LD, Wells JG, et al. Epidemiologic evidence that prior antimicrobial exposure decreases resistance to infection by antimicrobial-sensitive Salmonella. J Infect Dis 1990; 161:255.
  24. Thiennimitr P, Winter SE, Winter MG, et al. Intestinal inflammation allows Salmonella to use ethanolamine to compete with the microbiota. Proc Natl Acad Sci U S A 2011; 108:17480.
  25. Selsted ME, Miller SI, Henschen AH, Ouellette AJ. Enteric defensins: antibiotic peptide components of intestinal host defense. J Cell Biol 1992; 118:929.
  26. Michetti P, Mahan MJ, Slauch JM, et al. Monoclonal secretory immunoglobulin A protects mice against oral challenge with the invasive pathogen Salmonella typhimurium. Infect Immun 1992; 60:1786.
  27. Dasgupta S, Das S, Biswas A, et al. Small alarmones (p)ppGpp regulate virulence associated traits and pathogenesis of Salmonella enterica serovar Typhi. Cell Microbiol 2019; 21:e13034.
  28. Ledeboer NA, Frye JG, McClelland M, Jones BD. Salmonella enterica serovar Typhimurium requires the Lpf, Pef, and Tafi fimbriae for biofilm formation on HEp-2 tissue culture cells and chicken intestinal epithelium. Infect Immun 2006; 74:3156.
  29. Weening EH, Barker JD, Laarakker MC, et al. The Salmonella enterica serotype Typhimurium lpf, bcf, stb, stc, std, and sth fimbrial operons are required for intestinal persistence in mice. Infect Immun 2005; 73:3358.
  30. Cellular Microbiology, Cossart P, Boquet P, Normark S, Rappuoli R (Eds), ASM Press, Washington, DC 2000.
  31. Kohbata S, Yokoyama H, Yabuuchi E. Cytopathogenic effect of Salmonella typhi GIFU 10007 on M cells of murine ileal Peyer's patches in ligated ileal loops: an ultrastructural study. Microbiol Immunol 1986; 30:1225.
  32. Hughes EA, Galán JE. Immune response to Salmonella: location, location, location? Immunity 2002; 16:325.
  33. Pace J, Hayman MJ, Galán JE. Signal transduction and invasion of epithelial cells by S. typhimurium. Cell 1993; 72:505.
  34. Finlay BB, Gumbiner B, Falkow S. Penetration of Salmonella through a polarized Madin-Darby canine kidney epithelial cell monolayer. J Cell Biol 1988; 107:221.
  35. Halle S, Bumann D, Herbrand H, et al. Solitary intestinal lymphoid tissue provides a productive port of entry for Salmonella enterica serovar Typhimurium. Infect Immun 2007; 75:1577.
  36. Steele-Mortimer O. The Salmonella-containing vacuole: moving with the times. Curr Opin Microbiol 2008; 11:38.
  37. Zhang H, Zoued A, Liu X, et al. Type I interferon remodels lysosome function and modifies intestinal epithelial defense. Proc Natl Acad Sci U S A 2020; 117:29862.
  38. Kingsley RA, Humphries AD, Weening EH, et al. Molecular and phenotypic analysis of the CS54 island of Salmonella enterica serotype typhimurium: identification of intestinal colonization and persistence determinants. Infect Immun 2003; 71:629.
  39. Hueck CJ, Hantman MJ, Bajaj V, et al. Salmonella typhimurium secreted invasion determinants are homologous to Shigella Ipa proteins. Mol Microbiol 1995; 18:479.
  40. Galán JE. Molecular genetic bases of Salmonella entry into host cells. Mol Microbiol 1996; 20:263.
  41. Lee CA, Jones BD, Falkow S. Identification of a Salmonella typhimurium invasion locus by selection for hyperinvasive mutants. Proc Natl Acad Sci U S A 1992; 89:1847.
  42. Behlau I, Miller SI. A PhoP-repressed gene promotes Salmonella typhimurium invasion of epithelial cells. J Bacteriol 1993; 175:4475.
  43. Ochman H, Soncini FC, Solomon F, Groisman EA. Identification of a pathogenicity island required for Salmonella survival in host cells. Proc Natl Acad Sci U S A 1996; 93:7800.
  44. Shea JE, Hensel M, Gleeson C, Holden DW. Identification of a virulence locus encoding a second type III secretion system in Salmonella typhimurium. Proc Natl Acad Sci U S A 1996; 93:2593.
