Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Feb 1.
Published in final edited form as: FEMS Immunol Med Microbiol. 2011 Dec 19;64(1):13–20. doi: 10.1111/j.1574-695X.2011.00909.x

The biological basis of severe outcomes in Anaplasma phagocytophilum infection

J Stephen Dumler 1,*
PMCID: PMC3256277  NIHMSID: NIHMS339756  PMID: 22098465

Abstract

Anaplasma phagocytophilum causes granulocytic anaplasmosis, an acute disease in humans that is also often subclinical. However, 36% are hospitalized, 7% need intensive care, and the case fatality rate is 0.6%. The biological basis for severe disease is not understood. Despite A. phagocytophilum’s mechanisms to subvert neutrophil antimicrobial responses, whether these mechanisms lead to disease is unclear. In animals, inflammatory lesions track with IFNγ and IL-10 expression, and infection of Ifng −/− mice leads to increased pathogen load but inhibition of inflammation. Suppression of STAT signaling in horses impacts IL-10 and IFN-γ expression, and also suppresses disease severity. Similar inhibition of inflammation with infection of NKT-deficient mice suggests that innate immune responses are key for disease. With severe disease, tissues can demonstrate hemophagocytosis, and measures of macrophage activation/hemophagocytic syndromes (MAS/HPS) support the concept of HGA as an immunopathologic disease. MAS/HPS are related to defective cytotoxic lymphocytes that ordinarily diminish inflammation. Pilot studies in mice show cytotoxic lymphocyte activation with A. phagocytophilum infection, yet suppression of cytotoxic responses from both NKT and CD8 cells, consistent with the development of MAS/HPS. Whether severity relates to microbial factors or genetically-determined diversity in human immune and inflammatory response needs more investigation.

Keywords: Anaplasma, rickettsia, immunopathology, hemophagocytic syndrome, macrophage activation syndrome, cytotoxic lymphocyte

Introduction

Human granulocytic anaplasmosis (HGA) is a tick-transmitted zoonosis caused by the intraneutrophilic rickettsia, Anaplasma phagocytophilum (Dumler, et al., 2007, Bakken & Dumler, 2008). The infection occurs broadly across mammalian species, but disease manifestations are in part species-dependent, with humans, horses, and dogs recognized to have mild to severe clinical signs and/or symptoms with infection. Small mammals, including laboratory mice, can sustain infection without clinical signs, yet develop inflammatory histopathologic lesions nearly identical to those observed in humans, horses, and dogs, underscoring their utility as models to study disease processes (Bunnell, et al., 1999, Lepidi, et al., 2000, Choi, et al., 2007).

In humans, HGA is now the 3rd most common vector-borne infection in the U.S. and Europe, and is increasingly recognized as an important vector-borne infection in Asia, especially in China (Zhang, et al., 2008, Zhang, et al., 2008). Among cross-sectional serosurveys conducted in North America, Europe and Asia, 3.7% have A. phagocytophilum antibodies, and that proportion increases with activities predisposing to or predicting tick exposures and bites (Dumler, et al., 2005, Dumler, et al., 2007). However, in areas with high seroprevalences, such as in the northeast and Upper Midwest USA, active surveillance identifies far fewer clinically-evident infections (Bakken, et al., 1996, IJdo, et al., 2000). Features of human disease include fever, myalgia, headache, malaise, thrombocytopenia, leukopenia, anemia, and mild to moderate hepatic injury sufficient to result in increased aspartate and alanine aminotransferase activities in serum (Dumler, et al., 2007, Dumler, et al., 2007, Bakken & Dumler, 2008). These undifferentiated clinical features result in a very broad differential diagnosis and make final definitive diagnosis of HGA difficult. This is confounded by the diversity of disease presentations in humans; while many patients have mild clinical signs and symptoms, infection results in hospitalization for 36%, ICU admission in 7%, and death in 0.6% of cases (Bakken, et al., 1994, Bakken, et al., 1998, Bakken & Dumler, 2000, Chapman, et al., 2006). Severe complications include septic shock-like syndromes, acute respiratory distress syndrome, immune compromise and opportunistic infections, among others (Dumler, et al., 2007).

A. phagocytophilum subversion and disease pathogenesis

The underlying pathogenetic events that lead to disease manifestations are not well understood for A. phagocytophilum. Early investigations linked disease severity with pathogen load, and disease manifestations are often easily reversed with specific antimicrobial treatment (Bakken, et al., 1994, Bakken & Dumler, 2006, Wormser, et al., 2006, Dumler, et al., 2007, Bakken & Dumler, 2008). However, these observations were contradicted in subsequent studies where the degree of disease and pathologic injury did not correlate well with bacterial burden, and pathologic findings suggested the potential for a cytokine- or immune-mediated process (Walker & Dumler, 1995, Walker & Dumler, 1996, Jahangir, et al., 1998, Bunnell, et al., 1999, Lepidi, et al., 2000). For example, the hallmark of infection that plausibly reflects direct pathogen-mediated cytolytic injury is leukopenia, especially neutropenia, since A. phagocytophilum infects almost exclusively neutrophils in vivo. However, among infected patients, the mean infected leukocyte count is 0.3 × 109 cells/L, whereas the mean total leukocyte count for HGA patients is 3.7 × 109 cells/L. Since the mean leukocyte count with infection is approximately 50% of that for uninfected patients, 4.1 × 109 cells/L, or 15-fold more leukocytes are lost than can be accounted for by direct A. phagocytophilum lysis alone (Bakken, et al., 2001, Dumler, et al., 2007). Thus, another explanation for the hematologic derangements is needed.

