We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Host-cell interactions with pathogenic Rickettsia species

    Sanjeev K Sahni

    † Author for correspondence

    Department of Microbiology & Immunology, P.O. Box 672, University of Rochester School of Medicine & Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, USA and, Department of Medicine, Hematology–Oncology Unit, University of Rochester School of Medicine & Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, USA.

    &
    Elena Rydkina

    Department of Microbiology & Immunology, P.O. Box 672, University of Rochester School of Medicine & Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, USA.

    Published Online:https://doi.org/10.2217/fmb.09.6

    Pathogenic Rickettsia species are Gram-negative, obligate intracellular bacteria responsible for the spotted fever and typhus groups of diseases around the world. It is now well established that a majority of sequelae associated with human rickettsioses are the outcome of the pathogen’s affinity for endothelium lining the blood vessels, the consequences of which are vascular inflammation, insult to vascular integrity and compromised vascular permeability, collectively termed ‘Rickettsial vasculitis’. Signaling mechanisms leading to transcriptional activation of target cells in response to Rickettsial adhesion and/or invasion, differential activation of host-cell signaling due to infection with spotted fever versus typhus subgroups of Rickettsiae, and their contributions to the host’s immune responses and determination of cell fate are the major subtopics of this review. Also included is a succinct analysis of established in vivo models and their use for understanding Rickettsial interactions with host cells and pathogenesis of vasculotropic rickettsioses. Continued progress in these important but relatively under-explored areas of bacterial pathogenesis research should further highlight unique aspects of Rickettsial interactions with host cells, elucidate the biological basis of endothelial tropism and reveal novel chemotherapeutic and vaccination strategies for debilitating Rickettsial diseases.

    Bibliography

    • Walker DH: Rickettsiae and Rickettsial infections: the current state of knowledge. Clin. Infect. Dis.45(Suppl. 1),S39–S44 (2007).
    • Labruna MB, Camargo LM, Camargo EP, Walker DH: Detection of a spotted fever group Rickettsia in the tick Haemaphysalis juxtakochi in Rondonia, Brazil. Vet. Parasitol.127,169–174 (2005).
    • Gillespie JJ, Beier MS, Rahman MS et al.: Plasmids and Rickettsial evolution: insight from Rickettsia felis. PLoS One2,e266 (2007).
    • Gillespie JJ, Williams K, Shukla M et al.: Rickettsia phylogenomics: Unwinding the intricacies of obligate intracellular life. PLoS One3,e2018 (2008).
    • Dantas-Torres F: Rocky Mountain spotted fever. Lancet Infect. Dis.7,724–732 (2007).
    • Parola P, Raoult D: Tropical rickettsioses. Clin. Dermatol.24,191–200 (2006).
    • Rovery C, Raoult D: Mediterranean spotted fever. Infect. Dis. Clin. North Am.22,515–530 (2008).
    • Sousa R, França A, Dória Nòbrega S et al.: Host- and microbe-related risk factors for and pathophysiology of fatal Rickettsia conorii infection in Portuguese patients. J. Infect. Dis.198,576–585 (2008).
    • Paddock CD, Finley RW, Wright CS et al.: Rickettsia parkeri rickettsiosis and its clinical distinction from Rocky Mountain spotted fever. Clin. Infect. Dis.47,1188–1196 (2008).
    • 10  Apperson CS, Engber B, Nicholson WL et al.: Tick-borne diseases in North Carolina: is ‘Rickettsia amblyommii’ a possible cause of rickettsiosis reported as Rocky Mountain spotted fever? Vector Borne Zoonotic Dis.8,597–606 (2008).
    • 11  Dumler JS, Walker DH: Rocky Mountain spotted fever – changing ecology and persisting virulence. N. Engl. J. Med.353,551–553 (2005).
    • 12  Raoult D, Woodward T, Dumler JS: The history of epidemic typhus. Infect. Dis. Clin. North Am.18,127–140 (2004).
    • 13  Civen R, Ngo V: Murine typhus: an unrecognized suburban vectorborne disease. Clin. Infect. Dis.46,913–918 (2008).