  45. Hensel M, Shea JE, Waterman SR, et al. Genes encoding putative effector proteins of the type III secretion system of Salmonella pathogenicity island 2 are required for bacterial virulence and proliferation in macrophages. Mol Microbiol 1998; 30:163.
  46. Watson PR, Paulin SM, Bland AP, et al. Characterization of intestinal invasion by Salmonella typhimurium and Salmonella dublin and effect of a mutation in the invH gene. Infect Immun 1995; 63:2743.
  47. Galyov EE, Wood MW, Rosqvist R, et al. A secreted effector protein of Salmonella dublin is translocated into eukaryotic cells and mediates inflammation and fluid secretion in infected ileal mucosa. Mol Microbiol 1997; 25:903.
  48. Francis CL, Ryan TA, Jones BD, et al. Ruffles induced by Salmonella and other stimuli direct macropinocytosis of bacteria. Nature 1993; 364:639.
  49. Hannemann S, Galán JE. Salmonella enterica serovar-specific transcriptional reprogramming of infected cells. PLoS Pathog 2017; 13:e1006532.
  50. Ginocchio CC, Rahn K, Clarke RC, Galán JE. Naturally occurring deletions in the centisome 63 pathogenicity island of environmental isolates of Salmonella spp. Infect Immun 1997; 65:1267.
  51. McCormick BA, Colgan SP, Delp-Archer C, et al. Salmonella typhimurium attachment to human intestinal epithelial monolayers: transcellular signalling to subepithelial neutrophils. J Cell Biol 1993; 123:895.
  52. Tükel C, Raffatellu M, Chessa D, et al. Neutrophil influx during non-typhoidal salmonellosis: who is in the driver's seat? FEMS Immunol Med Microbiol 2006; 46:320.
  53. McCormick BA, Miller SI, Carnes D, Madara JL. Transepithelial signaling to neutrophils by salmonellae: a novel virulence mechanism for gastroenteritis. Infect Immun 1995; 63:2302.
  54. Jung HC, Eckmann L, Yang SK, et al. A distinct array of proinflammatory cytokines is expressed in human colon epithelial cells in response to bacterial invasion. J Clin Invest 1995; 95:55.
  55. Miao EA, Alpuche-Aranda CM, Dors M, et al. Cytoplasmic flagellin activates caspase-1 and secretion of interleukin 1beta via Ipaf. Nat Immunol 2006; 7:569.
  56. Sansonetti PJ, Phalipon A, Arondel J, et al. Caspase-1 activation of IL-1beta and IL-18 are essential for Shigella flexneri-induced inflammation. Immunity 2000; 12:581.
  57. Tsuji NM, Tsutsui H, Seki E, et al. Roles of caspase-1 in Listeria infection in mice. Int Immunol 2004; 16:335.
  58. Mariathasan S, Weiss DS, Dixit VM, Monack DM. Innate immunity against Francisella tularensis is dependent on the ASC/caspase-1 axis. J Exp Med 2005; 202:1043.
  59. Lara-Tejero M, Sutterwala FS, Ogura Y, et al. Role of the caspase-1 inflammasome in Salmonella typhimurium pathogenesis. J Exp Med 2006; 203:1407.
  60. Finlay BB, Falkow S. Virulence factors associated with Salmonella species. Microbiol Sci 1988; 5:324.
  61. Rahman MM, Guard-Petter J, Carlson RW. A virulent isolate of Salmonella enteritidis produces a Salmonella typhi-like lipopolysaccharide. J Bacteriol 1997; 179:2126.
  62. Kawasaki K, Ernst RK, Miller SI. 3-O-deacylation of lipid A by PagL, a PhoP/PhoQ-regulated deacylase of Salmonella typhimurium, modulates signaling through Toll-like receptor 4. J Biol Chem 2004; 279:20044.
  63. Khan SA, Everest P, Servos S, et al. A lethal role for lipid A in Salmonella infections. Mol Microbiol 1998; 29:571.
  64. Guo L, Lim KB, Gunn JS, et al. Regulation of lipid A modifications by Salmonella typhimurium virulence genes phoP-phoQ. Science 1997; 276:250.
  65. Bader MW, Sanowar S, Daley ME, et al. Recognition of antimicrobial peptides by a bacterial sensor kinase. Cell 2005; 122:461.