Considerable investigation regarding how A. phagocytophilum subverts neutrophil function to survive within this key host defense cell has been conducted (Carlyon & Fikrig, 2006, Rikihisa, 2011). However, enhanced intracellular survival or increased pathogen load does not necessarily belie clinical signs or disease. A. phagocytophilum uses several strategies (Table 1) that conceivably could also contribute to disease pathogenesis: 1) its ability to alter neutrophil mobilization of phagocyte oxidase by disorganized assembly, superoxide dismutase activity, and reduced transcription of key phagocyte oxidase genes such as CYBB and RAC1, appear to be involved (Choi & Dumler, 2003, Carlyon, et al., 2004, IJdo & Mueller, 2004); 2) its ability to induce upregulated chemokine expression and neutrophil vesicle discharge leads to the recruitment of new neutrophils hosts to support pathogen population expansion (Klein, et al., 2000, Akkoyunlu, et al., 2001, Scorpio, et al., 2004), whereas inhibition of this process by blockade of the key chemokine IL-8 receptor (CXCR1) or pharmacologic alteration of IL-10:IFN-γ balance by dexamethasone, leads to reduced pathogen numbers in tissues, but does not appear to alter or reduce host pathologic responses (Scorpio, et al., 2004, Davies, et al., in press); 3) its ability to introduce microbial effectors, such as AnkA, into the neutrophil via a type IV secretion system, and to alter local signaling (ROCK1 and Syk) that facilitates A. phagocytophilum endocytic entry and over the course of infection, leads at minimum to reduced CYBB transcription and reduced microbial killing in the infected cell (Lin, et al., 2007, Thomas & Fikrig, 2007, Garcia-Garcia, et al., 2009, Garcia-Garcia, et al., 2009); and 4) its ability to upregulate cellular histone deacetylase transcription and activity that likely promotes transcriptional program alterations associated with pathogen survival and population expansion (Garcia-Garcia, et al., 2009). While each of these has been shown to play a role in pathogen survival in vitro, and for some, in vivo, their role in disease pathogenesis is poorly understood. For example, although blockade of CXCR1 leads to reduced pathogen load, the degree of histopathologic injury in the murine model, which correlates strongly with disease manifestations in humans and horses, is not altered, and infection in a phagocyte oxidase deficient condition does not lead to increased pathogen load but actually ameliorates inflammation (Scorpio, et al., 2004, Scorpio, et al., 2006).

Table 1.

A. phagocytophilum strategies for intracellular survival and propagation, and their effect on histopathology and disease.

A. phagocytophilum-related pathogetic mechanism Target or effector molecule Effect on A. phagocytophilum loada Effect of gene deletion/pharmacologic/antibody inhibition on histopathologic lesionsb References
Phagocyte oxidase Disorganized assembly or transcriptional repression of host CYBB/RAC2 0 (in vivo); ↑↑↑ (in vitro) ↓↓ (phox −/− B6 mice in vivo) (Banerjee, et al., 2000, Banerjee, et al., 2000, Carlyon, et al., 2002, von Loewenich, et al., 2004, Scorpio, et al., 2006)
Chemokine/cytokine expression IL8, IL-10:IFNγ ↑↑ 0 (CXCR1 antibody-treated vs. control antibody-treated in vivo); ↓↓ (in dexamethasone-treated horses) (Akkoyunlu & Fikrig, 2000, Akkoyunlu, et al., 2001, Scorpio, et al., 2004, Davies, et al., in press)
A. phagocytophilum granulocyte entry A. phagocytophilum
AnkA
Host ROCK1 and Syk
↑↑ to ↑↑↑ ↓↓ to ↓↓↓ (vs. ROCK1 or Syk- silencing or vs. AnkA control antibody in vitro) (Lin, et al., 2007, Thomas & Fikrig, 2007)
Histone deacetylase (HDAC) HDAC1 and 2 ↑↑ (vs. HDAC-silenced cells or NaB or trichostatin inhibition in vitro) Not done (Garcia-Garcia, et al., 2009)
a

↑ - increased bacterial load or survival

b

↓ - decreased or “0” no change in histopathologic inflammation compared to unmodified control

Evidence for immunopathogenesis of disease with A. phagocytophilum

The immunologic responses to A. phagocytophilum include both humoral and cellular; however, immune control of A. phagocytophilum is still under investigation. A. phagocytophilum is cleared with kinetics that mimic rising antibody titers (Bakken, et al., 1996, Bakken, et al., 2002); yet there are clear examples of patients with high antibody titers, substantial bacteremia, and disease (Bakken, et al., 1994, Bakken, et al., 1996). While SCID mice cannot control infection, neither do they develop clinical signs, suggesting a dichotomous relationship between immune processes that protect and those that may cause injury (Hodzic, et al., 1998, Bunnell, et al., 1999). In contrast, A. phagocytophilum tissue and blood loads do not change with genetic deletion of iNOS (Nos2 −/−), TLR2 or MyD88, and only variably so for phagocyte oxidase (Cybb −/−) (Banerjee, et al., 2000, von Loewenich, et al., 2004). Yet, activation of these same innate immune pathways by infection, leads to hepatic inflammatory injury, in part dependent upon mouse strain genetic background (Scorpio, et al., 2006).