    • 14  Walker DH: Principles of the malicious use of infectious agents to create terror: reasons for concern for organisms of the genus Rickettsia. Ann. NY Acad. Sci.990,739–742 (2003).
    • 15  Azad AF: Pathogenic Rickettsiae as bioterrorism agents. Clin. Infect. Dis.45(Suppl. 1),S52–S55 (2007).
    • 16  Valbuena G, Walker DH: The endothelium as a target for infections. Annu. Rev. Pathol.1,171–198 (2006).
    • 17  Sahni SK: Endothelial cell infection and hemostasis. Thromb. Res.119,531–549 (2007).
    • 18  Walker DH, Ismail N: Emerging and re-emerging rickettsioses: endothelial cell infection and early disease events. Nat. Rev. Microbiol.6,375–386 (2008).▪▪ A recent review summarizing the threats posed by pathogenic Rickettsia species to human health, host defense and Rickettsial virulence mechanisms, and manipulation of host cells by Rickettsiae.
    • 19  Aird WC: Endothelium in health and disease. Pharmacol. Rep.60,139–143 (2008).
    • 20  Eremeeva ME, Santucci LA, Popov VL, Walker DH, Silverman DJ: Rickettsia rickettsii infection of human endothelial cells: oxidative injury and reorganization of the cytoskeleton. In: Rickettsiae and Rickettsial diseases at the turn of the third millennium. Raoult D, Brouqui P (Eds). Elsevier Press, Paris, France, 128–144 (1999).
    • 21  Walker DH, Hudnall SD, Szaniawski WK, Feng HM: Monoclonal antibody-based immunohistochemical diagnosis of Rickettsialpox: the macrophage is the principal target. Mod. Pathol.12,529–533 (1999).
    • 22  Pober JS, Sessa WC: Evolving functions of endothelial cells in inflammation. Nat. Rev. Immunol.7,803–815 (2007).
    • 23  Hackstadt T: The biology of Rickettsiae. Infect. Agents Dis.5,127–143 (1996).
    • 24  Bleck TP: Central nervous system involvement in Rickettsial diseases. Neurol. Clin.17,801–812 (1999).
    • 25  Egermayer P: The role of infectious agents in pulmonary and systemic vascular disease. Expert Opin Pharmacother.2,1093–1097, (2001).
    • 26  Walker TS: Rickettsial interactions with human endothelial cells in vitro: adherence and entry. Infect. Immun.44,205–210 (1984).
    • 27  Li H, Walker DH: rOmpA is a critical protein for the adhesion of Rickettsia rickettsii to host cells. Microb. Pathog.24,289–298 (1998).
    • 28  Renesto P, Samson L, Ogata H et al.: Identification of two putative Rickettsial adhesins by proteomic analysis. Res. Microbiol.157,605–612 (2006).
    • 29  Martinez JJ, Seveau S, Veiga E, Matsuyama S, Cossart P: Ku70, a component of DNA-dependent protein kinase, is a mammalian receptor for Rickettsia conorii. Cell123,1013–1023 (2005).▪ First report of identification of a receptor involved in Rickettsial internalization into mammalian host cells.
    • 30  Martinez JJ, Cossart P: Early signaling events involved in the entry of Rickettsia conorii into mammalian cells. J. Cell Sci.117,5097–5106 (2004).
    • 31  Rydkina E, Turpin LC, Sahni SK: Activation of p38 mitogen-activated protein kinase module facilitates in vitro host cell invasion by Rickettsia rickettsii. J. Med. Microbiol.57,1172–1175 (2008).
    • 32  Chan YG, Cardwell MM, Hermanas TM, Uchiyama T, Martinez JJ: Rickettsial outer-membrane protein B (rOmpB) mediates bacterial invasion through Ku70 in an actin, c-Cbl, clathrin and caveolin 2-dependent manner. Cell. Microbiol.11,629–644 (2009).
    • 33  Teysseire N, Boudier J-A, Raoult D: Rickettsia conorii entry into Vero cells. Infect. Immun.63,366–374 (1995).