  66. Nishino K, Latifi T, Groisman EA. Virulence and drug resistance roles of multidrug efflux systems of Salmonella enterica serovar Typhimurium. Mol Microbiol 2006; 59:126.
  67. Aguero J, Faundez G, Nunez M, et al. Choleriform syndrome and production of labile enterotoxin (CT/LT1)-like antigen by species of Salmonella infantis and Salmonella haardt isolated from the same patient. Rev Infect Dis 1991; 13:420.
  68. Chopra AK, Houston CW, Peterson JW, et al. Cloning and expression of the Salmonella enterotoxin gene. J Bacteriol 1987; 169:5095.
  69. Chopra AK, Huang JH, Xu X, et al. Role of Salmonella enterotoxin in overall virulence of the organism. Microb Pathog 1999; 27:155.
  70. Rahman H, Tschape H. Induction of Salmonella enterotoxin (stn) gene expression by epithelial cells (IEC-6). Indian J Exp Biol 1999; 37:1101.
  71. Lee S, Yang YA, Milano SK, et al. Salmonella Typhoid Toxin PltB Subunit and Its Non-typhoidal Salmonella Ortholog Confer Differential Host Adaptation and Virulence. Cell Host Microbe 2020; 27:937.
  72. García-del Portillo F. Salmonella intracellular proliferation: where, when and how? Microbes Infect 2001; 3:1305.
  73. Yeom J, Shao Y, Groisman EA. Small proteins regulate Salmonella survival inside macrophages by controlling degradation of a magnesium transporter. Proc Natl Acad Sci U S A 2020; 117:20235.
  74. Chen M, Sun H, Boot M, et al. Itaconate is an effector of a Rab GTPase cell-autonomous host defense pathway against Salmonella. Science 2020; 369:450.
  75. Rosenberg G, Yehezkel D, Hoffman D, et al. Host succinate is an activation signal for Salmonella virulence during intracellular infection. Science 2021; 371:400.
  76. Alpuche-Aranda CM, Racoosin EL, Swanson JA, Miller SI. Salmonella stimulate macrophage macropinocytosis and persist within spacious phagosomes. J Exp Med 1994; 179:601.
  77. Miller SI, Kukral AM, Mekalanos JJ. A two-component regulatory system (phoP phoQ) controls Salmonella typhimurium virulence. Proc Natl Acad Sci U S A 1989; 86:5054.
  78. Groisman EA, Chiao E, Lipps CJ, Heffron F. Salmonella typhimurium phoP virulence gene is a transcriptional regulator. Proc Natl Acad Sci U S A 1989; 86:7077.
  79. Angelakopoulos H, Hohmann EL. Pilot study of phoP/phoQ-deleted Salmonella enterica serovar typhimurium expressing Helicobacter pylori urease in adult volunteers. Infect Immun 2000; 68:2135.
  80. Fields PI, Swanson RV, Haidaris CG, Heffron F. Mutants of Salmonella typhimurium that cannot survive within the macrophage are avirulent. Proc Natl Acad Sci U S A 1986; 83:5189.
  81. Guiney DG, Fang FC, Krause M, et al. Biology and clinical significance of virulence plasmids in Salmonella serovars. Clin Infect Dis 1995; 21 Suppl 2:S146.
  82. Libby SJ, Adams LG, Ficht TA, et al. The spv genes on the Salmonella dublin virulence plasmid are required for severe enteritis and systemic infection in the natural host. Infect Immun 1997; 65:1786.
  83. Ahmer BM, Tran M, Heffron F. The virulence plasmid of Salmonella typhimurium is self-transmissible. J Bacteriol 1999; 181:1364.
  84. Chiu CH, Chu C, Ou JT. Lack of evidence of an association between the carriage of virulence plasmid and the bacteremia of Salmonella typhimurium in humans. Microbiol Immunol 2000; 44:741.
  85. Chang CC, Ou JT. Excess production of interleukin-12 subunit p40 stimulated by the virulence plasmid of Salmonella enterica serovar Typhimurium in the early phase of infection in the mouse. Microb Pathog 2002; 32:15.
  86. Conlan JW. Neutrophils prevent extracellular colonization of the liver microvasculature by Salmonella typhimurium. Infect Immun 1996; 64:1043.
  87. Anand AJ, Glatt AE. Salmonella osteomyelitis and arthritis in sickle cell disease. Semin Arthritis Rheum 1994; 24:211.