The cellular immune hallmark of A. phagocytophilum in humans, horses, and in mice is the early appearance of IFN-γ and IL-10 in serum or plasma (Dumler, et al., 2000, Martin, et al., 2000, Martin, et al., 2001, Scorpio, et al., 2008). While the proinflammatory cytokines TNF-α and IL-1β can be induced from peripheral blood leukocytes after infection ex vivo, these cytokines are generally not detected at high quantities in serum or plasma with active infection (Dumler, et al., 2000, Kim & Rikihisa, 2000, Dumler, et al., 2007). In the murine model of A. phagocytophilum infection, both IFN-γ and IL-10 are present in plasma during active infection, and appear as early as several hours post infection, peaking between days 2 and 7, typically considered an interval prior to adaptive immune response (Akkoyunlu & Fikrig, 2000, Martin, et al., 2001, Choi, et al., 2007, Pedra, et al., 2008). As anticipated, IFN-γ peaks shortly after maximum A. phagocytophilum bacteremia in mice; however, histopathologic injury, as reflected in hepatic inflammatory lesions, tracks more closely with IFN-γ levels than with bacteremia (Martin, et al., 2001). Among IFN-γ knockout mice, the level of bacteremia is 7–8 times higher than in wild type mice, yet IFN-γ knockout mice lack hepatic histopathology that is present in wild type mice. Interestingly, IL-10 knockout mice do not have altered bacteremia compared to wild type mice, yet develop significantly worse hepatic inflammatory lesions, providing key evidence for an immunopathologic role in the balance between Th1 and Th2 responses, or with relative roles of both IFN-γ and IL-10.

Given the clear importance of IFN-γ and IL-10 in histopathologic inflammatory lesions in the murine model of HGA, and their early appearance post infection, potentially important cellular sources include CD8 T lymphocytes, and the innate immune effector cells, NK and NKT cells (Choi, et al., 2007, Pedra, et al., 2008). Splenic CD8 T lymphocytes from infected mice become more activated than from mock-infected mice by day 4 post infection, when IFN-γ is peaking in plasma. However, the NK1.1-labeled splenic lymphocyte population, that includes both NK and NKT cells, is even more significantly activated with infection vs. mock-infection, peaking in near precise concordance with plasma IFN-γ levels. Infection in Jα18 −/− (Choi, et al., 2007) and CD1d −/− (unpublished data) mice that lack NKT cells leads to complete loss of hepatic inflammatory lesions on days 4 to 7 post infection, and antibody depletion of NK cells also suppresses hepatic inflammation at day 7 compared to infection in wild type or control antibody-treated mice. These results clearly indicate that innate immune induction of inflammatory lesions, which correspond to disease manifestations, is a key component of disease pathogenesis with HGA.

A role for cytotoxic lymphocytes in disease pathogenesis with A. phagocytophilum?

How do cytotoxic cells, like NK, NKT, and CD8 cells contribute to inflammatory disease manifestations with activation? The underpinning for IFN-γ driven effector response is two-fold: induction of proinflammatory cytokines and chemokines that activate local effector cells for phagocytosis and enhanced cellular killing and for recruitment of more effector cells; and production of effector molecules for microbial killing, including the oxygen-dependent production of superoxide radicals and hypochlorite and of nitric oxide and peroxynitrates (Ismail, et al., 2002, Mosser, 2003, Mantovani, et al., 2004, Lang, 2005, Browning, et al., 2006). When sequestered, these effector molecules and pathways are potent in their antimicrobial capacity; however, when excessive or exposed to the external host milieu, they can lead to “collateral” host inflammatory injury (Grom, 2004, Larroche & Mouthon, 2004, Janka & zur Stadt, 2005, Menasche, et al., 2005, Ravelli, et al., 2005).

The balance between microbial killing and host injury is a major determinant of disease outcome in many infections. One class of effector cells recruited and activated for many infectious agents, particularly intracellular infectious agents like viruses and intracellular bacteria is the cytotoxic lymphocyte. Cytotoxic lymphocytes have a potent capacity to both amplify the response by further release of IFN-γ, to directly kill cellular targets identified by specific cytotoxic cell-target cell interactions. This occurs as with CD8 T lymphocyte recognition of foreign peptides presented in the context of MHC class I molecules, or with NK lymphocyte recognition of altered HLA expression or TLR2 signaling, or with NKT cell recognition of specific glycolipids in the context of MHC class I CD1d (Scharton & Scott, 1993, Crowe, et al., 2003, Davis & Dustin, 2004, Godfrey & Kronenberg, 2004, Kronenberg, 2005, Mattner, et al., 2005, Voskoboinik, et al., 2006).