    • 34  Whitworth T, Popov VL, Yu XJ, Walker DH, Bouyer DH: Expression of the Rickettsia prowazekii pld or tlyC gene in Salmonella enterica serovar Typhimurium mediates phagosomal escape. Infect. Immun.73,6668–6673 (2005).
    • 35  Silverman DJ: Rickettsia rickettsii-induced cellular injury of human vascular endothelium in vitro. Infect. Immun.44,545–553 (1984).▪ Description of an in vitro model of endothelial call infection with Rickettsia rickettsii and potential mechanisms of pathogen-induced cell injury.
    • 36  Silverman DJ: Adherence of platelets to human endothelial cells infected by Rickettsia rickettsii. J. Infect. Dis.153,694–700 (1986).
    • 37  Teysseire N, Arnoux D, George G, Sampol J, Raoult D: von Willebrand factor release and thrombomodulin and tissue factor expression in Rickettsia conorii-infected endothelial cells. Infect. Immun.60,4388–4393 (1992).
    • 38  Sporn LA, Haidaris PJ, Shi RJ, Nemerson Y, Silverman DJ, Marder VJ: Rickettsia rickettsii infection of cultured human endothelial cells induces tissue factor expression. Blood83,1527–1534 (1994).
    • 39  Kaplanski GN, Teysseire N, Farnarier C et al.: IL-6 and IL-8 production from cultured human endothelial cells stimulated by infection with Rickettsia conorii via a cell-associated IL-1α-dependent pathway. J. Clin. Invest.96,2839–2844 (1995).
    • 40  Sporn LA, Marder VJ: Interleukin-1α production during Rickettsia rickettsii infection of cultured endothelial cells: potential role in autocrine cell stimulation. Infect. Immun.64,1609–1613 (1996).
    • 41  Dignat-George F, Teysseire N, Mutin M et al.: Rickettsia conorii infection enhances vascular cell adhesion molecule-1 and intercellular adhesion molecule-1-dependent mononuclear cell adherence to endothelial cells. J. Infect. Dis.175,1142–1152 (1997).
    • 42  Sporn LA, Lawrence SO, Silverman DJ, Marder VJ: E-selectin-dependent neutrophil adhesion to Rickettsia rickettsii-infected endothelial cells. Blood81,2406–2412 (1993).
    • 43  Drancourt M, Allessi M-C, Levy P-Y, Juhan-Vague I, Raoult D: Selection of tissue-type plasminogen activator and plasminogen activator inhibitor by Rickettsia conroii and Rickettsia rickettsii-infected cultured endothelial cells. Infect. Immun.58,2459–2463 (1990).
    • 44  Shi R-J, Simpson-Haidaris PJ, Marder VJ, Silverman DJ, Sporn LA: Post-transcriptional regulation of endothelial cell plasminogen activator inhibitor-1 expression during Rickettsia rickettsii infection. Microb. Pathog.28,127–133 (2000).
    • 45  Sporn LA, Shi R-J, Lawrence SO, Silverman DJ, Marder VJ: Rickettsia rickettsii infection of cultured endothelial cells induces release of large von Willebrand factor multimers from Weibel–Palade bodies. Blood78,2595–2602 (1991).
    • 46  Walker TS, Brown JS, Hoover CS, Morgan DA: Endothelial prostaglandin secretion: effects of typhus Rickettsiae. J. Infect. Dis.162,1136–1144 (1990).
    • 47  Bierhaus A, Nawroth PP: Modulation of the vascular endothelium during infection – the role of NF-κB activation. Contrib. Microbiol.10,86–105 (2003).
    • 48  Spiecker M, Darius H, Liao JK: A functional role of IκBε in endothelial cell activation. J. Immunol.164,3316–3322 (2000).
    • 49  Zandi E, Rothwarf DM, Delhase M, Hayakawa M, Karin M: The IκB kinase complex (IKK) contains two kinase subunits, IKKα and IKKβ, necessary for IκB phosphorylation and NF-κB activation. Cell91,243–252 (1997).
    • 50  Sporn LA, Sahni SK, Lerner NB et al.: Rickettsia rickettsii infection of cultured human endothelial cells induces NF-κB activation. Infect. Immun.65,2786–2791 (1997).