  88. Mabey DC, Brown A, Greenwood BM. Plasmodium falciparum malaria and Salmonella infections in Gambian children. J Infect Dis 1987; 155:1319.
  89. Gopinath R, Keystone JS, Kain KC. Concurrent falciparum malaria and Salmonella bacteremia in travelers: report of two cases. Clin Infect Dis 1995; 20:706.
  90. Gendrel D, Kombila M, Beaudoin-Leblevec G, Richard-Lenoble D. Nontyphoidal salmonellal septicemia in Gabonese children infected with Schistosoma intercalatum. Clin Infect Dis 1994; 18:103.
  91. Wheat LJ, Rubin RH, Harris NL, et al. Systemic salmonellosis in patients with disseminated histoplasmosis. Case for 'macrophage blockade' caused by Histoplasma capsulatum. Arch Intern Med 1987; 147:561.
  92. Miao EA, Andersen-Nissen E, Warren SE, Aderem A. TLR5 and Ipaf: dual sensors of bacterial flagellin in the innate immune system. Semin Immunopathol 2007; 29:275.
  93. Ramos JM, García-Corbeira P, Aguado JM, et al. Clinical significance of primary vs. secondary bacteremia due to nontyphoid Salmonella in patients without AIDS. Clin Infect Dis 1994; 19:777.
  94. Gruenewald R, Blum S, Chan J. Relationship between human immunodeficiency virus infection and salmonellosis in 20- to 59-year-old residents of New York City. Clin Infect Dis 1994; 18:358.
  95. Levine WC, Buehler JW, Bean NH, Tauxe RV. Epidemiology of nontyphoidal Salmonella bacteremia during the human immunodeficiency virus epidemic. J Infect Dis 1991; 164:81.
  96. Thamlikitkul V, Dhiraputra C, Paisarnsinsup T, Chareandee C. Non-typhoidal Salmonella bacteraemia: clinical features and risk factors. Trop Med Int Health 1996; 1:443.
  97. Wolfe MS, Louria DB, Armstrong D, Blevins A. Salmonellosis in patients with neoplastic disease. A review of 100 episodes at Memorial Cancer Center over a 13-year period. Arch Intern Med 1971; 128:546.
  98. Tumbarello M, Tacconelli E, Caponera S, et al. The impact of bacteraemia on HIV infection. Nine years experience in a large Italian university hospital. J Infect 1995; 31:123.
  99. Mastroeni P, Simmons C, Fowler R, et al. Igh-6(-/-) (B-cell-deficient) mice fail to mount solid acquired resistance to oral challenge with virulent Salmonella enterica serovar typhimurium and show impaired Th1 T-cell responses to Salmonella antigens. Infect Immun 2000; 68:46.
  100. Forrest BD. Impairment of immunogenicity of Salmonella typhi Ty21a due to preexisting cross-reacting intestinal antibodies. J Infect Dis 1992; 166:210.
  101. MacLennan CA, Gilchrist JJ, Gordon MA, et al. Dysregulated humoral immunity to nontyphoidal Salmonella in HIV-infected African adults. Science 2010; 328:508.
  102. Endt K, Stecher B, Chaffron S, et al. The microbiota mediates pathogen clearance from the gut lumen after non-typhoidal Salmonella diarrhea. PLoS Pathog 2010; 6:e1001097.
  103. Thiemann S, Smit N, Roy U, et al. Enhancement of IFNγ Production by Distinct Commensals Ameliorates Salmonella-Induced Disease. Cell Host Microbe 2017; 21:682.
  104. Eloe-Fadrosh EA, McArthur MA, Seekatz AM, et al. Impact of oral typhoid vaccination on the human gut microbiota and correlations with s. Typhi-specific immunological responses. PLoS One 2013; 8:e62026.
  105. Claudi B, Spröte P, Chirkova A, et al. Phenotypic variation of Salmonella in host tissues delays eradication by antimicrobial chemotherapy. Cell 2014; 158:722.
  106. Helaine S, Cheverton AM, Watson KG, et al. Internalization of Salmonella by macrophages induces formation of nonreplicating persisters. Science 2014; 343:204.
  107. Neiger MR, González JF, Gonzalez-Escobedo G, et al. Pathoadaptive Alteration of Salmonella Biofilm Formation in Response to the Gallbladder Environment. J Bacteriol 2019; 201.
Topic 2700 Version 25.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