When the typical homeostatic mechanism of inflammatory dampening occurs after the inflammatory stimulus has been controlled and reduced in quantity, inflammatory signaling ceases and the underlying pathologic processes is altered to that of repair and reconstitution of function. However, significant and unremitting inflammatory injury occurs in macrophage activation and hemophagocytic syndromes (Francois, et al., 1997, Fisman, 2000, Grom, 2004, Larroche & Mouthon, 2004, Janka & zur Stadt, 2005, Menasche, et al., 2005, Behrens, et al., 2007, Arceci, 2008), genetic and acquired conditions that share the phenotype of i) active cytotoxic lymphocyte participation, ii) leukopenia, iii) thrombocytopenia, iv) anemia, and in hemophagocytic syndromes, v) the presence of hemophagocytic cells. Macrophage activation and hemophagocytic syndromes are believed to result from an underlying genetic or acquired defect in the ability of the cytotoxic lymphocyte, whether a CD8 T lymphocyte, an NK cell, or an NKT lymphocyte, to mobilize or deliver functional secretory granules that contain both perforin (which creates a membrane pore in the target cell) and granzyme (which enters via the perforin pore and causes DNA degradation, inducing apoptosis and death of the target cell) (Fisman, 2000, Grom, 2004, Larroche & Mouthon, 2004, Janka & zur Stadt, 2005, Menasche, et al., 2005, Arceci, 2008). Such genetic conditions include PRF1 (perforin) mutations, RAB27A mutations (secretory vesicle trafficking; Griscelli syndrome), LYST mutations (lysosomal trafficking regulator; Chediak-Higashi syndrome), and SH2D1A mutations (development of NKT cells; X-linked lymphoproliferative syndrome). When the target is an infected cell, the pathogen is killed. However, these cytotoxic cells do not directly recognize intact pathogen; rather pathogen molecular patterns are the ligands in these immunologic synapses (Davis & Dustin, 2004, Voskoboinik, et al., 2006). Thus, antigen presenting cells, whether MHCI- or CD1d-restricted, are also targets. This process is believed to be a normal homeostatic mechanism to contain unrestrained inflammatory activation. Thus, when cytotoxic cells are activated, they release the macrophage activating, proinflammatory IFN-γ that drives further inflammatory injury, but killing of antigen presenting cell targets initiates homeostatic control; with defective vesicular trafficking or inactive perforin mutants, the inability to deliver perforin and granzyme and to kill antigen presenting cells permits an unrestrained, amplifying proinflammatory response without benefit of cytolytic homeostatic resolution (Fisman, 2000, Grom, 2004, Larroche & Mouthon, 2004, Menasche, et al., 2005, Arceci, 2008). The result is a poorly controlled, in cases, relentless downward spiral of inflammatory injury and severe or fatal disease.

In fact, HGA patients have many features of macrophage activation, as evidenced by high levels of IFN-γ, IL-10, IL-12, and serum ferritin, as well as the key clinical features leukopenia, thrombocytopenia, anemia, and hepatic injury, and periodically including the detection of hemophagocytic cells in bone marrow, spleen, lymph node and liver, where examined (Lepidi, et al., 2000, Dumler, et al., 2007) (Figure 2). Moreover, HGA severity is correlated with the level of serum ferritin, serum IL-12, and the ratio of IL-10 to IFN-γ (Dumler, et al., 2007). The latter feature is highly similar to observations in the murine model (Martin, et al., 2001, Choi & Dumler, 2007). Since mice do not develop disease, the horse model, where clinical disease is strikingly similar to that in humans, is studied. Here, infected horses are studied after the IFN-γ:IL-10 axis is pharmacologically manipulated by dexamethasone treatment that alters STAT3 and STAT1 signaling to reduce propagation of effector signaling – functionally polarizing the response to favor anti-inflammatory IL-10 domination. As anticipated, horses treated with dexamethasone had increased IL-10:IFN-γ and IL-10:IL-12 ratios, higher overall bacteremias than untreated animals, but significantly less clinical signs and disease (Davies, et al., in press).

Figure 2.

Figure 2

Splenic NKT cells from A. phagocytophilum-infected mice (Ap/HL-60 cells) have abrogated cytotoxic responses, based on reduced delivery of vesicles (CD107a) to membranes after degranulation stimulated by ionomycin C, as compared with mock-infected (HL-60 cells) mice at days 7 and 10 post-infection. P values were calculated using the Wilcoxon test for nonparametric analysis, and a value <0.05 was considered significant.

It is reasonably understood why genetically-linked syndromes that result from specific mutations in key pathways important for secretory vacuole delivery or effector function result in a macrophage activation or hemophagocytic syndrome. However, why this occurs with acquired infections, especially those associated with bacteria is not well understood (Francois, et al., 1997, Fisman, 2000, Larroche & Mouthon, 2004, Janka & zur Stadt, 2005). To determine whether such a principle might apply to A. phagocytophilum, the murine model was studied using ex vivo splenic lymphocytes from infected and mock-infected animals. These cells were examined for their ability to discharge secretory vacuole contents (CD107a) when activated in vitro, a well-recognized correlate of cellular cytotoxicity. Ex vivo splenic NKT cells from infected animals stimulate for degranulation were significantly less cytotoxic than mock-infected controls at days 7 and 10, as were CD8 T lymphocytes at day 4 (Dumler, unpublished data). While neither NKT nor CD8 T lymphocytes are targets of A. phagocytophilum infection, these preliminary results suggest the basis for an acquired cytotoxicity defect with A. phagocytophilum infection that plausibly provides a tool to examine the periodic occurrence of severe inflammatory disease complications in HGA, and to investigate mechanisms of acquired cytotoxic lymphocyte function with non-viral infections.

Conclusions

In summary, HGA is an important emerging tick-borne infection that occurs world-wide. While most infections seem only moderate in severity and are easily treated with doxycycline, a minority will have very significant and debilitating disease, despite otherwise adequate antimicrobial treatment. The inflammatory basis for disease is most likely a result of immunopathologic injury after IFN-γ activation of effector cells not adequately tempered by the dampening effects of IL-10 (Martin, et al., 2001, Dumler, et al., 2007). A subset of patients that does poorly owing to serious inflammatory injury, could occur because of a higher degree of polarization in this IL-10:IFN-γ axis, resulting in a macrophage activation-like syndrome, that if accompanied by defects in cytotoxic effector molecule delivery, whether genetically-predisposed or acquired via infection, could lead to the most polar of severe infection complications (Dumler, et al., 2007, Davies, et al., in press). How this happens, what ligands drive the inflammatory process, the relative role of innate and adaptive immunity, and how they can be controlled are now important questions to address (Choi & Dumler, 2007). The ability to toggle disease progression by targeting anti-inflammatory treatment during infection in horses could provide additional guidance to help for those severely affected (Scorpio, et al., 2006, Davies, et al., in press). Moreover, the dissection of the fundamental biological processes that drive this syndrome with HGA at the level of cytotoxic lymphocyte dysfunction, offers a new possibility to study the pathogenesis of acquired non-viral hemophagocytic syndromes. Once again, obligate intracellular bacteria have lessons to teach, and as good students, we should pay careful attention.