    • 51  Clifton DR, Rydkina E, Freeman RS, Sahni SK: NF-κB activation during Rickettsia rickettsii infection of endothelial cells involves the activation of catalytic IκB kinases IKKα and IKKβ and phosphorylation-proteolysis of the inhibitor protein IκBα. Infect. Immun.73,155–165 (2005).
    • 52  Sahni SK, Van Antwerp DJ, Eremeeva ME, Silverman DJ, Marder VJ, Sporn LA: Proteasome-independent activation of nuclear factor κB in cytoplasmic extracts from human endothelial cells by Rickettsia rickettsii. Infect. Immun.66,1827–1833 (1998).▪ Experimental evidence supporting the hypothesis that Rickettsiae may interact directly with inactive nuclear factor-κB present in the host cytoplasm.
    • 53  Sahni SK, Rydkina E, Joshi SG, Sporn LA, Silverman DJ: Interactions of Rickettsia rickettsii with endothelial nuclear factor-κB in a “cell-free” system. Ann. NY Acad. Sci.990,635–641 (2003).
    • 54  Heinzen RA: Rickettsial actin-based motility: behavior and involvement of cytoskeletal regulators. Ann. NY Acad. Sci.990,535–547 (2003).
    • 55  Gouin E, Welch MD, Cossart P: Actin-based motility of intracellular pathogens. Curr. Opin Microbiol.8,35–45 (2005).
    • 56  Andersson SGE, Zomorodipour A, Andersson JO et al.: The genome sequence of Rickettsia prowazekii and the origin of mitochondria. Nature396,133–140 (1998).▪ The very first report of the entire genome sequence of a pathogenic Rickettsia species.
    • 57  Ogata H, Audic S, Renesto-Audiffren P et al.: Mechanisms of evolution in Rickettsia conorii and R. prowazekii. Science293,2093–2098 (2001).
    • 58  Amiri H, Davids W, Andersson SG: Birth and death of orphan genes in Rickettsia. Mol. Biol. Evol.20,1575–1587 (2003).
    • 59  Rydkina E, Silverman DJ, Sahni SK: Similarities and differences in host cell signaling following infection with different Rickettsia species. Ann. NY Acad. Sci.1063,203–206 (2005).
    • 60  Rydkina E, Sahni A, Silverman DJ, Sahni SK: Comparative analysis of host-cell signaling mechanisms activated in response to infection with Rickettsia conorii and Rickettsia typhi. J. Med. Microbiol.56,896–906 (2007).
    • 61  Ventura C, Maioli M: Protein kinase C control of gene expression. Crit. Rev. Eukaryotic Gene Exp.11,243–267 (2001).
    • 62  Shirakawa F, Mizel SB: in vitro activation and nuclear translocation of NF-κB catalyzed by cyclic AMP-dependent protein kinase and protein kinase C. Mol. Cell. Biol.9,2424–2430 (1989).
    • 63  Sahni SK, Turpin LC, Brown TL, Sporn LA: Involvement of protein kinase C in Rickettsia rickettsii-induced transcriptional activation of the host endothelial cell. Infect. Immun.67,6418–6423 (1999).
    • 64  Bogatcheva NV, Dudek SM, Garcia JG, Verin AD: Mitogen-activated protein kinases in endothelial pathophysiology. J. Investig. Med.51,341–352 (2003).
    • 65  Schulze-Osthoff K, Ferrari D, Riehemann K, Wesselborg S: Regulation of NF-κB activation by MAP kinase cascades. Immunobiol.198,35–49 (1997).
    • 66  Roux PP, Blenis J: ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions. Microbiol. Mol. Biol. Rev.68,320–344 (2004).
    • 67  Rydkina E, Silverman DJ, Sahni SK: Activation of p38 stress-activated protein kinase during Rickettsia rickettsii infection of human endothelial cells: role in the induction of chemokine response. Cell. Microbiol.7,1519–1530 (2005).
    • 68  Eremeeva ME, Dasch GA, Silverman DJ: Interaction of Rickettsiae with eukaryotic cells. Adhesion, entry, intracellular growth, and host cell responses. Subcell. Biochem.33,479–516 (2000).