Figure 1.

Figure 1

Splenic histopathology from a patient who died after HGA. Note the hemo- and leukophagocytic cells denoted by the white arrows. Hematoxylin & eosin stain; original magnification x 400.

Acknowledgments

This work was supported in part by grant R01 AI44102 from the National Institutes of Allergy and Infectious Diseases. Thanks to many colleagues including Johan S. Bakken, M.D., Diana G. Scorpio, DVM MPH, Kyoung Seong Choi, PhD DVM, and John E. Madigan, DVM for long help and support.

Literature Cited

  1. Akkoyunlu M, Fikrig E. Gamma interferon dominates the murine cytokine response to the agent of human granulocytic ehrlichiosis and helps to control the degree of early rickettsemia. Infect Immun. 2000;68:1827–1833. doi: 10.1128/iai.68.4.1827-1833.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Akkoyunlu M, Malawista SE, Anguita J, Fikrig E. Exploitation of interleukin-8-induced neutrophil chemotaxis by the agent of human granulocytic ehrlichiosis. Infect Immun. 2001;69:5577–5588. doi: 10.1128/IAI.69.9.5577-5588.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Arceci RJ. When T cells and macrophages do not talk: the hemophagocytic syndromes. Curr Opin Hematol. 2008;15:359–367. doi: 10.1097/MOH.0b013e3282f97f88. [DOI] [PubMed] [Google Scholar]
  4. Bakken JS, Dumler JS. Human granulocytic ehrlichiosis. Clin Infect Dis. 2000;31:554–560. doi: 10.1086/313948. [DOI] [PubMed] [Google Scholar]
  5. Bakken JS, Dumler JS. Clinical diagnosis and treatment of human granulocytotropic anaplasmosis. Ann N Y Acad Sci. 2006;1078:236–247. doi: 10.1196/annals.1374.042. [DOI] [PubMed] [Google Scholar]
  6. Bakken JS, Dumler S. Human granulocytic anaplasmosis. Infect Dis Clin North Am. 2008;22:433–448. viii. doi: 10.1016/j.idc.2008.03.011. [DOI] [PubMed] [Google Scholar]
  7. Bakken JS, Haller I, Riddell D, Walls JJ, Dumler JS. The serological response of patients infected with the agent of human granulocytic ehrlichiosis. Clin Infect Dis. 2002;34:22–27. doi: 10.1086/323811. [DOI] [PubMed] [Google Scholar]
  8. Bakken JS, Dumler JS, Chen SM, Eckman MR, Van Etta LL, Walker DH. Human granulocytic ehrlichiosis in the upper Midwest United States. A new species emerging? JAMA. 1994;272:212–218. [PubMed] [Google Scholar]
  9. Bakken JS, Krueth J, Wilson-Nordskog C, Tilden RL, Asanovich K, Dumler JS. Clinical and laboratory characteristics of human granulocytic ehrlichiosis. JAMA. 1996;275:199–205. [PubMed] [Google Scholar]
  10. Bakken JS, Aguero-Rosenfeld ME, Tilden RL, et al. Serial measurements of hematologic counts during the active phase of human granulocytic ehrlichiosis. Clin Infect Dis. 2001;32:862–870. doi: 10.1086/319350. [DOI] [PubMed] [Google Scholar]
  11. Bakken JS, Goellner P, Van Etten M, et al. Seroprevalence of human granulocytic ehrlichiosis among permanent residents of northwestern Wisconsin. Clin Infect Dis. 1998;27:1491–1496. doi: 10.1086/515048. [DOI] [PubMed] [Google Scholar]
  12. Banerjee R, Anguita J, Fikrig E. Granulocytic ehrlichiosis in mice deficient in phagocyte oxidase or inducible nitric oxide synthase. Infect Immun. 2000;68:4361–4362. doi: 10.1128/iai.68.7.4361-4362.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Banerjee R, Anguita J, Roos D, Fikrig E. Cutting edge: infection by the agent of human granulocytic ehrlichiosis prevents the respiratory burst by down-regulating gp91phox. J Immunol. 2000;164:3946–3949. doi: 10.4049/jimmunol.164.8.3946. [DOI] [PubMed] [Google Scholar]
  14. Behrens EM, Beukelman T, Paessler M, Cron RQ. Occult macrophage activation syndrome in patients with systemic juvenile idiopathic arthritis. J Rheumatol. 2007;34:1133–1138. [PubMed] [Google Scholar]
  15. Browning MD, Garyu JW, Dumler JS, Scorpio DG. Role of reactive nitrogen species in development of hepatic injury in a C57BL/6 mouse model of human granulocytic anaplasmosis. Comp Med. 2006;56:55–62. [PubMed] [Google Scholar]
  16. Bunnell JE, Trigiani ER, Srinivas SR, Dumler JS. Development and distribution of pathologic lesions are related to immune status and tissue deposition of human granulocytic ehrlichiosis agent-infected cells in a murine model system. J Infect Dis. 1999;180:546–550. doi: 10.1086/314902. [DOI] [PubMed] [Google Scholar]
  17. Carlyon JA, Fikrig E. Mechanisms of evasion of neutrophil killing by Anaplasma phagocytophilum. Curr Opin Hematol. 2006;13:28–33. doi: 10.1097/01.moh.0000190109.00532.56. [DOI] [PubMed] [Google Scholar]
  18. Carlyon JA, Chan WT, Galan J, Roos D, Fikrig E. Repression of rac2 mRNA expression by Anaplasma phagocytophila is essential to the inhibition of superoxide production and bacterial proliferation. J Immunol. 2002;169:7009–7018. doi: 10.4049/jimmunol.169.12.7009. [DOI] [PubMed] [Google Scholar]
  19. Carlyon JA, Abdel-Latif D, Pypaert M, Lacy P, Fikrig E. Anaplasma phagocytophilum utilizes multiple host evasion mechanisms to thwart NADPH oxidase-mediated killing during neutrophil infection. Infect Immun. 2004;72:4772–4783. doi: 10.1128/IAI.72.8.4772-4783.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Chapman AS, Bakken JS, Folk SM, et al. Diagnosis and management of tickborne rickettsial diseases: Rocky Mountain spotted fever, ehrlichioses, and anaplasmosis--United States: a practical guide for physicians and other health-care and public health professionals. MMWR Recomm Rep. 2006;55:1–27. [PubMed] [Google Scholar]
  21. Choi KS, Dumler JS. Early induction and late abrogation of respiratory burst in A. phagocytophilum-infected neutrophils. Ann N Y Acad Sci. 2003;990:488–493. doi: 10.1111/j.1749-6632.2003.tb07415.x. [DOI] [PubMed] [Google Scholar]
  22. Choi KS, Dumler JS. Mitogenic component in polar lipid-enriched Anaplasma phagocytophilum membranes. Clin Vaccine Immunol. 2007;14:1260–1265. doi: 10.1128/CVI.00204-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Choi KS, Scorpio DG, Barat NC, Dumler JS. Msp2 variation in Anaplasma phagocytophilum in vivo does not stimulate T cell immune responses or interferon-gamma production. FEMS Immunol Med Microbiol. 2007:374–386. doi: 10.1111/j.1574-695X.2007.00214.x. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  24. Choi KS, Webb T, Oelke M, Scorpio DG, Dumler JS. Differential innate immune cell activation and proinflammatory response in Anaplasma phagocytophilum infection. Infect Immun. 2007;75:3124–3130. doi: 10.1128/IAI.00098-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Crowe NY, Uldrich AP, Kyparissoudis K, et al. Glycolipid antigen drives rapid expansion and sustained cytokine production by NK T cells. J Immunol. 2003;171:4020–4027. doi: 10.4049/jimmunol.171.8.4020. [DOI] [PubMed] [Google Scholar]
  26. Davies RS, Madigan JE, Hodzic E, Borjesson DL, Dumler JS. Dexamethasone-induced cytokine changes associated with diminished disease severity in horses infected with Anaplasma phagocytophilum. Clin Vaccine Immunol. doi: 10.1128/CVI.05034-11. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Davis DM, Dustin ML. What is the importance of the immunological synapse? Trends Immunol. 2004;25:323–327. doi: 10.1016/j.it.2004.03.007. [DOI] [PubMed] [Google Scholar]
  28. Dumler JS, Barat NC, Barat CE, Bakken JS. Human granulocytic anaplasmosis and macrophage activation. Clin Infect Dis. 2007;45:199–204. doi: 10.1086/518834. [DOI] [PubMed] [Google Scholar]
  29. Dumler JS, Madigan JE, Pusterla N, Bakken JS. Ehrlichioses in humans: epidemiology, clinical presentation, diagnosis, and treatment. Clin Infect Dis. 2007;45(Suppl 1):S45–51. doi: 10.1086/518146. [DOI] [PubMed] [Google Scholar]
  30. Dumler JS, Trigiani ER, Bakken JS, Aguero-Rosenfeld ME, Wormser GP. Serum cytokine responses during acute human granulocytic ehrlichiosis. Clin Diagn Lab Immunol. 2000;7:6–8. doi: 10.1128/cdli.7.1.6-8.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Dumler JS, Choi KS, Garcia-Garcia JC, et al. Human granulocytic anaplasmosis and. Anaplasma phagocytophilum Emerg Infect Dis. 2005;11:1828–1834. doi: 10.3201/eid1112.050898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Fisman DN. Hemophagocytic syndromes and infection. Emerg Infect Dis. 2000;6:601–608. doi: 10.3201/eid0606.000608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Francois B, Trimoreau F, Vignon P, Fixe P, Praloran V, Gastinne H. Thrombocytopenia in the sepsis syndrome: role of hemophagocytosis and macrophage colony-stimulating factor. Am J Med. 1997;103:114–120. doi: 10.1016/s0002-9343(97)00136-8. [DOI] [PubMed] [Google Scholar]
  34. Garcia-Garcia JC, Barat NC, Trembley SJ, Dumler JS. Epigenetic silencing of host cell defense genes enhances intracellular survival of the rickettsial pathogen Anaplasma phagocytophilum. PLoS Pathog. 2009;5:e1000488. doi: 10.1371/journal.ppat.1000488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Garcia-Garcia JC, Rennoll-Bankert KE, Pelly S, Milstone AM, Dumler JS. Silencing of host cell CYBB gene expression by the nuclear effector AnkA of the intracellular pathogen Anaplasma phagocytophilum. Infect Immun. 2009;77:2385–2391. doi: 10.1128/IAI.00023-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Godfrey DI, Kronenberg M. Going both ways: immune regulation via CD1d-dependent NKT cells. J Clin Invest. 2004;114:1379–1388. doi: 10.1172/JCI23594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Grom AA. Natural killer cell dysfunction: A common pathway in systemic-onset juvenile rheumatoid arthritis, macrophage activation syndrome, and hemophagocytic lymphohistiocytosis? Arthritis Rheum. 2004;50:689–698. doi: 10.1002/art.20198. [DOI] [PubMed] [Google Scholar]