    • 69  Rydkina E, Sahni A, Silverman DJ, Sahni SK: Rickettsia rickettsii infection of cultured human endothelial cells induces heme oxygenase 1 expression. Infect. Immun.70,4045–4052 (2002).
    • 70  Silverman DJ, Bond SB: Infection of human vascular endothelial cells by Rickettsia rickettsii. J. Infect. Dis.149,201–206 (1984).
    • 71  Eremeeva ME, Silverman DJ: Effects of the antioxidant α-lipoic acid on human umbilical vein endothelial cells infected with Rickettsia rickettsii. Infect. Immun.66,2290–2299 (1998).▪ First report of experimental findings in support of a protective role for an antioxidant compound, α-lipoic acid, during in vitro host-cell infection with R. rickettsii.
    • 72  Eremeeva ME, Silverman DJ: Rickettsia rickettsii infection of the EA.hy 926 endothelial cell line: morphological response to infection and evidence for oxidative injury. Microbiology144,2037–2048 (1998).
    • 73  Rydkina E, Sahni SK, Santucci LA, Turpin LC, Baggs RB, Silverman DJ: Selective modulation of antioxidant enzyme activities in host tissues during Rickettsia conorii infection. Microb. Pathog.36,293–301 (2004).▪ Initial in vivo evidence for involvement of oxidative stress mechanisms and potential protective functions of the antioxidant compound α-lipoic acid in the pathogenesis of Rocky Mountain spotted fever.
    • 74  Maines MD, Gibbs PE: 30 some years of heme oxygenase: from a “molecular wrecking ball” to a “mesmerizing” trigger of cellular events. Biochem. Biophys. Res. Commun.338,568–577 (2005).
    • 75  Rydkina E, Sahni A, Baggs RB, Silverman DJ, Sahni SK: Infection of human endothelial cells with spotted fever group Rickettsiae stimulates cyclooxygenase 2 expression and release of vasoactive prostaglandins. Infect. Immun.74,5067–5074 (2006).
    • 76  Turco J, Winkler HH: Selection of α/β interferon- and γ interferon-resistant Rickettsiae by passage of Rickettsia prowazekii in L929 cells. Infect. Immun.58,3279–3285 (1990).
    • 77  Turco J, Winkler HH: Effect of mouse lymphokines and cloned mouse interferon-γ on the interaction of Rickettsia prowazekii with mouse macrophage-like RAW264.7 cells. Infect. Immun.45,303–308 (1984).
    • 78  Turco J, Winkler HH: γ-interferon-induced inhibition of the growth of Rickettsia prowazekii in fibroblasts cannot be explained by the degradation of tryptophan or other amino acids. Infect. Immun.53,38–46 (1986).
    • 79  Turco J, Winkler HH: Isolation of Rickettsia prowazekii with reduced sensitivity to γ-interferon. Infect. Immun.57,1765–1772 (1989).
    • 80  Turco J, Winkler HH: Comparison of properties of virulent, avirulent, and interferon-resistant Rickettsia prowazekii strains. Infect. Immun.59,1647–1655 (1991).
    • 81  Manor E, Sarov I: Inhibition of Rickettsia conorii growth by recombinant tumor necrosis factor α: enhancement of inhibition by γ-interferon. Infect. Immun.58,1886–1890 (1990).
    • 82  Feng HM, Walker DH: Mechanisms of intracellular killing of Rickettsia conorii in infected human endothelial cells, hepatocytes, and macrophages. Infect. Immun.68,6729–6736 (2000).
    • 83  Walker DH, Olano JP, Feng HM: Critical role of cytotoxic T lymphocytes in immune clearance of Rickettsial infection. Infect. Immun.69,1841–1846 (2001).
    • 84  Feng HM, Popov VL, Walker DH: Depletion of γ-interferon and tumor necrosis factor α in mice with Rickettsia conorii-infected endothelium: impairment of rickettsicidal nitric oxide production resulting in fatal, overwhelming Rickettsial disease. Infect. Immun.62,1952–1960 (1994).
    • 85  Walker DH, Popov VL, Feng HM: Establishment of a novel endothelial target mouse model of a typhus group rickettsiosis: evidence for critical roles for γ interferon and CD8 T lymphocytes. Lab. Invest.80,1361–1372 (2000).▪ First detailed description of a susceptible mouse strain with Rickettsia typhi as a useful laboratory model mimicking disseminated endothelial infection seen during typhus group rickettsioses in humans.
    • 86  Billings AN, Feng HM, Olano JP, Walker DH: Rickettsial infection in murine models activates an early anti-Rickettsial effect mediated by NK cells and associated with production of γ interferon. Am. J. Trop. Med. Hyg.65,52–56 (2001).
    • 87  Clifton DR, Rydkina E, Huyck H et al.: Expression and secretion of chemotactic cytokines IL-8 and MCP-1 by human endothelial cells after Rickettsia rickettsii infection: regulation by nuclear transcription factor NF-κB. Int. J. Med. Microbiol.295,267–278 (2005).
    • 88  Valbuena G, Walker DH: Expression of CX3CL1 (fractalkine) in mice with endothelial-target Rickettsial infection of the spotted-fever group. Virchows Arch.446,21–27 (2005).
    • 89  Radulovic S, Price PW, Beier MS, Gaywee J, Macaluso JA, Azad A: Rickettsia-macrophage interactions: host cell responses to Rickettsia akari and Rickettsia typhi. Infect. Immun.70,2576–2582 (2002).
    • 90  Valbuena G, Bradford W, Walker DH: Expression analysis of the T-cell-targeting chemokines CXCL9 and CXCL10 in mice and humans with endothelial infections caused by Rickettsiae of the spotted fever group. Am. J. Pathol.163,1357–1369 (2003).
    • 91  Walker TS, Dersch MW, White WE: Effects of typhus Rickettsiae on peritoneal and alveolar macrophages: Rickettsiae stimulate leukotriene and prostaglandin secretion. J. Infect. Dis.163,568–573 (1991).
    • 92  Walker TS, Mellott GE: Rickettsial stimulation of endothelial platelet-activating factor synthesis. Infect. Immun.61,2024–2029 (1993).
    • 93  Bechah Y, Capo C, Raoult D, Mege JL: Infection of endothelial cells with virulent Rickettsia prowazekii increases the transmigration of leukocytes. J. Infect. Dis.197,142–147 (2008).
    • 94  Clifton DR, Goss RA, Sahni SK et al.: NF-κB-dependent inhibition of apoptosis is essential for host cell survival during Rickettsia rickettsii infection. Proc. Natl. Acad. Sci. USA95,4646–4651 (1998).▪ Original report documenting a stratagem exploited by intracellular Rickettsiae to delay apoptosis early during infection in order to prolong host-cell survival.
    • 95  Riedl SJ, Shi Y: Molecular mechanisms of caspase regulation during apoptosis. Nat. Rev. Mol. Cell. Biol.5,897–907 (2004).
    • 96  Joshi SG, Francis CW, Silverman DJ, Sahni SK: Nuclear factor-κB protects against host cell apoptosis during Rickettsia rickettsii infection by inhibiting activation of apical and effector caspases and maintaining mitochondrial integrity. Infect. Immun.71,4127–4136 (2003).
    • 97  Chan SL, Yu VC: Proteins of the Bcl-2 family in apoptosis signaling: from mechanistic insights to therapeutic opportunities. Clin. Exp. Pharmacol. Physiol.31,119–128 (2004).
    • 98  Scorrano L, Korsmeyer SJ: Mechanisms of cytochrome c release by proapoptotic Bcl-2 family members. Biochem. Biophys. Res. Commun.304,437–444 (2003).
    • 99  Joshi SG, Francis CW, Silverman DJ, Sahni SK: NF-κB activation suppresses host cell apoptosis during Rickettsia rickettsii infection via regulatory effects on intracellular localization or levels of apoptogenic and anti-apoptotic proteins. FEMS Microbiol. Lett.234,333–341 (2004).