  38. Hodzic E, Ijdo JW, Feng S, et al. Granulocytic ehrlichiosis in the laboratory mouse. J Infect Dis. 1998;177:737–745. doi: 10.1086/514236. [DOI] [PubMed] [Google Scholar]
  39. IJdo JW, Mueller AC. Neutrophil NADPH oxidase is reduced at the Anaplasma phagocytophilum phagosome. Infect Immun. 2004;72:5392–5401. doi: 10.1128/IAI.72.9.5392-5401.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Jdo IJW, Meek JI, Cartter ML, et al. The emergence of another tickborne infection in the 12-town area around Lyme, Connecticut: human granulocytic ehrlichiosis. J Infect Dis. 2000;181:1388–1393. doi: 10.1086/315389. [DOI] [PubMed] [Google Scholar]
  41. Ismail N, Olano JP, Feng HM, Walker DH. Current status of immune mechanisms of killing of intracellular microorganisms. FEMS Microbiol Lett. 2002;207:111–120. doi: 10.1111/j.1574-6968.2002.tb11038.x. [DOI] [PubMed] [Google Scholar]
  42. Jahangir A, Kolbert C, Edwards W, Mitchell P, Dumler JS, Persing DH. Fatal pancarditis associated with human granulocytic Ehrlichiosis in a 44-year-old man. Clin Infect Dis. 1998;27:1424–1427. doi: 10.1086/515014. [DOI] [PubMed] [Google Scholar]
  43. Janka G, zur Stadt U. Familial and acquired hemophagocytic lymphohistiocytosis. Hematology. 2005;2005:82–88. doi: 10.1182/asheducation-2005.1.82. [DOI] [PubMed] [Google Scholar]
  44. Kim HY, Rikihisa Y. Expression of interleukin-1beta, tumor necrosis factor alpha, and interleukin-6 in human peripheral blood leukocytes exposed to human granulocytic ehrlichiosis agent or recombinant major surface protein P44. Infect Immun. 2000;68:3394–3402. doi: 10.1128/iai.68.6.3394-3402.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Klein MB, Hu S, Chao CC, Goodman JL. The agent of human granulocytic ehrlichiosis induces the production of myelosuppressing chemokines without induction of proinflammatory cytokines. J Infect Dis. 2000;182:200–205. doi: 10.1086/315641. [DOI] [PubMed] [Google Scholar]
  46. Kronenberg M. Toward an understanding of NKT cell biology: progress and paradoxes. Annu Rev Immunol. 2005;23:877–900. doi: 10.1146/annurev.immunol.23.021704.115742. [DOI] [PubMed] [Google Scholar]
  47. Lang R. Tuning of macrophage responses by Stat3-inducing cytokines: molecular mechanisms and consequences in infection. Immunobiology. 2005;210:63–76. doi: 10.1016/j.imbio.2005.05.001. [DOI] [PubMed] [Google Scholar]
  48. Larroche C, Mouthon L. Pathogenesis of hemophagocytic syndrome (HPS) Autoimmun Rev. 2004;3:69–75. doi: 10.1016/S1568-9972(03)00091-0. [DOI] [PubMed] [Google Scholar]
  49. Lepidi H, Bunnell JE, Martin ME, Madigan JE, Stuen S, Dumler JS. Comparative pathology, and immunohistology associated with clinical illness after Ehrlichia phagocytophila-group infections. Am J Trop Med Hyg. 2000;62:29–37. doi: 10.4269/ajtmh.2000.62.29. [DOI] [PubMed] [Google Scholar]
  50. Lin M, den Dulk-Ras A, Hooykaas PJ, Rikihisa Y. Anaplasma phagocytophilum AnkA secreted by type IV secretion system is tyrosine phosphorylated by Abl-1 to facilitate infection. Cell Microbiol. 2007;9:2644–2657. doi: 10.1111/j.1462-5822.2007.00985.x. [DOI] [PubMed] [Google Scholar]
  51. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends in Immunology. 2004;25:677–686. doi: 10.1016/j.it.2004.09.015. [DOI] [PubMed] [Google Scholar]
  52. Martin ME, Bunnell JE, Dumler JS. Pathology, immunohistology, and cytokine responses in early phases of human granulocytic ehrlichiosis in a murine model. J Infect Dis. 2000;181:374–378. doi: 10.1086/315206. [DOI] [PubMed] [Google Scholar]
  53. Martin ME, Caspersen K, Dumler JS. Immunopathology and ehrlichial propagation are regulated by interferon-gamma and interleukin-10 in a murine model of human granulocytic ehrlichiosis. Am J Pathol. 2001;158:1881–1888. doi: 10.1016/s0002-9440(10)64145-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Mattner J, Debord KL, Ismail N, et al. Exogenous and endogenous glycolipid antigens activate NKT cells during microbial infections. Nature. 2005;434:525–529. doi: 10.1038/nature03408. [DOI] [PubMed] [Google Scholar]