    • 100  Bechelli JB, Rydkina E, Colonne PM, Sahni SK: Rickettsia rickettsii infection protects human microvascular endothelial cells against staurosporine-induced apoptosis by a cIAP2-independent mechanism. J. Infect. Dis. (2009) (Epub ahead of print).
    • 101  Valbuena G, Walker DH: Changes in the adherens junctions of human endothelial cells infected with spotted fever group Rickettsiae. Virchows Arch.446,379–382 (2005).
    • 102  Woods ME, Olano JP: Host defenses to Rickettsia rickettsii infection contribute to increased microvascular permeability in human cerebral endothelial cells. J. Clin. Immunol.28,174–185 (2008).▪ In vitro evidence indicating involvement of both the direct effects of infection on endothelial cells and inflammatory host cell-derived mediators in inducing changes in vascular permeability.
    • 103  Yoshikai Y: Roles of prostaglandins and leukotrienes in acute inflammation caused by bacterial infection. Curr. Opin Infect. Dis.14,257–263 (2001).
    • 104  Eisemann CS, Nypaver MJ, Osterman JV: Susceptibility of inbred mice to Rickettsiae of the spotted fever group. Infect. Immun.43,143–148 (1984).
    • 105  Walker DH: Endothelial-target Rickettsial infection. Lab. Anim. Sci.47,483–485 (1997).▪ A commentary highlighting the salient features of a mouse model of Rocky Mountain spotted fever and applicability to investigating the pathogenesis of vasculotropic Rickettsial diseasaes.
    • 106  Feng HM, Wen J, Walker DH: Rickettsia australis infection: a murine model of a highly invasive vasculopathic rickettsiosis. Am. J. Pathol.142,1471–1482 (1993).
    • 107  Valbuena G, Walker DH: Effect of blocking the CXCL9/10-CXCR3 chemokine system in the outcome of endothelial-target Rickettsial infections. Am. J. Trop. Med. Hyg.71,393–399 (2004).
    • 108  Eremeeva ME, Liang Z, Paddock C et al.: Rickettsia rickettsii infection in the pine vole, Microtus pinetorum: kinetics of infection and quantitation of antioxidant enzyme gene expression by RT-PCR. Ann. NY Acad. Sci.990,468–473 (2003).
    • 109  Ormsbee R, Peacock M, Gerloff R, Tallent G, Wike D: Limits of Rickettsial infectivity. Infect. Immun.19,239–245 (1978).
    • 110  Crist AE Jr, Wisseman CL Jr, Murphy JR: Characteristics of lymphoid cells that adoptively transfer immunity to Rickettsia mooseri infection in mice. Infect. Immun.44,55–60 (1984).
    • 111  Gonder JC, Kenyon RH, Pedersen CE Jr: Epidemic typhus infection in cynomolgus monkeys (Macaca fascicularis). Infect. Immun.30,219–223 (1980).
    • 112  Bechah Y, Capo C, Grau GE, Raoult D, Mege JL: A murine model of infection with Rickettsia prowazekii: implications for pathogenesis of epidemic typhus. Microbes Infect.9,898–906 (2007).▪ An original report documenting the susceptibility of BALB/c mice to infection with Rickettsia prowazekii and the potential for exploitation of this mouse model to study the pathogenesis of epidemic typhus.
    • 113  Bechah Y, Capo C, Mege JL, Raoult D: Rickettsial diseases: from Rickettsiaarthropod relationships to pathophysiology and animal models. Future Microbiol.3,223–236 (2008).
    • 114  Davi G, Giammarresi C, Vigneri S et al.: Demonstration of Rickettsia conorii-induced coagulative and platelet activation in vivo in patients with Mediterranean spotted fever. Thromb. Haemost.74,631–634 (1995).
    • 115  Elghetany MT, Walker DH: Hemostatic changes in Rocky Mountain spotted fever and Mediterranean spotted fever. Am. J. Clin. Pathol.112,159–168 (1999).
    • 116  Yamada T, Harber P, Pettit GW, Wing DA, Oster CN: Activation of the kallikrein–kinin system in Rocky Mountain spotted fever. Ann. Intern. Med.88,764–768 (1978).
    • 117  Davi G, Giammarresi C, Vitale G, Mansueto S: Enhanced thromboxane biosynthesis in patients with Mediterranean spotted fever. Blood83,3101 (1994).
    • 118  Cillari E, Milano S, D’Agostino P et al.: Depression of CD4 T cell subsets and alteration in cytokine profile in boutonneuse fever. J. Infect. Dis.174,1051–1057 (1996).
    • 119  Vitale G, Mansueto S, Gambino G et al.: The acute phase response in Sicilian patients with boutonneuse fever admitted to hospitals in Palermo, 1992–1997. J. Infect.42,33–39 (2001).
    • 120  Sessler CN, Schwartz M, Windsor AC, Fowler AA: Increased serum cytokines and intercellular adhesion molecule-1 in fulminant Rocky Mountain spotted fever. Crit. Care Med.23,973–976 (1995).
    • 121  Mahara F: Japanese spotted fever: report of 31 cases and review of the literature. Emerg. Infect. Dis.3,105–111 (1997).
    • 122  Iwasaki H, Mahara F, Takada N, Fujita H, Ueda T: Fulminant Japanese spotted fever associated with hypercytokinemia. J. Clin. Microbiol.39,2341–2343 (2001).
    • 123  Jensenius M, Ueland T, Fournier PE et al.: Systemic inflammatory responses in African tick-bite fever. J. Infect. Dis.187,1332–1336 (2003).
    • 124  Damås JK, Davi G, Jensenius M et al.: Relative chemokine and adhesion molecule expression in Mediterranean spotted fever and African tick bite fever. J. Infect.58(1),68–75 (2009).
    • 125  de Sousa R, Ismail N, Nobrega SD et al.: Intralesional expression of mRNA of interferon-γ, tumor necrosis factor-α, interleukin-10, nitric oxide synthase, indoleamine-2,3-dioxygenase, and RANTES is a major immune effector in Mediterranean spotted fever rickettsiosis. J. Infect. Dis.196,770–781 (2007).
    • 126  Walker DH, Kirkman NH: Rocky Mountain spotted fever and deficiency in glucose-6-phosphate dehydrogenase. J. Infect. Dis.142,771 (1980).
    • 127  Renesto P, Rovery C, Schrenzel J et al.: Rickettsia conorii transcriptional response with in inoculation eschar. PLoS One3,e3681 (2008).
    • 128  Gouin E, Egile C, Dehoux P et al.: The RickA protein of Rickettsia conorii activates the Arp2/3 complex. Nature427,457–461 (2004).
    • 129  Jeng RL, Goley ED, D’Alessio JA et al.: A Rickettsia WASP-like protein activates the Arp2/3 complex and mediates actin-based motility. Cell. Microbiol.6,761–769 (2004).
    • 130  Malek JA, Wierzbowski JM, Tao W et al.: Protein interaction mapping on a functional shotgun sequence of Rickettsia sibirica. Nucleic Acids Res.32,1059–1064 (2004).
    • 131  Walker DH, Yu XJ: Progress in Rickettsial genome analysis from pioneering of Rickettsia prowazekii to the recent Rickettsia typhi. Ann. NY Acad. Sci.1063,13–25 (2005).
    • 132  Nagai H, Kagan JC, Zhu X, Kahn RA, Roy CR: A bacterial guanine nucleotide exchange factor activates ARF on Legionella phagosomes. Science295,679–682 (2002).
    • 133  Ogata H, Renesto P, Audic S et al.: The genome sequence of Rickettsia felis identifies the first putative conjugative plasmid in an obligate intracellular parasite. PLoS Biol.3,e248 (2005).
    • 134  Baldridge GD, Burkhardt NY, Felsheim RF, Kurtti TJ, Munderloh UG: Transposon insertion reveals pRM, a plasmid of Rickettsia monacensis. Appl. Environ. Microbiol.73,4984–4995 (2007).
    • 135  Baldridge GD, Burkhardt NY, Felsheim RF, Kurtti TJ, Munderloh UG: Plasmids of the pRM/pRF family occur in diverse Rickettsia species. Appl. Environ. Microbiol.74,645–652 (2008).