  55. Menasche G, Feldmann J, Fischer A, de Saint Basile G. Primary hemophagocytic syndromes point to a direct link between lymphocyte cytotoxicity and homeostasis. Immunol Rev. 2005;203:165–179. doi: 10.1111/j.0105-2896.2005.00224.x. [DOI] [PubMed] [Google Scholar]
  56. Mosser DM. The many faces of macrophage activation. J Leukoc Biol. 2003;73:209–212. doi: 10.1189/jlb.0602325. [DOI] [PubMed] [Google Scholar]
  57. Pedra JH, Mattner J, Tao J, et al. c-Jun NH2-terminal kinase 2 inhibits gamma interferon production during Anaplasma phagocytophilum infection. Infect Immun. 2008;76:308–316. doi: 10.1128/IAI.00599-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Ravelli A, Magni-Manzoni S, Pistorio A, et al. Preliminary diagnostic guidelines for macrophage activation syndrome complicating systemic juvenile idiopathic arthritis. J Pediatrics. 2005;146:598–604. doi: 10.1016/j.jpeds.2004.12.016. [DOI] [PubMed] [Google Scholar]
  59. Rikihisa Y. Mechanisms of obligatory intracellular infection with. Anaplasma phagocytophilum Clin Microbiol Rev. 2011;24:469–489. doi: 10.1128/CMR.00064-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Scharton TM, Scott P. Natural killer cells are a source of interferon gamma that drives differentiation of CD4+ T cell subsets and induces early resistance to Leishmania major in mice. J Exp Med. 1993;178:567–577. doi: 10.1084/jem.178.2.567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Scorpio DG, Akkoyunlu M, Fikrig E, Dumler JS. CXCR2 blockade influences Anaplasma phagocytophilum propagation but not histopathology in the mouse model of human granulocytic anaplasmosis. Clin Diagn Lab Immunol. 2004;11:963–968. doi: 10.1128/CDLI.11.5.963-968.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Scorpio DG, von Loewenich FD, Gobel H, Bogdan C, Dumler JS. Innate immune response to Anaplasma phagocytophilum contributes to hepatic injury. Clin Vaccine Immunol. 2006;13:806–809. doi: 10.1128/CVI.00092-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Scorpio DG, von Loewenich FD, Gobel H, Bogdan C, Dumler JS. Innate immune response to Anaplasma phagocytophilum contributes to hepatic injury. Clin Vaccine Immunol. 2006;13:806–809. doi: 10.1128/CVI.00092-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Scorpio DG, Leutenegger C, Berger J, Barat N, Madigan JE, Dumler JS. Sequential analysis of Anaplasma phagocytophilum msp2 transcription in murine and equine models of human granulocytic anaplasmosis. Clin Vaccine Immunol. 2008;15:418–424. doi: 10.1128/CVI.00417-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Thomas V, Fikrig E. Anaplasma phagocytophilum specifically induces tyrosine phosphorylation of ROCK1 during infection. Cell Microbiol. 2007;9:1730–1737. doi: 10.1111/j.1462-5822.2007.00908.x. [DOI] [PubMed] [Google Scholar]
  66. von Loewenich FD, Scorpio DG, Reischl U, Dumler JS, Bogdan C. Frontline: control of Anaplasma phagocytophilum, an obligate intracellular pathogen, in the absence of inducible nitric oxide synthase, phagocyte NADPH oxidase, tumor necrosis factor, Toll-like receptor (TLR)2 and TLR4, or the TLR adaptor molecule MyD88. Eur J Immunol. 2004;34:1789–1797. doi: 10.1002/eji.200425029. [DOI] [PubMed] [Google Scholar]
  67. Voskoboinik I, Smyth MJ, Trapani JA. Perforin-mediated target-cell death and immune homeostasis. Nat Rev Immunol. 2006;6:940–952. doi: 10.1038/nri1983. [DOI] [PubMed] [Google Scholar]
  68. Walker DH, Dumler JS. Will pathologists play as important a role in the future as they have in the past against the challenge of infectious diseases. Infect Agents Dis. 1995;4:167–170. [PubMed] [Google Scholar]
  69. Walker DH, Dumler JS. Emergence of the ehrlichioses as human health problems. Emerg Infect Dis. 1996;2:18–29. doi: 10.3201/eid0201.960102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Wormser GP, Dattwyler RJ, Shapiro ED, et al. The clinical assessment, treatment, and prevention of lyme disease, human granulocytic anaplasmosis, and babesiosis: clinical practice guidelines by the Infectious Diseases Society of America. Clin Infect Dis. 2006;43:1089–1134. doi: 10.1086/508667. [DOI] [PubMed] [Google Scholar]
  71. Zhang L, Shan A, Mathew B, et al. Rickettsial Seroepidemiology among farm workers, Tianjin, People’s Republic of China. Emerg Infect Dis. 2008;14:938–940. doi: 10.3201/eid1406.071502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Zhang L, Liu Y, Ni D, et al. Nosocomial transmission of human granulocytic anaplasmosis in China. JAMA. 2008;300:2263–2270. doi: 10.1001/jama.2008.626. [DOI] [PubMed] [Google Scholar]

RESOURCES